LHRH Human

Luteinizing Hormone Releasing Hormone Human
Shipped with Ice Packs
In Stock

Description

Lutenizing Hormone Releasing Hormone Human Synthetic is a single, non-glycosylated, polypeptide chain containing 10 amino acids and having a molecular mass of 1182.2 Dalton. The molecular formula is C55H75N17O13.C2H4O2. The CAS Number is 71447-49-9.

Product Specs

Introduction
Gonadotropin-releasing hormone 1 (GNRH1), also called Luteinizing-hormone releasing hormone (LHRH), is a hormone that triggers the release of other hormones, namely follicle-stimulating hormone (FSH) and luteinizing hormone (LH), from the pituitary gland. GNRH1 itself is produced and released by a part of the brain called the hypothalamus. When GNRH1 reaches the pituitary gland, it stimulates the production and release of FSH and LH. The pattern of GNRH1 release, in terms of the amount and frequency, influences how much FSH and LH are released. Additionally, hormones like androgens and estrogens also provide feedback that affects this process. Low-frequency pulses of GNRH1 mainly lead to FSH release, while high-frequency pulses primarily trigger LH release. The way GNRH1 is secreted differs between males and females. In males, GNRH1 is released in pulses at a consistent frequency. However, in females, the frequency of these pulses changes throughout the menstrual cycle, with a significant surge in GNRH1 right before ovulation. The pulsatile release of GNRH1 is essential for proper reproductive function in all vertebrates. This single hormone plays a crucial role in a complex series of events in females, including the growth of follicles (which contain eggs), the release of an egg (ovulation), and the maintenance of the corpus luteum (a structure that forms after ovulation). In males, GNRH1 is vital for the production of sperm.
Description
Synthetic human Lutenizing Hormone Releasing Hormone is a single chain molecule, not containing any sugar molecules, composed of 10 amino acids. Its molecular weight is 1182.2 Daltons, and its chemical formula is C55H75N17O13.C2H4O2. It is uniquely identified by the CAS Number 71447-49-9.
Physical Appearance
It appears as a white powder, sterilized by filtration and dried by freezing (lyophilized).
Formulation
The LHRH, in a concentrated solution of 1 mg/ml, undergoes a freeze-drying process to remove water, resulting in a powder form. No additional substances are added during this process.
Solubility
To reconstitute the freeze-dried LHRH, it is recommended to dissolve it in sterile water with a pH of 9 (adjusted using sodium hydroxide) at a concentration not less than 100 micrograms per milliliter. This solution can then be diluted further with other aqueous solutions if needed. Alternatively, LHRH can also be dissolved in a 1% solution of glacial acetic acid.
Stability
Lyophilized LHRH, though stable at room temperature for up to 3 weeks, should ideally be stored in dry conditions at a temperature below -18°C. Once reconstituted, LHRH can be stored at 4°C for 2-7 days. For longer storage periods, it should be kept at or below -18°C. To further enhance long-term storage stability, adding a carrier protein like 0.1% HSA or BSA is recommended. Repeated cycles of freezing and thawing should be avoided.
Purity
The purity is determined to be greater than 98.0% using a technique called Reverse Phase High Performance Liquid Chromatography (RP-HPLC).
Synonyms
Progonadoliberin-1, Progonadoliberin I, LHRH, GRH, GNRH, LNRH.
Amino Acid Sequence
Pyr-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH2.

Q&A

What is the biological role of human LHRH in reproductive physiology?

Human LHRH is a hypothalamic peptide involved in the regulation of reproductive functions by controlling the synthesis and release of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the anterior pituitary gland . These gonadotropins then act on the gonads (testes in males, ovaries in females) to stimulate sex hormone production and gametogenesis. LHRH is released in a pulsatile manner from the hypothalamus, with the frequency and amplitude of these pulses determining the differential secretion of LH and FSH. This pulsatile secretion pattern is critical for maintaining normal reproductive function, as continuous exposure to LHRH leads to receptor desensitization and downregulation, which forms the basis for therapeutic applications of LHRH agonists.

How do LHRH analogs differ in their mechanism of action?

