PEG precipitation distinguishes bioactive monomeric PRL from inactive macroprolactin. Key parameters from recent studies:
Assay Platform | Total PRL (mIU/L) | Monomeric PRL (mIU/L) | PEG Recovery (%) |
---|---|---|---|
Architect | 84–409 | 58–224 | 63 |
Kryptor | 59–291 | 90–331 | 122 |
Cobas | 58–316 | 58–257 | 64 |
Recovery thresholds: <50% PEG recovery indicates macroprolactinemia .
Clinical utility: Corrects falsely elevated PRL readings in 12–26% of hyperprolactinemia cases .
Polymorphisms in the PRL gene promoter and exon regions correlate with reproductive traits:
SNP Locus | Genotype | Egg Production Impact |
---|---|---|
g.8052T>C | CC | ↑ Daily egg yield |
g.7835A>G | AG | Neutral |
24-bp indel | H3 | ↑ Total egg number |
g.8052T>C in exon 5 significantly enhances hen-day production (P < 0.05) .
The 24-bp indel at -358 bp increases laying persistence in White Leghorns .
Chicken PRL enhances immune cell activity:
Lymphocyte proliferation: Dose-dependent mitogenesis in thymus/spleen cells .
NK cell activation: +15–20% increase in infected poultry treated with PRL .
Anti-inflammatory effects: Suppresses LPS-induced TNF-α and IL-6 via NFκB inhibition .
Prolactin, a neuroendocrine hormone primarily produced by the pituitary gland, is also synthesized in other cells like those found in the placenta, brain, and uterus. Its secretion is triggered by various stimuli such as eating, nursing, mating, estrogen treatment, and ovulation. While prolactin's primary function is to stimulate and maintain lactation, it also plays a role in breast cancer development, regulation of reproductive functions, and immunoregulation.
Recombinant Pegylated Chicken Prolactin, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 199 amino acids and an additional alanine at the N-terminal. This pegylated prolactin is mono-pegylated with a molecular mass of approximately 39 kDa. However, under non-denaturing conditions, it exhibits a behavior similar to a 220 kDa protein due to its increased hydrodynamic volume.
The purification of the pegylated prolactin protein is achieved through proprietary chromatographic techniques.
The protein was lyophilized from a concentrated solution (1 mg/ml) containing 0.02% sodium bicarbonate (NaHCO3).
To reconstitute the lyophilized prolactin, it is recommended to dissolve it in sterile 0.4% sodium bicarbonate (NaHCO3) at pH 8.5, to a concentration of at least 100 µg/ml. This solution can then be further diluted in other aqueous solutions as needed.
Lyophilized prolactin, while stable at room temperature for up to 3 weeks, should ideally be stored desiccated at temperatures below -18°C. After reconstitution, prolactin should be stored at 4°C for 2-7 days. For long-term storage, freezing at -18°C is recommended.
For enhanced stability during long-term storage, adding a carrier protein like 0.1% HSA (Human Serum Albumin) or BSA (Bovine Serum Albumin) is recommended.
Avoid repeated freeze-thaw cycles to maintain protein integrity.
The purity is determined to be greater than 95% based on gel filtration analysis.
The biological activity of Pegylated Chicken Prolactin was assessed in vitro through its ability to induce proliferation in Nb2 cells and Baf/3 cells stably transfected with Chicken Prolactin receptors. While its activity is lower than non-pegylated Chicken Prolactin in vitro, it is expected to exhibit higher in vivo activity due to its prolonged circulation time resulting from pegylation.
Chicken prolactin (chPRL) is a 23 kDa polypeptide hormone primarily secreted by the anterior pituitary cells, with high sequence homology across different poultry varieties. In poultry, prolactin plays crucial roles in reproductive processes including nesting behavior, hatching, egg production, and broodiness regulation . Unlike in mammals where prolactin is primarily associated with lactation, in chickens it regulates reproductive cycles and parental behaviors. Physiologically, elevated prolactin levels are associated with reduced egg production as they trigger and maintain incubation behavior. The hormone is also present in immune tissues such as the thymus, spleen, and lymphocytes, suggesting immunomodulatory functions beyond reproduction . The gene encoding chicken prolactin is highly conserved across poultry species, with polymorphisms in this gene being linked to variations in reproductive performance among different breeds.
