Prolactin Ovine Antagonist inhibits PRLR signaling via two mechanisms:
Competitive binding: Occupies PRLR sites, blocking native prolactin from initiating signaling cascades .
Prevention of receptor dimerization: The R129G mutation destabilizes the receptor’s extracellular domain, impeding JAK2/STAT5 pathway activation .
High prolactin concentrations (≥500 ng/mL) induce cytotoxicity and oxidative stress in ovine ovarian granulosa cells (GCs) . Prolactin Ovine Antagonist counteracts these effects:
Parameter | 500 ng/mL Prolactin | With Antagonist |
---|---|---|
Cell Viability | ↓ 40% | Restored to baseline |
Reactive Oxygen Species | ↑ 2.5-fold | Normalized |
Apoptosis Rate | 4.01% ± 0.22 | Reduced to 1.60% ± 0.06 |
Antagonist treatment reverses prolactin-induced dysregulation in GCs:
Downregulated pathways: Mitogen-activated protein kinase 12 (MAPK12) signaling, pentose phosphate pathway .
Upregulated proteins: BCL2L1 (anti-apoptotic), UBA52 (ubiquitin-related repair) .
While primarily a research tool, this antagonist informs drug development for hyperprolactinemia. Current ergoline-based therapies (e.g., bromocriptine) carry cardiovascular risks . Prolactin Ovine Antagonist’s non-ergoline structure and targeted action position it as a safer candidate for suppressing prolactinomas or idiopathic hyperprolactinemia .
Storage: Lyophilized form stable at -18°C; reconstituted solution usable for 2–7 days at 4°C .
Carrier proteins: Addition of 0.1% HSA/BSA recommended for long-term storage .
Prolactin Ovine Antagonist is a recombinant protein produced in E. coli that competitively inhibits the binding of endogenous prolactin to its receptors. It consists of a single, non-glycosylated polypeptide chain containing 199 amino acids with a molecular mass of 23kDa . In research settings, this antagonist serves as a valuable tool for investigating prolactin's diverse physiological functions by selectively blocking prolactin signaling pathways. Studies have demonstrated that central infusion of prolactin antagonists can effectively prevent prolactin-mediated effects, such as the maintenance of CS-induced prolactin secretory rhythms in experimental models .
When designing experiments with Prolactin Ovine Antagonist, researchers should consider both the administration route (central vs. peripheral) and duration of treatment, as these factors significantly influence experimental outcomes. For instance, continuous central infusion of a prolactin antagonist for 3 days has been shown to prevent the expression of CS-induced prolactin secretory rhythm, while shorter administration periods (1 day) may not produce the same inhibitory effects .
For optimal experimental outcomes, Prolactin Ovine Antagonist should be reconstituted according to specific parameters that maintain its biological activity. The lyophilized protein powder should be reconstituted in sterile 18MΩ-cm H₂O at a concentration not less than 100μg/ml . This initial stock solution can then be further diluted to the desired working concentrations in appropriate aqueous buffers.
The standard preparation protocol includes:
Allow the lyophilized product to reach room temperature
Add sterile 18MΩ-cm H₂O to achieve a minimum concentration of 100μg/ml
Gently agitate until completely dissolved (avoid vigorous shaking)
Prepare working dilutions in appropriate culture media or buffer solutions
Use reconstituted protein promptly or aliquot and store appropriately
It's worth noting that the formulation typically contains 0.02%-0.03% NaHCO₃ as a stabilizing agent . For experiments requiring precise concentrations, researchers should consider preparing serial dilutions ranging from 4-500 ng/mL, which have been successfully employed in previous studies investigating dose-dependent effects on ovarian granulosa cells .
Prolactin (PRL) exerts significant influence on follicular development, with research demonstrating an inverse relationship between PRL concentration and follicle counts. Regression analysis has shown that as PRL concentration increases, the number of follicles gradually decreases, with R² values exceeding 0.70 for various regression models (linear: 0.709, quadratic: 0.7704, cubic: 0.7795) . This strong correlation underscores prolactin's regulatory role in ovarian function.
