CGB Human

CGB Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to CGB Human

Chorionic Gonadotropin Beta (CGB) is a glycoprotein subunit critical for human chorionic gonadotropin (hCG), a hormone essential for pregnancy. Produced by trophoblastic cells in the placenta, CGB forms a heterodimer with a common alpha subunit (α-hCG) to mediate luteal progesterone production, angiogenesis, and immunotolerance during early pregnancy .

Biological Role and Function

CGB’s beta subunit confers functional specificity to hCG, enabling it to bind hCG/LH receptors on ovarian and endometrial cells. Key roles include:

  • Progesterone Maintenance: Stimulation of corpus luteum to sustain pregnancy .

  • Angiogenesis and Placental Development: Promotion of uterine vascularization and syncytiotrophoblast differentiation .

  • Immunotolerance: Suppression of maternal immune responses to fetal cells .

CGB is the first molecule synthesized by the embryo as early as the eight-cell stage, with RNA detected in blastocysts .

Genetic and Evolutionary Insights

The CGB gene cluster on chromosome 19q13.33 includes six genes (CGB1-CGB8), four of which encode functional CGB isoforms.

GeneFunctionalitySequence SimilarityNotes
CGBFunctional97-99% (CGB5, CGB7, CGB8)Expressed in placental trophoblasts
CGB1/CGB2Pseudogenes85% (CGB)Result from frameshift mutations; encode truncated 132-a.a. proteins
LHBFunctional94% (CGB)Encodes luteinizing hormone beta subunit

Key Evolutionary Note: CGB genes originated from duplication of LHB and share high homology with great apes .

Clinical and Research Applications

CGB serves as a biomarker in oncology and reproductive health:

**5.1. Cancer Detection

CGB expression is elevated in gynecological tumors (e.g., ovarian, endometrial, cervical cancers) and circulating tumor cells (CTCs) .

Cancer TypeCGB ExpressionMethodClinical Relevance
Ovarian68% Urinary beta-coreTumor burden/prognosis
Endometrial51% Real-time RT-PCRMetastasis monitoring
Cervical46% Serum assaysEarly detection

CTC Detection: Real-time RT-PCR identifies CGB transcripts in peripheral blood, distinguishing cancer patients from controls .

**5.2. Reproductive Health

CGB is used to diagnose ectopic pregnancies and monitor hCG levels during assisted reproductive technologies (ART) .

Production and Stability

Recombinant CGB is produced in E. coli with a His-tag for purification .

ParameterDetails
Purity>95% (chromatographic techniques)
Storage-20°C (long-term); 4°C (short-term)
Stabilizers20 mM Tris-HCl (pH 8.0), 0.4M urea, 10% glycerol
HandlingAvoid freeze-thaw cycles; add 0.1% HSA/BSA for long-term storage

Research Findings and Studies

Recent studies highlight CGB’s role in tumor biology:

**7.1. Circulating Tumor Cells

A 2024 study used CGB and GNRH1 mRNA to detect CTCs in gynecological cancers, achieving 93% sensitivity for CGB .

**7.2. Gene Expression Patterns

In silico analysis of CGB1/CGB2 promoters identified transcription factor binding sites (e.g., SP1, AP2), suggesting regulatory roles in placental development .

Product Specs

Introduction
Glycoprotein hormones are made up of two subunits: a common alpha subunit and a distinct beta subunit that determines its biological function. CGB, part of the glycoprotein hormone beta chain family, serves as the beta 3 subunit of Chorionic Gonadotropin (CG). Produced by trophoblastic cells in the placenta, CG stimulates the ovaries. The gene coding for CG's beta subunit is one of six genes arranged in tandem and inverted pairs on chromosome 19q13.3, adjacent to the gene for the luteinizing hormone beta subunit.
Description
Recombinant Human CGB, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 168 amino acids (specifically, amino acids 21 to 165). It has a molecular mass of 17.9 kDa. This CGB protein is engineered with a 23 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The CGB protein solution is provided at a concentration of 0.5 mg/ml and contains 20mM Tris-HCl buffer (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freezing and thawing.
Purity
Purity exceeds 85.0% as determined by SDS-PAGE analysis.
Synonyms

CGB3, CGB5, CGB7, CGB8, hCGB,  CG-beta, CGB.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSSKEPLRP RCRPINATLA VEKEGCPVCI TVNTTICAGY CPTMTRVLQG VLPALPQVVC NYRDVRFESI RLPGCPRGVN PVVSYAVALS CQCALCRRST TDCGGPKDHP LTCDDPRFQD SSSSKAPPPS LPSPSRLPGP SDTPILPQ.