PEGylation is a biochemical process that involves the covalent attachment of polyethylene glycol (PEG) molecules to proteins or peptides. In chicken prolactin research, PEGylation serves to modify the pharmacokinetic and pharmacodynamic properties of the native hormone. The primary benefits of this modification include prolonged circulation time in the bloodstream, reduced immunogenicity, increased stability, and protection against proteolytic degradation .
Research has demonstrated that PEGylated chicken prolactin (PEG-chPRL) remains in circulation approximately 16 hours longer than non-PEGylated chPRL, despite showing reduced in vitro biological activity (approximately 10-fold lower in cell bioassays) . This extended half-life makes PEGylated prolactin particularly valuable for in vivo experiments, as it reduces the required dosing frequency while potentially improving efficacy. The research application of PEGylation thus represents a strategic approach to enhancing the utility of chicken prolactin for experimental studies, particularly those investigating long-term hormonal effects in poultry.
Researchers distinguish between monomeric prolactin (the biologically active 23 kDa form) and macroprolactin (higher molecular weight aggregates with reduced bioactivity) using several methodological approaches, with PEG precipitation being most commonly employed in research settings.
The differentiation protocol typically involves:
PEG precipitation: Mixing serum with an equal volume of 25% (wt/vol) PEG 6000, incubating for 10 minutes, followed by centrifugation at 1810 × g for 30 minutes .
Comparative analysis: The supernatant containing monomeric prolactin is analyzed alongside a non-precipitated but similarly diluted aliquot of the same serum .
Recovery calculation: Macroprolactin presence is indicated when post-PEG recovery is less than a method-specific cutoff (typically 40-50%) .
This distinction is crucial in research settings because macroprolactin can cause falsely elevated prolactin measurements in immunoassays despite having limited biological activity. The gold standard method is gel filtration chromatography (GFC), which separates prolactin molecules based on size, but this is less practical for routine screening of multiple samples .
Different immunoassay methods show varying reactivity toward macroprolactin, necessitating method-specific reference intervals. The optimal recovery cutoff varies by method: 40% for some assays (AutoDelfia, Architect, Cobas) and higher thresholds for others (50% for Immulite, 80% for Kryptor) . This methodological consideration is essential for accurate prolactin quantification in research studies.
The production of recombinant chicken prolactin (chPRL) for research purposes primarily employs bacterial expression systems, with Escherichia coli being the most commonly utilized host. The methodological approach involves:
Gene cloning: The chicken prolactin gene is cloned into a suitable expression vector.
Bacterial transformation: E. coli is transformed with the recombinant vector.
Protein expression: The bacteria express the recombinant protein, typically as inclusion bodies.
Solubilization and refolding: The inclusion bodies are solubilized and the protein is refolded to achieve its native conformation .
For purification of monomeric chPRL, researchers typically employ a multi-step chromatographic approach:
Initial purification using ion-exchange chromatography
Further purification via size exclusion chromatography to isolate the monomeric form
Additional chromatographic steps to achieve high purity
The success of proper refolding can be verified through circular dichroism (CD) spectroscopy, which shows that properly refolded chPRL has CD spectra similar to human prolactin, indicating conservation of secondary structure elements . This methodological approach yields biologically active monomeric chPRL suitable for research applications and further modifications such as PEGylation.
The optimal method for PEGylating chicken prolactin involves a controlled chemical reaction followed by specialized purification techniques to isolate the mono-PEGylated form. The process typically follows these methodological steps:
Chemical conjugation: Recombinant monomeric chPRL is reacted with activated PEG molecules (typically mPEG-aldehyde or mPEG-NHS) under conditions that favor attachment at specific amino acid residues, usually the N-terminal or lysine residues.