In experimental models, Prolactin Ovine Antagonist serves as a crucial tool for parsing the specific contributions of prolactin signaling to follicular development. By selectively blocking prolactin activity, researchers can investigate altered patterns of steroid hormone secretion, particularly estradiol (E₂) and progesterone (P₄), which are significantly affected by prolactin levels . This approach helps elucidate the molecular mechanisms that mediate prolactin's effects on reproductive physiology.
When designing follicular development studies using Prolactin Ovine Antagonist, researchers should consider incorporating techniques such as immunofluorescence staining, cell proliferation assays, and hormone secretion analysis to comprehensively assess antagonist effects on follicular dynamics.
When investigating dose-dependent effects of Prolactin Ovine Antagonist, a methodical experimental design is essential. Based on established protocols, researchers should consider implementing a concentration gradient approach with at least five concentration points (e.g., 0, 4, 20, 100, and 500 ng/mL) to effectively capture the full spectrum of biological responses .
For cell culture experiments, granulosa cells should be seeded at an appropriate density (typically 1×10⁵ cells per well for 24-well plates) and cultured in DMEM/F12 medium supplemented with 10% fetal bovine serum and 1% streptomycin/penicillin mixture . After 24 hours of treatment with varying concentrations of the antagonist, researchers can assess multiple parameters including:
Cell viability using CCK-8 assay
Apoptosis rates using Annexin V-FITC/PI staining and flow cytometry
Oxidative stress markers (MDA, ROS, SOD, T-AOC)
Hormone secretion (E₂ and P₄) via ELISA
Gene expression changes through RT-qPCR
The experimental design should include appropriate controls and a minimum of 3-4 biological replicates to ensure statistical robustness. For interventional studies, researchers should consider both the timing and duration of antagonist administration, as demonstrated by the differential effects observed with 1-day versus 3-day continuous infusion protocols .
Prolactin has been established as a modulator of oxidative stress in ovarian cells, with concentration-dependent effects on multiple oxidative stress markers. When investigating these relationships, researchers should employ a comprehensive panel of oxidative stress assessments, including measurements of reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and total antioxidant capacity (T-AOC) .
Research has demonstrated that high prolactin concentrations (500 ng/mL) induce significant cytotoxicity and oxidative stress in ovine ovarian granulosa cells . When designing experiments to study the protective effects of Prolactin Ovine Antagonist against oxidative damage, researchers should:
Establish baseline oxidative stress parameters in control conditions
Pre-treat cells with varying concentrations of Prolactin Ovine Antagonist
Challenge with known oxidative stress inducers or high prolactin concentrations
Measure multiple oxidative stress markers at different time points
Correlate oxidative stress parameters with cell viability and apoptosis rates
This multi-parameter approach enables researchers to develop a comprehensive understanding of how Prolactin Ovine Antagonist modulates the complex relationship between prolactin signaling and oxidative stress in reproductive tissues.
The molecular mechanisms governing Prolactin Antagonist effects on receptor expression involve complex interactions between long (L-PRLR) and short (S-PRLR) prolactin receptor isoforms. To investigate these mechanisms, researchers should employ both gene knockdown (CRISPR/Cas9) and overexpression approaches.
For CRISPR/Cas9-mediated receptor knockdown, researchers should:
Design appropriate sgRNAs targeting specific PRLR isoforms
Transfect cells with recombinant plasmids using Lipofectamine 3000
Verify knockdown efficiency via RT-qPCR at 48 hours post-transfection
Assess downstream effects on hormone secretion and cell viability
For overexpression studies, lentiviral vectors should be employed with optimized multiplicity of infection (MOI ≈ 400) to achieve efficient transfection . After establishing these modified cellular models, researchers can systematically investigate how Prolactin Ovine Antagonist differentially affects signaling through the altered receptor expression patterns.
The relative expression of receptor isoforms can be quantified using the 2^(-ΔΔCT) method, with appropriate housekeeping genes as internal controls . Functional validation of specific signaling pathways (such as MAPK12) provides additional insights into the molecular mechanisms mediating prolactin antagonist effects.