Q&A

What is CBG and how does it interact with human biological systems?

Cannabigerol (CBG) is a non-psychoactive cannabinoid found in Cannabis sativa, often referred to as "the mother of all cannabinoids" due to its role as a precursor to many other cannabinoids. In human biological systems, CBG exhibits multiple receptor interactions:

Receptor TypeInteraction TypePotential Physiological Effects
CB1 and CB2 (G-protein-coupled)AgonistModulation of endocannabinoid system
α2 adrenoceptorsStimulatorySympathetic nervous system effects
TRPV8AntagonistPotential anti-cancer effects (prostate), pain reduction

These interactions occur throughout the body as endocannabinoid receptors are present not only in the brain but also in bones, skeletal muscle, skin, fat tissue, immune cells, liver, pancreas, heart, blood vessels, gastrointestinal tract, and kidney .

What is the Clinical Genetics Branch (CGB) and what are its primary research areas?

The Clinical Genetics Branch (CGB) is a research division within the National Cancer Institute that studies individuals and populations at high genetic risk of cancer. Its multidisciplinary approach combines clinical, genetic, genomic, epidemiologic, behavioral, statistical, and laboratory scientific research modalities .

Primary research areas include:

  • Clinical, genetic, and genomic cancer research

  • Etiologic studies with potential clinical applications

  • Population-based early detection and cancer prevention strategies

  • Management of cancer in high-risk families and populations

The Clinical Epidemiology Unit (CEU) within CGB specifically conducts research with potential clinical and public health applications and leads studies evaluating population-based early detection and cancer prevention strategies .

How do human cytochrome P450 enzymes metabolize CBG?

Human cytochrome P450 enzymes play a critical role in CBG metabolism through the following process:

  • Several specific P450 isoforms (CYP2J2, CYP3A4, CYP2D6, CYP2C8, and CYP2C9) are primarily responsible for CBG metabolism

  • The major metabolite formed is cyclo-CBG through oxidation at the prenyl chain

  • Metabolism occurs preferentially at the prenyl chain rather than other positions

  • Both in vitro and in vivo studies confirm cyclo-CBG as the major metabolite

Research indicates that both CBG and its oxidized metabolites demonstrate anti-inflammatory activity in human cells, specifically reducing inflammation in BV2 microglial cells stimulated with LPS .

What are Single-Case Experimental Designs (SCEDs) and how are they applied in human research?

Single-Case Experimental Designs (SCEDs) are multiphase experimental designs where extensive data is collected on individual subjects who serve as their own controls. They are particularly valuable for identifying optimal treatments for individual people rather than relying on group averages .

Key characteristics of SCEDs include:

Design ElementMethodological RequirementsResearch Implications
Phase structureMinimum 5 data points per phaseEnsures sufficient data for analysis
Stability criteriaData points within 15% of phase medianControls for natural variability
ReplicationMinimum of 3 replications of treatment effectsStrengthens causal inferences
RandomizationOrder of presentation can be randomizedEnhances experimental control

Three primary SCED types used in human research are:

  • Reversal designs (e.g., ABAB) - comparing baseline (A) to treatment (B) phases repeatedly

  • Multiple baseline designs - staggering treatment across participants

  • Combined designs - incorporating elements of both approaches

How can researchers differentiate between direct CBG effects and those of its metabolites in human studies?

Differentiating between direct CBG effects and those of its metabolites requires sophisticated methodological approaches:

  • Isolated metabolite testing: Synthesize and test purified CBG metabolites (particularly cyclo-CBG) against parent CBG

  • Enzyme inhibition studies: Employ selective cytochrome P450 inhibitors while monitoring biological responses

  • Pharmacokinetic-pharmacodynamic modeling: Correlate plasma concentrations of both CBG and metabolites with biological effects

  • Receptor binding assays: Compare binding affinities to relevant receptors (CB1, CB2, α2 adrenoceptors, TRPV8)

  • Time-course analysis: Examine temporal relationships between administration, metabolite formation, and responses

This comprehensive approach helps establish whether observed effects are attributable to the parent compound or its metabolites, which is essential for accurate interpretation of research findings.

What statistical considerations are most important when analyzing SCEDs in human research?