LHRH agonists and antagonists both target LHRH receptors but operate through fundamentally different mechanisms:

LHRH Agonists:

  • Initially activate LHRH receptors, causing a temporary surge (flare) in LH, FSH, and subsequently testosterone

  • With continued administration, cause receptor desensitization and downregulation

  • Eventually lead to suppression of gonadotropin release and sex hormone production

  • Examples include Zoladex (goserelin) and Buserelin

LHRH Antagonists:

  • Immediately block LHRH receptor signaling without initial activation

  • Cause rapid and sustained inhibition of LH, FSH, and testosterone without the initial surge

  • Do not require receptor downregulation for therapeutic effect

  • Provide more immediate hormone suppression

The choice between agonists and antagonists depends on the clinical context, with antagonists preferred when rapid hormone suppression is needed or when the initial flare response could be detrimental to the patient.

What experimental methods are recommended for detecting LHRH and its receptors?

Multiple complementary approaches are recommended for comprehensive LHRH and LHRH receptor detection:

  • RT-PCR for mRNA detection:

    • Highly sensitive for detecting LHRH receptor transcript expression

    • Can detect low copy numbers using appropriate primers

    • Studies have successfully detected LHRH-R-I mRNA in 83% of bladder cancer specimens

  • Immunohistochemistry (IHC):

    • Allows visualization of receptor distribution within tissues

    • Can assess heterogeneity of expression across tumor sections

    • Permits correlation with histopathological features

    • Studies show variable intensity of LHRH-R expression correlated with tumor grade

  • Radioligand binding assays:

    • Gold standard for confirming functional receptor protein expression

    • Provides quantitative data on receptor density and binding affinity

    • Can distinguish specific from non-specific binding

  • Western blot analysis:

    • Confirms receptor protein expression and molecular weight

    • Allows semi-quantitative comparison between samples

A comprehensive approach using multiple methods is strongly recommended for conclusive receptor characterization. For example, studies on bladder cancer employed RT-PCR, IHC, and radioligand binding assays in combination to convincingly demonstrate LHRH receptor expression .

What are the key considerations for designing experiments to study LHRH effects?

When designing experiments to study LHRH effects, researchers should consider several important factors:

  • Experimental design selection:

    • Between-subjects designs: Different treatment groups receive different LHRH analogs or concentrations

    • Within-subjects designs: Same cell population measured at different time points

  • Control conditions:

    • Vehicle controls are essential for comparisons

    • Include non-LHRH receptor expressing cells as negative controls

    • Use competitive binding with antagonists to confirm specificity of observed effects

  • Random assignment:

    • Ensure random distribution of cells/animals to different treatment conditions

    • This controls for extraneous variables and reduces systematic bias

  • Time course considerations:

    • LHRH effects may vary over time, particularly with agonists

    • Antiproliferative effects of LHRH agonists on DU 145 cells were observed after 4 days of treatment

  • Dose-response relationships:

    • Test multiple concentrations to establish dose-dependency

    • Include both sub-therapeutic and supra-therapeutic doses

  • Verification of receptor-mediated effects:

    • Co-administration of antagonists with agonists to confirm specificity

    • For example, the inhibitory action of Zoladex and Buserelin on DU 145 cells was completely counteracted by LHRH antagonist co-treatment

  • Appropriate outcome measures:

    • Select endpoints (proliferation, apoptosis, gene expression) based on specific research questions

    • Include multiple endpoints when possible to capture comprehensive effects

How can researchers distinguish direct LHRH effects from indirect hormonal effects?

Distinguishing direct LHRH effects from indirect hormonal effects requires careful experimental design:

  • In vitro systems:

    • Use hormone-free media with charcoal-stripped serum

    • Employ cell models that lack gonadotropin receptors but express LHRH receptors

    • Create co-culture experiments with/without pituitary cells to compare direct vs. indirect pathways

  • Receptor blocking studies:

    • Use specific LHRH receptor antagonists to block direct effects

    • Compare outcomes with/without antagonist co-treatment

    • The antiproliferative effects of LHRH agonists on DU 145 cells were completely counteracted by LHRH antagonist co-treatment, confirming direct receptor-mediated action

  • Receptor expression manipulation:

    • siRNA knockdown of LHRH receptors

    • Receptor overexpression studies

    • Comparison of effects in receptor-positive vs. receptor-negative cells

  • Signaling pathway analysis:

    • Monitor immediate post-receptor signaling events

    • Conduct time-course studies (rapid effects likely direct, delayed effects may be indirect)

    • Implement pathway inhibitor studies

  • In vivo approaches:

    • Tissue-specific receptor knockout models

    • Hypophysectomized animal models (lacking pituitary)

    • Compare local vs. systemic administration of LHRH analogs

The study showing that LHRH antagonist treatment of DU 145 cells cultured in serum-free conditions resulted in increased cell proliferation strongly suggests a direct inhibitory role of LHRH in local regulation of cancer cell growth, independent of systemic hormonal effects .