Reaction optimization: Parameters including pH, temperature, molar ratio of PEG to protein, and reaction time are carefully controlled to maximize the yield of mono-PEGylated species while minimizing undesired di- or poly-PEGylated products.
Purification: For the critical separation of mono-PEGylated chPRL from unreacted prolactin and other PEGylated forms, hydrophobic interaction chromatography (HIC) has proven particularly effective . This represents an improvement over size exclusion chromatography, which was traditionally used but provided less efficient separation.
The hydrophobic interaction chromatography approach leverages the altered surface properties of the PEGylated protein. The research has demonstrated that HIC can successfully isolate PEG-chPRL to homogeneity and is likely applicable to purification of other PEGylated proteins as well . This methodological advancement significantly improves the efficiency of generating highly pure mono-PEGylated prolactin for research applications.
Researchers employ multiple complementary methodologies to accurately assess and compare the biological activity of non-PEGylated versus PEGylated chicken prolactin:
In vitro assessment methods:
Receptor binding assays: Measuring the interaction between prolactin forms and human prolactin receptor extracellular domain (hPRLR-ECD). Studies show that PEG-chPRL typically exhibits approximately 2-fold lower binding affinity compared to non-PEGylated chPRL .
Cell proliferation assays: Utilizing Nb2-11C cells (a rat lymphoma cell line dependent on prolactin for growth) to measure PRLR-mediated proliferation. Research indicates that PEG-chPRL shows approximately 10-fold lower activity in stimulating cell proliferation compared to non-PEGylated chPRL .
Structural analysis: Circular dichroism (CD) spectroscopy to compare secondary structure elements. The CD spectra of non-PEGylated and PEGylated chPRL are typically almost identical, indicating that PEGylation does not significantly alter protein folding .
In vivo assessment methods:
Pharmacokinetic studies: Measuring serum concentration over time after administration. Studies demonstrate that PEG-chPRL remains in circulation approximately 16 hours longer than non-PEGylated chPRL .
Physiological response measurement: Assessing downstream biological effects such as alterations in corticosteroid levels. Research shows that PEG-chPRL injections in chickens produce significantly more profound effects on subsequent corticosteroid levels compared to non-PEGylated chPRL, despite lower in vitro activity .
This multi-faceted evaluation approach allows researchers to comprehensively characterize the relationship between in vitro activity, pharmacokinetics, and in vivo efficacy, providing crucial insights for experimental design when using these research reagents.
Prolactin gene polymorphisms significantly influence egg production in chickens through alterations in the hormone's expression or functionality. Specific single nucleotide polymorphisms (SNPs) in the prolactin gene have been correlated with variation in egg-laying performance across different chicken breeds.
Key genetic findings:
The exon 5 region of the chicken prolactin gene contains several polymorphic sites that impact reproductive traits. Research on IPB-D1 chickens identified three significant SNPs:
g.7835A>G
g.7886A>T
g.8052T>C
Among these, the g.8052T>C mutation demonstrated a statistically significant association with egg production traits . This specific polymorphism may serve as a genetic marker to enhance selection for improved egg production in breeding programs.
Methodological approach for identification:
Researchers employ a systematic genetic analysis pipeline:
DNA extraction: Genomic DNA is isolated from blood or tissue samples using methods such as the phenol-chloroform extraction technique .
PCR amplification: Target regions of the prolactin gene are amplified using polymerase chain reaction with sequence-specific primers. For exon 5 analysis, primers generate amplicons of approximately 557 bp .
DNA sequencing: Amplified fragments undergo sequencing to identify polymorphic sites.
Genetic analysis: Genotype and allele frequencies are calculated, and Hardy-Weinberg equilibrium is assessed to confirm polymorphism validity .
Association analysis: Statistical methods correlate specific genotypes with quantitative production traits to determine significant associations.
These methodological approaches allow researchers to identify genetic markers with potential application in marker-assisted selection programs aimed at improving egg production in chicken breeding.