The administration route of Prolactin Ovine Antagonist significantly influences its physiological effects, particularly regarding prolactin secretory rhythms. Central (intracerebroventricular, icv) and peripheral (systemic) administration produce distinct outcomes that should be carefully considered when designing experiments.
For central administration studies:
Implant guide cannulas into the lateral cerebral ventricle
Allow approximately one week for recovery
Connect osmotic pumps for continuous infusion or perform discrete injections
Collect serial blood samples at appropriate intervals (typically every 2-4 hours)
For peripheral administration:
Insert catheter tubing into the jugular vein
Administer the antagonist systemically at predetermined time points
Collect blood samples according to the same schedule as central administration
Research has demonstrated that continuous central infusion of prolactin antagonist (such as S179D) for 3 days prevents the expression of CS-induced prolactin secretory rhythm, while shorter administration periods (1 day) may not block this rhythm . This temporal dependence highlights the importance of carefully designed administration protocols in prolactin antagonist research.
Integration of proteomic and metabolomic approaches offers comprehensive insights into the molecular mechanisms underlying Prolactin Ovine Antagonist effects. When implementing these advanced analytical techniques, researchers should consider a multi-omics workflow that captures both immediate signaling events and downstream metabolic alterations.
For proteomic analysis:
Perform protein extraction and quantification from control and treated samples
Implement high-resolution LC-MS/MS analysis for protein identification
Apply label-free quantification or iTRAQ/TMT labeling for relative quantification
Conduct bioinformatic analysis to identify differentially expressed proteins
Validate key findings using Western blotting or targeted proteomics
For metabolomic analysis:
Extract metabolites using appropriate solvent systems
Employ both targeted and untargeted metabolomic approaches
Analyze samples using LC-MS or GC-MS depending on the metabolite classes of interest
Identify altered metabolic pathways through pathway enrichment analysis
Correlate metabolite changes with proteomic findings to establish integrated pathway models
These multi-omics approaches have successfully identified key differences between control and high prolactin concentration treatments in ovarian granulosa cells, revealing both cytotoxicity mechanisms and adaptive responses .
CRISPR/Cas9 technology offers unprecedented precision for investigating prolactin receptor signaling pathways. When optimizing this approach for prolactin research, consider the following methodological recommendations:
Design multiple sgRNAs targeting different regions of the PRLR gene to ensure successful editing
Transfect cells at 50-60% confluence to maximize editing efficiency
Verify editing efficiency using both fluorescence observation (at 48 hours) and RT-qPCR
Establish both knockout and knockdown models to investigate dose-dependent effects
Generate cell lines with selective targeting of L-PRLR versus S-PRLR isoforms
The optimal transfection protocol involves seeding 1×10⁵ cells per well in 6-well plates and transfecting with recombinant plasmids using Lipofectamine 3000 . For comprehensive analysis of receptor signaling dynamics, researchers should combine gene editing with pharmacological interventions using Prolactin Ovine Antagonist at varying concentrations.
This integrated approach enables systematic investigation of how specific receptor isoforms mediate the diverse physiological effects of prolactin and how these pathways can be selectively modulated by antagonists.
Temporal dynamics of Prolactin Antagonist effects require carefully designed longitudinal experiments that capture both immediate and delayed responses. When investigating these temporal patterns, researchers should:
Implement time-course experimental designs with multiple sampling points
Combine short-term (24-48 hours) and long-term (3-7 days) observation periods
Utilize continuous monitoring systems where possible (e.g., real-time cell analysis)
Consider both pulsatile and continuous antagonist administration protocols
Correlate temporal changes in cellular responses with alterations in gene expression patterns
Evidence indicates that the duration of prolactin antagonist administration significantly impacts physiological outcomes. For instance, continuous central infusion of a prolactin antagonist for 3 days prevents the expression of CS-induced prolactin secretory rhythm, while shorter administration (1 day) does not block this rhythm .
When designing temporal dynamics studies, researchers should consider implementing automated sampling systems and continuous monitoring technologies to capture transient responses that might be missed with traditional endpoint analyses.