What statistical approaches are most appropriate for analyzing LHRH receptor expression data?

When analyzing LHRH receptor expression data across different samples and conditions, researchers should implement these statistical approaches:

  • Correlation analyses:

    • Pearson or Spearman correlation for examining relationships between continuous variables

    • In bladder cancer studies, Pearson analysis showed LHRH receptor expression was negatively correlated (r = -0.91) with pathological grade

  • Categorical data analysis:

    • Chi-square or Fisher's exact test for comparing proportions of receptor-positive cases across groups

    • Odds ratios to quantify associations between receptor status and clinical parameters

  • Continuous expression analysis:

    • ANOVA or Kruskal-Wallis for comparing expression levels across multiple groups

    • t-tests or Mann-Whitney U tests for pairwise comparisons

    • ANCOVA to adjust for covariates (age, tumor size, etc.)

  • Multivariable approaches:

    • Multiple regression to adjust for confounding variables

    • Principal component analysis to reduce dimensionality when analyzing multiple receptor types

  • Advanced statistical considerations:

    • Adjust for multiple comparisons (Bonferroni, Benjamini-Hochberg, etc.)

    • Account for intra-tumor heterogeneity with mixed-effects models

    • Implement bootstrapping for robust confidence intervals when sample sizes are small

The approach used in bladder cancer research, revealing a negative correlation between LHRH receptor expression and tumor grade, demonstrates how correlation analysis can reveal important biological relationships . Selection of statistical methods should be guided by the specific data type, sample size, and research question.

What evidence supports LHRH receptor expression in different cancer types?

Evidence for LHRH receptor expression has been documented across multiple cancer types using various detection methods:

Prostate Cancer:

  • Functional LHRH receptors confirmed in both androgen-dependent and androgen-independent prostate cancer cell lines

  • LHRH receptor expression demonstrated in primary prostate cancer tissue samples

  • Expression found in DU 145 cells (androgen-independent prostate cancer cell line)

Bladder Cancer:

  • LHRH-R-I mRNA detected in 20 of 24 (83%) human bladder cancer specimens

  • Protein expression confirmed by immunohistochemistry in all examined samples

  • Radioligand binding studies demonstrated specific LHRH-R-I binding

  • Expression appears negatively correlated with tumor grade (r = -0.91)

The following table summarizes LHRH-R expression data from bladder cancer studies:

Detection MethodPositive ResultsTotal SamplesPositive (%)
LHRH-R-I mRNA202483%
LHRH-I mRNA192479%
Both LHRH and LHRH-R152462.5%
IHC for LHRH-R1212100%

The expression of LHRH receptors in cancer tissues outside the reproductive system suggests potential broader applications for LHRH-targeted therapies beyond traditional hormone-dependent cancers .

How do LHRH agonists exert direct antiproliferative effects on cancer cells?

LHRH agonists can exert direct antiproliferative effects on cancer cells through several receptor-mediated mechanisms independent of their action on the pituitary-gonadal axis:

  • Direct receptor-mediated growth inhibition:

    • LHRH agonists Zoladex and Buserelin demonstrated significant and dose-dependent antiproliferative action on DU 145 prostate cancer cells after 4 days of treatment

    • This effect was receptor-mediated, as it was completely counteracted by co-treatment with an LHRH antagonist

  • Interference with growth factor signaling:

    • LHRH receptor activation can inhibit EGF-induced mitogenic signaling

    • Downregulation of growth factor receptors

    • Inhibition of tyrosine kinase activity

  • Cell cycle regulation:

    • Induction of cell cycle arrest, often in G0/G1 phase

    • Modulation of cyclins and cyclin-dependent kinases

  • Apoptosis induction:

    • Activation of pro-apoptotic pathways

    • Downregulation of anti-apoptotic proteins

The evidence for direct antiproliferative effects includes the discovery of low-affinity binding sites for LHRH agonists on cancer cell membranes, particularly evident when cells are cultured in serum-free conditions . The identification of autocrine/paracrine LHRH loops in androgen-independent prostate cancer cells further supports these direct actions .