The relationship between prolactin gene expression, broodiness, and reproductive performance in chickens represents a complex physiological interplay with significant breed-specific variations. This relationship can be characterized through several key mechanisms:
Physiological mechanisms:
Prolactin levels and reproductive cycles: Elevated prolactin concentrations typically correlate with reduced egg production and the onset of broodiness (incubation behavior). During the reproductive cycle, prolactin levels fluctuate, with dramatic increases observed during incubation phases .
Breed-specific variations: Commercial egg-laying breeds have been selectively bred to minimize broodiness behavior, resulting in lower prolactin responsiveness compared to native breeds, which often exhibit stronger maternal behaviors and consequently reduced egg production during brooding periods .
Molecular regulation: The prolactin gene's expression is regulated by complex promoter elements and transcription factors that respond differently across breeds. Local chicken breeds often experience reproductive system disruption during incubation periods, resulting in fewer eggs laid compared to commercial layers .
Research methodologies:
Researchers investigate these relationships using:
Quantitative PCR: Measuring prolactin mRNA expression levels across different reproductive phases and breeds.
Immunoassays: Tracking circulating prolactin hormone levels throughout reproductive cycles.
Genetic association studies: Correlating specific prolactin gene variants with reproductive performance metrics.
Transcriptional analysis: Examining differences in gene promoter activity and response elements between breeds.
The prolactin gene regulates both egg production and incubation behavior in laying chickens, with genetic polymorphisms potentially serving as selection markers to improve reproductive traits . Understanding these relationships provides valuable insights for breeding programs aimed at optimizing reproductive performance in different chicken populations.
Researchers employ multiple sophisticated methodological approaches to establish evolutionary relationships between chicken prolactin and other species' prolactin molecules:
Comparative sequence analysis methodologies:
Phylogenetic analysis: Constructing evolutionary trees based on nucleotide and amino acid sequences to determine divergence patterns and evolutionary distances between species.
Sequence alignment tools: Using multiple sequence alignment software to identify conserved domains, functional motifs, and species-specific variations.
Comparative genomics: Analyzing syntenic regions and gene arrangements across species to understand evolutionary conservation and divergence.
Molecular clock approaches: Estimating the timing of evolutionary divergence between prolactin variants across species.
Structural biology approaches:
3D structure comparison: Comparing crystal structures or structural models to identify conserved functional domains versus variable regions.
Structure-function relationship studies: Correlating structural conservation with functional conservation across species.
Functional evolutionary implications:
The evolutionary analysis reveals that prolactin and growth hormone belong to the same hormone family and originate from a common ancestor . This shared evolutionary history explains several important functional characteristics:
Structural similarities with cytokines: Both prolactin and growth hormone share structural similarities with cytokines and their receptor superfamilies, suggesting an evolutionary and functional relationship with the immune system .
Signaling pathway conservation: The JAK/STAT, MAPK, and PI3K/AKT signaling pathways activated by these hormones are highly conserved across species, reflecting their fundamental biological importance .
Dual functionality: The evolutionary analysis helps explain why chicken prolactin, while primarily associated with reproduction, also exhibits important immunological functions across various tissues.
This evolutionary perspective provides critical insights into the multifunctional nature of prolactin across species and explains the hormone's diverse roles beyond reproduction in chickens, particularly its involvement in immune system regulation.
Chicken prolactin exerts significant immunomodulatory effects beyond its reproductive functions, with evidence supporting its role in multiple aspects of avian immune regulation. Researchers have uncovered several key mechanisms through methodical investigation:
Established immunological roles:
Immune tissue presence: Prolactin is produced not only by the pituitary but also locally within immune tissues, including the thymus, spleen, and by lymphocytes themselves, suggesting paracrine and autocrine immunoregulatory functions .
Cytokine-like properties: Prolactin shares structural similarities with cytokines and their receptor superfamilies, indicating an evolutionary relationship with immune system mediators .