Researchers working with Prolactin Ovine Antagonist may encounter several technical challenges that can affect experimental outcomes. These issues and their solutions include:
Solubility Issues:
Activity Loss During Storage:
Challenge: Decreased antagonist activity after reconstitution
Solution: Prepare fresh solutions or store as single-use aliquots at -80°C to minimize freeze-thaw cycles
Variable Cellular Responses:
Administration Challenges in Animal Models:
Interference with Assay Systems:
Challenge: Antagonist interference with hormone ELISAs
Solution: Include appropriate controls with known concentrations of antagonist to account for potential matrix effects
By anticipating these challenges and implementing the suggested solutions, researchers can enhance the reliability and reproducibility of their Prolactin Ovine Antagonist experiments.
Contradictory findings regarding Prolactin Antagonist effects across different experimental models require careful interpretation that considers multiple factors:
Species-Specific Differences:
Prolactin receptor structure, distribution, and signaling pathways vary significantly between species
When comparing results across species, consider evolutionary conservation of specific pathways rather than absolute effects
Receptor Isoform Expression:
Dose-Response Relationships:
Temporal Considerations:
Methodology Differences:
Variations in cell isolation, culture conditions, and analytical techniques contribute to disparate findings
Standardize methodological approaches and explicitly report all experimental parameters
When encountering contradictory data, researchers should systematically evaluate these factors and consider designing bridging experiments that directly address the specific variables that differ between contradictory studies.
Several emerging technologies offer significant potential for advancing Prolactin Ovine Antagonist research:
Single-Cell Omics:
Single-cell RNA sequencing can reveal cell-specific responses to prolactin antagonists, particularly in heterogeneous tissues like ovaries
This approach enables identification of differential sensitivity across cell populations
CRISPR-Based Epigenome Editing:
Organoid and Microfluidic Systems:
Three-dimensional organoid cultures better recapitulate in vivo tissue architecture and cell-cell interactions
Microfluidic systems allow precise control of antagonist concentrations and continuous monitoring of cellular responses
Multiplexed Biosensors:
Real-time monitoring of multiple signaling pathways simultaneously using fluorescent biosensors
Enables dynamic tracking of antagonist effects on downstream signaling cascades
Artificial Intelligence for Data Integration:
These technologies, especially when used in combination, promise to provide unprecedented insights into the complex mechanisms through which Prolactin Ovine Antagonist modulates physiological processes.
Despite significant advances, several crucial questions regarding Prolactin Ovine Antagonist remain unresolved:
Receptor Subtype Selectivity:
Do current antagonists exhibit preferential binding to specific prolactin receptor isoforms?
How can antagonist selectivity be engineered to target specific signaling pathways?
Blood-Brain Barrier Dynamics:
Temporal Signaling Networks:
Oxidative Stress Mechanisms:
Therapeutic Potential:
Could Prolactin Ovine Antagonist serve as a model for developing therapeutics for prolactin-related disorders?
What modifications would be necessary to optimize pharmacokinetic properties while maintaining antagonist activity?
Addressing these questions will require interdisciplinary approaches combining molecular biology, structural biology, systems pharmacology, and translational research methodologies.
Prolactin Ovine Antagonist Recombinant is designed to act as an antagonist to prolactin, meaning it binds to prolactin receptors but does not activate them. This can be useful in research and therapeutic contexts where inhibition of prolactin activity is desired. The antagonist form of prolactin can help in studying the hormone’s role in various physiological processes and in developing treatments for conditions where prolactin’s activity needs to be modulated .
The recombinant form of prolactin, including its antagonist, is widely used in laboratory research to study prolactin’s effects on cell proliferation, differentiation, and other cellular processes. It is also used in the development of therapeutic strategies for conditions such as prolactinomas (prolactin-secreting tumors), breast cancer, and other disorders related to prolactin dysregulation .
Lyophilized (freeze-dried) Prolactin Ovine Antagonist Recombinant is stable at room temperature for up to three weeks, but for long-term storage, it should be kept desiccated below -18°C. Upon reconstitution, it should be stored at 4°C for short-term use (2-7 days) and below -18°C for long-term use. It is recommended to add a carrier protein (such as 0.1% HSA or BSA) to prevent freeze-thaw cycles .