What is the significance of autocrine/paracrine LHRH systems in cancer tissues?

The discovery of autocrine/paracrine LHRH systems in cancer tissues has profound implications for understanding cancer biology and developing new therapeutic approaches:

  • Evidence for autocrine/paracrine systems:

    • RT-PCR has demonstrated mRNA for LHRH expression in cancer cells, including DU 145 prostate cancer cells and bladder cancer cells

    • Co-expression of both LHRH and its receptor within the same tumor tissue (62.5% of bladder cancer samples expressed both LHRH and LHRH-R)

    • Treatment with LHRH antagonists in serum-free conditions increases cancer cell proliferation, suggesting an inhibitory role for endogenous LHRH

  • Functional significance:

    • Local growth regulation independent of systemic hormonal control

    • Potential tumor-suppressive role in some contexts

    • Possible involvement in tumor differentiation (negative correlation between LHRH-R expression and tumor grade in bladder cancer)

    • May represent an endogenous growth-limiting mechanism

  • Therapeutic implications:

    • Potential for LHRH analog therapy even in cancers not traditionally considered hormone-dependent

    • Rationale for developing cytotoxic LHRH analogs for targeted therapy

    • May explain differential responses to LHRH agonists vs. antagonists in some cancer types

These data indicate that LHRH may function as a local growth regulator in various tissues beyond its classical role as a reproductive hormone, opening new avenues for therapeutic intervention in cancer treatment.

How can cytotoxic LHRH analogs be developed for targeted cancer therapy?

The development of cytotoxic LHRH analogs represents an advanced approach to targeted cancer therapy, leveraging LHRH receptors as molecular targets for delivering cytotoxic payloads:

  • Design considerations:

    • Selection of appropriate LHRH analog (agonist or antagonist) as targeting moiety

    • Choice of cytotoxic agent (e.g., doxorubicin, docetaxel derivatives)

    • Linker chemistry (stable in circulation, cleavable in target cells)

    • Optimization of drug-to-peptide ratio

    • AN-207 is an example of a cytotoxic LHRH analog investigated for PC-82 human prostate cancer

  • In vitro evaluation:

    • Comparison of cytotoxicity in LHRH receptor-positive vs. negative cells

    • Demonstration of receptor-mediated uptake (competition with unconjugated LHRH)

    • Mechanism of action studies (apoptosis, cell cycle effects)

    • Resistance profiling

  • In vivo studies:

    • Pharmacokinetics and biodistribution

    • Efficacy in appropriate tumor models

    • Toxicology studies (off-target effects, maximum tolerated dose)

    • Comparison with unconjugated cytotoxic agent and non-targeted controls

  • Clinical translation:

    • Phase I/II trials to assess safety, pharmacokinetics, and preliminary efficacy

    • Patient selection strategies based on LHRH receptor expression

    • Combination strategies with other therapeutic modalities

The high incidence of LHRH receptors in cancers such as bladder cancer (83% at mRNA level) suggests their potential as molecular targets for such targeted therapies . The negative correlation between LHRH receptor expression and tumor grade observed in bladder cancer indicates that patient selection might be important for optimizing outcomes with these approaches.

How do LHRH receptors in cancer cells differ from those in pituitary cells?