Signaling pathway activation: Like cytokines, prolactin activates JAK/STAT, MAPK, and PI3K/AKT signaling pathways in immune cells, affecting their development, proliferation, and function .
Immune competence maintenance: Prolactin deficiency can lead to impaired host immune function, suggesting its essential role in maintaining immune system integrity .
Research methodologies for investigation:
Receptor expression analysis: Characterizing prolactin receptor expression patterns across different immune cell populations using flow cytometry, immunohistochemistry, and qPCR.
In vitro immune cell functional assays: Treating isolated chicken immune cells with recombinant prolactin to assess changes in proliferation, cytokine production, and effector functions.
In vivo immune challenge studies: Administering PEGylated or non-PEGylated prolactin to chickens before pathogen challenge to evaluate its impact on immune response parameters and disease outcomes.
Knockout/knockdown approaches: Using molecular techniques to reduce prolactin or prolactin receptor expression in specific tissues to determine functional consequences on immune responses.
These methodological approaches collectively demonstrate that prolactin functions as an important immunoregulatory hormone in chickens, potentially serving as a bidirectional communication mediator between the neuroendocrine and immune systems.
The hypothesis that prolactin (PRL) and its receptor (PRLR) might be involved in viral pathogenesis in poultry represents an emerging research direction with significant implications for avian health. Based on current evidence and mechanistic understanding, researchers have proposed several potential mechanisms:
Proposed mechanisms of involvement:
Viral receptor function: Prolactin receptors might serve as direct viral receptors or co-receptors, facilitating viral entry into host cells . This hypothesis is supported by the observation that certain viral infections alter prolactin levels and receptor expression patterns.
Signaling pathway hijacking: Viruses may exploit prolactin-activated signaling pathways (JAK/STAT, MAPK, PI3K/AKT) to enhance their replication or evade immune responses .
Immunomodulatory effects: Viral infection-induced changes in prolactin levels may contribute to immune dysregulation, potentially benefiting viral persistence or pathogenesis.
Experimental design approaches to test these hypotheses:
Receptor binding studies:
Surface plasmon resonance or co-immunoprecipitation assays to detect direct interactions between viral proteins and PRLR
Competitive binding assays using labeled viruses and soluble PRLR
Cell entry experiments:
PRLR knockdown/knockout in susceptible cell lines followed by viral challenge
Overexpression of PRLR variants to identify domains critical for potential viral interactions
Cell-cell fusion assays to evaluate PRLR's role in membrane fusion events during viral entry
In vivo experimental approaches:
Administration of PEGylated prolactin antagonists before viral challenge
Genetic modification of the PRLR gene using CRISPR/Cas9 to evaluate infection susceptibility
Temporal measurement of prolactin levels during viral infection progression
Comparative studies across viral pathogens:
Systematic evaluation of multiple avian viruses for PRLR-dependency
Correlation between viral tropism and PRLR expression patterns across tissues
These experimental approaches would provide valuable insights into whether prolactin and its receptor play significant roles in viral pathogenesis in poultry, potentially opening new avenues for antiviral intervention strategies.
PEGylated chicken prolactin (PEG-chPRL) offers unique advantages as a research tool for investigating prolonged hormonal effects on immunological parameters. Its extended half-life and modified pharmacokinetic profile make it particularly valuable for studying chronic or long-term immunomodulatory effects that would be difficult to assess with native prolactin.
Methodological applications:
Long-term immune phenotyping studies:
Single administration of PEG-chPRL can maintain elevated prolactin levels for extended periods (>16 hours longer than native chPRL)
Researchers can track changes in immune cell populations, cytokine profiles, and functional responses over multiple days without requiring frequent dosing
This reduces handling stress in experimental animals, which itself can alter immune parameters
Dose-response relationship investigations:
The extended circulation time allows for more stable hormone levels, enabling more accurate assessment of dose-dependent effects
Researchers can establish clearer correlations between sustained prolactin concentrations and specific immunological outcomes
Challenge-recovery experimental designs:
PEG-chPRL administration followed by immune challenge (pathogen exposure, vaccination)
Monitoring of both immediate and delayed immune responses without confounding effects of repeated hormone administration
Assessment of recovery trajectories under sustained prolactin influence
Specific experimental protocol considerations:
The significantly more profound effects of PEG-chPRL on physiological parameters such as corticosteroid levels compared to non-PEGylated chPRL suggest that this research tool may reveal immunological effects that might be missed in short-term studies using native prolactin. This methodological approach allows researchers to better model chronic prolactin elevation scenarios that may occur in certain pathological or physiological states in poultry.