LHRH receptors in cancer cells exhibit several important differences from their pituitary counterparts in terms of signaling mechanisms, regulation, and functional outcomes:

  • Signaling pathway differences:

    • Pituitary LHRH-R primarily couples to Gq/11 proteins, activating phospholipase C, increasing IP3/DAG and intracellular calcium

    • Cancer LHRH-R may couple to different G-proteins or utilize G-protein-independent signaling

    • In cancer cells, LHRH-R activation often leads to inhibition of growth factor receptor signaling rather than stimulation of hormone release

  • Receptor regulation:

    • Pituitary LHRH-R undergoes homologous desensitization with prolonged agonist exposure

    • Cancer LHRH-R may show altered desensitization kinetics or mechanisms

    • Differences in receptor internalization, recycling, and degradation pathways

  • Functional outcomes:

    • Pituitary: stimulation of gonadotropin synthesis and release

    • Cancer cells: often antiproliferative, pro-apoptotic, or anti-metastatic effects

    • Context-dependent responses based on cellular environment

  • Therapeutic implications:

    • Differential sensitivity to LHRH agonists vs. antagonists

    • Potential for cancer-selective targeting based on receptor differences

    • Opportunities for developing tissue-specific LHRH analogs

Understanding these differences is crucial for developing targeted therapeutic approaches and predicting clinical responses. The observation that LHRH agonists exert direct antiproliferative effects on cancer cells while stimulating hormone release from pituitary cells highlights the context-dependent nature of LHRH receptor signaling .

What methodological approaches can overcome challenges in LHRH receptor detection?

Researchers often encounter technical challenges when detecting LHRH receptors. These methodological approaches can address common problems:

  • For low signal-to-noise ratio in IHC:

    • Optimize antigen retrieval methods (test different pH buffers, EDTA vs. citrate)

    • Implement signal amplification systems (tyramide, polymer-based detection)

    • Use positive control tissues with known high expression

    • Test multiple antibodies targeting different receptor epitopes

    • Employ stringent blocking protocols to reduce background

  • For discrepancies between mRNA and protein detection:

    • Verify primer specificity for RT-PCR (sequence validation)

    • Test for presence of splice variants

    • Consider post-transcriptional regulation mechanisms

    • Use freshly prepared samples to minimize degradation

  • For variability in radioligand binding assays:

    • Optimize membrane preparation protocols

    • Verify ligand specificity with competition assays

    • Control temperature and incubation conditions precisely

    • Account for non-specific binding with parallel assays

  • For inconsistent results between methods:

    • Standardize sample preparation across methodologies

    • Consider functional detection methods (calcium imaging, cAMP assays)

    • Implement multi-method validation protocols

    • Report results from multiple methodologies, as seen in bladder cancer studies

The comprehensive approach used in the bladder cancer study, which employed RT-PCR, IHC, and radioligand binding assays, represents a best practice for robust LHRH receptor detection and characterization .

How should researchers interpret contradictory findings in LHRH research?

When faced with contradictory findings in LHRH research, researchers should consider several factors to resolve discrepancies and interpret results appropriately:

  • Methodological differences:

    • Detection methods (sensitivity, specificity, threshold definitions)

    • Experimental conditions (in vitro vs. in vivo, culture conditions)

    • Timing of measurements (acute vs. chronic exposure)

    • Species differences (human vs. animal models)

    • Consider how experimental design affects outcomes

  • Sample heterogeneity:

    • Cell type variations within tissues

    • Patient characteristics (age, sex, comorbidities)

    • Prior treatments affecting receptor expression

    • Disease stage and grade (as seen in bladder cancer, where LHRH-R expression negatively correlates with grade)

    • Genetic and epigenetic variations

  • Context-dependent signaling:

    • Receptor coupling to different G-proteins in different cellular contexts

    • Cross-talk with other signaling pathways

    • Variations in downstream effector availability

    • Influence of tumor microenvironment

  • Receptor variants and isoforms:

    • Expression of different LHRH receptor subtypes or splice variants

    • Post-translational modifications affecting function

    • Altered trafficking or localization

When interpreting studies with contradictory results, researchers should systematically evaluate these factors and consider performing integrative analyses that combine data across multiple studies. The use of multiple complementary methods, as demonstrated in the bladder cancer study, provides a robust approach to validate findings and resolve potential contradictions .

What research gaps remain in understanding LHRH receptor function in cancer?

Despite significant advances, important research gaps remain in understanding LHRH receptor function in cancer:

  • Receptor heterogeneity:

    • Comprehensive characterization of LHRH receptor subtypes across cancer types

    • Functional significance of receptor splice variants

    • Correlation between receptor subtypes and treatment response

    • Spatial and temporal dynamics of receptor expression during cancer progression

  • Signaling pathway intricacies:

    • Complete mapping of cancer-specific LHRH receptor signaling networks

    • Crosstalk mechanisms with other receptor systems

    • Identification of key nodes determining antiproliferative vs. proliferative effects

    • Tissue-specific signaling differences

  • Resistance mechanisms:

    • Molecular basis of resistance to LHRH analog therapy

    • Compensatory pathways activated upon LHRH receptor blockade

    • Biomarkers predictive of resistance development

    • Strategies to overcome or prevent resistance

  • Autocrine/paracrine system regulation:

    • Factors controlling local LHRH production in cancer cells

    • Regulatory mechanisms of LHRH receptor expression

    • Complete characterization of LHRH-like peptides in the tumor microenvironment

    • Interaction between autocrine LHRH systems and other local growth regulators

  • Clinical translation challenges:

    • Optimal patient selection criteria for LHRH-targeted therapies

    • Predictive biomarkers for response to different LHRH analogs

    • Combination strategies to enhance efficacy

    • Long-term effects of LHRH analog therapy

Addressing these gaps will require integration of advanced technologies and cross-disciplinary approaches combining molecular biology, pharmacology, systems biology, and clinical investigation.

What emerging technologies will advance LHRH research in the next decade?

Several emerging technologies are poised to significantly advance LHRH research in the coming decade:

  • Single-cell and spatial technologies:

    • Single-cell RNA sequencing for receptor heterogeneity characterization

    • Spatial transcriptomics to map receptor distribution in tissue context

    • Digital spatial profiling for quantitative spatial analysis of LHRH receptors

    • Resolution of intratumoral heterogeneity of receptor expression

  • Advanced imaging approaches:

    • Super-resolution microscopy for receptor clustering analysis

    • Live-cell imaging of LHRH receptor trafficking and internalization

    • Whole-body imaging with receptor-specific probes

    • Multiplexed imaging for simultaneous detection of multiple signaling components

  • CRISPR-based technologies:

    • Precise genome editing of LHRH receptor genes

    • CRISPR activation/interference for receptor expression modulation

    • CRISPR screens to identify synthetic lethal interactions with LHRH pathway

    • Creation of improved in vivo models

  • Organoid and microphysiological systems:

    • Patient-derived organoids for personalized therapy testing

    • Hypothalamic-pituitary-gonadal axis-on-a-chip

    • Tumor microenvironment models incorporating hormone gradients

    • Testing of LHRH analogs in physiologically relevant 3D systems

  • Artificial intelligence and machine learning:

    • Predictive models for LHRH analog efficacy

    • Image analysis algorithms for receptor quantification

    • AI-designed LHRH analogs with optimized properties

    • Integration of multi-omic data for comprehensive pathway analysis

These technologies will enable more precise, comprehensive, and dynamic understanding of LHRH biology across multiple scales, from molecular interactions to systemic effects, potentially leading to breakthrough advances in both basic science understanding and therapeutic applications.

Product Science Overview

Introduction

Luteinizing Hormone Releasing Hormone (LHRH), also known as Gonadotropin-Releasing Hormone (GnRH), is a pivotal hormone in the regulation of the reproductive system. It is a decapeptide produced in the hypothalamus and plays a crucial role in the control of the secretion of two other important hormones: Luteinizing Hormone (LH) and Follicle-Stimulating Hormone (FSH) from the anterior pituitary gland .

Structure and Function

LHRH is composed of ten amino acids, making it a decapeptide. Its primary function is to stimulate the anterior pituitary gland to release LH and FSH. These hormones are essential for the proper functioning of the gonads (ovaries in females and testes in males). In females, LH and FSH regulate the menstrual cycle and ovulation, while in males, they are involved in the production of testosterone and spermatogenesis .

Mechanism of Action

The release of LHRH is pulsatile, meaning it is released in bursts rather than a steady stream. This pulsatile release is crucial for the proper functioning of the reproductive system. Continuous release of LHRH, on the other hand, can lead to desensitization of the pituitary gland and a subsequent decrease in LH and FSH secretion .

Clinical Significance

LHRH analogs, both agonists and antagonists, have been developed for clinical use. These analogs are used in the treatment of hormone-sensitive cancers such as prostate and breast cancer, as well as in assisted reproductive technologies (ART) to control ovulation .

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.