Different immunoassay platforms exhibit varying degrees of sensitivity to macroprolactin, which can significantly impact research findings. Understanding these methodological differences is crucial for selecting the appropriate assay system and interpreting results accurately.
Comparative analysis of immunoassay methods:
The following table summarizes key differences in macroprolactin detection across major immunoassay platforms:
Immunoassay Method | Mean Post-PEG Recovery | Optimal Recovery Cutoff | Sensitivity to Macroprolactin |
---|---|---|---|
AutoDelfia | 65% | 40% | Moderate |
Architect | 65% | 40% | Moderate |
Cobas | Not specified | 40% | Moderate |
Immulite | Higher than average | 50% | High |
Kryptor | 120% | 80% | Low |
Methodological considerations for assay selection:
Research question specificity:
For studies requiring absolute prolactin quantification, methods with lower macroprolactin interference (like Kryptor) may be preferable
For screening studies specifically investigating macroprolactin prevalence, methods with moderate sensitivity are more appropriate
Reference interval approach versus recovery cutoff approach:
Some laboratories use post-PEG monomeric reference intervals
Others report the presence of macroprolactin when post-PEG recovery falls below method-specific cutoffs (typically 40-50%)
Both approaches show similar classification accuracy for most methods, with marginal differences in discordant classification rates
Validation requirements:
PEG precipitation results in co-precipitation of up to 20% of monomeric prolactin
Method-specific reference intervals and recovery cutoffs must be established using a representative healthy population
Validation against gel filtration chromatography (GFC) is recommended for establishing method-specific parameters
Assay interference factors:
These considerations highlight the importance of method-specific validation and standardization when measuring prolactin in research settings, particularly when comparing results across different studies or laboratory platforms.
PEGylated chicken prolactin (PEG-chPRL) represents an advanced research tool with expanding applications in poultry science. Its modified pharmacokinetic profile opens new avenues for investigation while presenting unique methodological challenges that researchers must address.
Advanced research applications:
Reproductive cycle manipulation:
Long-acting PEG-chPRL allows researchers to sustain altered prolactin levels throughout critical phases of the reproductive cycle
This enables detailed investigation of threshold effects and temporal sensitivity to prolactin in breeding behaviors
Applications include studying the relationship between prolonged prolactin elevation and egg production interruption
Neuroendocrine-immune axis studies:
PEG-chPRL facilitates investigation of bidirectional communication between endocrine and immune systems
Researchers can examine how sustained prolactin exposure affects immune cell development and function over extended periods
This approach allows for exploring potential therapeutic applications in immune-related disorders
Developmental biology investigations:
The extended half-life enables studies of prolactin's influence on various developmental processes
Effects on organ development, cellular differentiation, and tissue maturation can be assessed with reduced dosing frequency
This may provide insights into prolactin's role in developmental programming
Methodological challenges and solutions:
Bioactivity standardization:
PEGylation heterogeneity:
Assay interference:
Challenge: PEG moieties can interfere with certain analytical techniques and immunoassays
Solution: Development of PEG-insensitive assays or correction factors for accurate quantification
Species-specific considerations:
Challenge: Findings from mammalian PEGylated hormones may not directly translate to avian systems
Solution: Comparative studies across species and careful validation of each application in the target avian model
Addressing these methodological challenges will enable researchers to fully leverage the advantages of PEG-chPRL for advanced poultry research applications, potentially yielding novel insights into avian physiology, development, and immunology.
An integrated research approach combining genetic, molecular, and physiological methodologies offers the most comprehensive framework for elucidating prolactin's complex roles in poultry. This multidisciplinary strategy connects genotype to phenotype across various biological levels and contexts.
Integrated research methodology framework:
Genetic-level integration:
Genome-wide association studies (GWAS) to identify prolactin pathway-related genetic variants associated with specific traits
Targeted sequencing of prolactin gene regulatory regions across diverse chicken breeds to identify functional polymorphisms
Correlation of specific SNPs (such as g.8052T>C in exon 5) with phenotypic outcomes like egg production
Application of these genetic markers in marker-assisted selection programs
Molecular-level approaches:
Transcriptomic analysis of prolactin-responsive tissues under different physiological states
Proteomic profiling to identify post-translational modifications and protein interaction networks
Epigenetic analysis of prolactin gene regulation under different environmental conditions
Structure-function studies using engineered variants (like PEGylated prolactin) to dissect specific domains and their functions
Physiological and systems-level integration:
Pharmacokinetic/pharmacodynamic modeling of prolactin's effects across multiple systems
Real-time physiological monitoring following administration of PEGylated versus non-PEGylated prolactin
Comparative assessment of breed-specific responses to prolactin manipulation
Network analysis to map interactions between prolactin and other hormonal systems
Integration challenges and solutions:
Data integration across scales:
Challenge: Connecting molecular events to whole-organism phenotypes
Solution: Hierarchical modeling approaches and intermediate phenotyping at tissue/organ levels
Temporal dynamics:
Contextual specificity:
Challenge: Prolactin effects vary by tissue, physiological state, and environmental context
Solution: Factorial experimental designs that systematically vary these parameters
Translational relevance:
Challenge: Connecting basic research findings to practical applications
Solution: Including production-relevant traits in study designs and validating findings in commercial settings
This integrated approach enables researchers to develop comprehensive models of prolactin function that account for genetic variation, molecular mechanisms, and physiological outcomes. Such models can significantly advance our understanding of prolactin's diverse roles in avian biology and inform breeding and management strategies for improved poultry production.
Recombinant chicken prolactin (chPRL) is produced using Escherichia coli (E. coli) as the expression system . The protein is expressed as a single, non-glycosylated polypeptide chain containing 199 amino acids, with an additional alanine at the N-terminal . The recombinant protein is then purified to homogeneity.
PEGylation refers to the process of attaching polyethylene glycol (PEG) chains to a protein or peptide. This modification can enhance the pharmacokinetic properties of the protein, such as increasing its stability and prolonging its circulation time in the bloodstream . For chicken prolactin, PEGylation results in a mono-PEGylated protein (PEG-chPRL) with a molecular mass of approximately 39 kDa .
The biological activity of PEG-chPRL has been evaluated through its interaction with the human prolactin receptor extracellular domain (hPRLR-ECD) and its ability to stimulate PRLR-mediated proliferation in Nb2-11C cells . Although PEG-chPRL exhibits lower activity in vitro compared to non-PEGylated chPRL, it has a significantly prolonged circulation time, remaining in the bloodstream for 16 hours longer than the non-PEGylated form .
The PEGylation of chPRL leads to favorable pharmacokinetic alterations, allowing for reduced dosing frequency due to its prolonged persistence in the circulation . In vivo experiments have shown that PEG-chPRL injections in chickens result in more profound effects on corticosteroid levels in the blood compared to non-PEGylated chPRL .
The development of PEG-chPRL opens new avenues for poultry research, particularly in understanding the hormonal regulation of reproductive behaviors and stress responses. The improved pharmacokinetic properties of PEG-chPRL make it a valuable tool for in vivo studies, potentially reducing the frequency of dosing and improving the overall efficacy of the experiments .
In conclusion, the PEGylation of recombinant chicken prolactin represents a significant advancement in poultry research, offering enhanced stability and prolonged activity. Future research will likely focus on further optimizing the PEGylation process and exploring its applications in various physiological studies.