When analyzing SCEDs in human research contexts, researchers should implement these statistical approaches:

  • Visual analysis: Systematic examination of level, trend, variability, immediacy of effect, overlap, and consistency across similar phases

  • Effect size calculations: Measures like percentage of non-overlapping data, improvement rate difference, or Tau-U

  • Randomization tests: Permutation-based inference to strengthen causal claims

  • Hierarchical linear modeling: For analyzing multiple cases simultaneously

  • Interrupted time series analysis: To model trend changes at phase transitions

Key considerations include accounting for autocorrelation in repeated measurements, establishing minimum phase lengths, confirming data stability, and utilizing multiple replications to demonstrate experimental control.

How can researchers design studies to evaluate CBG's anti-inflammatory properties in humans?

Designing rigorous studies to evaluate CBG's anti-inflammatory properties requires:

  • Experimental models:

    • In vitro: Human microglial cell models (e.g., BV2 cells) with LPS stimulation

    • Ex vivo: Human peripheral blood mononuclear cells

    • In vivo: Controlled human exposure studies with inflammatory challenges

  • Measurement parameters:

    • Inflammatory biomarkers: Pro-inflammatory cytokines (TNF-α, IL-6, IL-1β)

    • Signaling pathway activation: NF-κB, MAP kinases

    • Functional outcomes: Clinical measures of inflammation

  • Experimental design considerations:

    • Dose-response relationships (multiple CBG concentrations)

    • Time-course measurements to capture both acute and sustained effects

    • Comparison with established anti-inflammatory agents as positive controls

    • Inclusion of both CBG and its identified metabolites (particularly cyclo-CBG)

  • Mechanistic investigations:

    • Receptor antagonist studies to determine pathway dependence

    • Gene expression analysis to characterize broader anti-inflammatory effects

How should researchers implement reversal designs in CGB human studies?

When implementing reversal designs (e.g., ABAB) in human studies:

  • Phase structure:

    • A phases: Baseline/control condition

    • B phases: Intervention condition

    • Minimum 5 data points per phase

    • Phase lengths determined by data stability rather than fixed durations

  • Practical implementation:

    • Establish stability criteria before phase transitions (data points within 15% of median)

    • Ensure no trending in direction of treatment effect during baseline

    • Allow sufficient time between phases for washout or stabilization

    • Randomize phase change points when possible

  • Replication requirements:

    • Three replications of treatment effects represent the minimum standard

    • Example of replications in an ABAB design: A₁ vs. B₁, B₁ vs. A₂, A₂ vs. B₂

    • Consider both within-subject and across-subject replications

This approach provides strong evidence for causal relationships between interventions and outcomes while controlling for potential confounding variables.

What approaches can strengthen multiple baseline designs in CGB cancer genetics research?

To strengthen multiple baseline designs in Clinical Genetics Branch cancer genetics research:

  • Staggered implementation:

    • Introduce interventions at different timepoints across participants/families

    • Base timing on stability of baseline data rather than predetermined schedules

    • Consider genetic risk profiles when determining implementation sequence

  • Design variants for specific contexts:

    • Multiple baseline across participants (different high-risk individuals)

    • Multiple baseline across behaviors (different preventive behaviors)

    • Multiple baseline across settings (clinic vs. home-based interventions)

  • Statistical enhancement approaches:

    • Randomization of intervention timing

    • Hierarchical linear modeling for data analysis

    • Calculation of effect sizes across cases

These methods strengthen internal validity while maintaining the practical advantages of multiple baseline designs in cancer genetics research.

How can single-case experimental designs be integrated into larger clinical trials?

Integrating SCEDs into larger clinical trials offers several methodological advantages:

  • Hybrid design approaches:

    • Initial SCED phase to identify responders, followed by RCT

    • RCT with embedded SCEDs for detailed individual response patterns

    • SCEDs to determine optimal personalized dosing before group comparisons

  • Implementation strategies:

    • Use SCEDs to examine individual differences that may be masked in group designs

    • Apply SCED methodology to intensively study a subset of participants

    • Employ SCEDs for pilot testing interventions before large-scale trials

  • Statistical integration:

    • Multilevel modeling to combine individual and group-level data

    • Bayesian approaches to integrate prior information from SCEDs into RCT analysis

    • Meta-analytic techniques to synthesize findings across single cases

This integration allows researchers to capture both personalized effects and population-level outcomes, particularly valuable in heterogeneous conditions.

What methodological challenges exist in measuring CBG metabolites in human samples?

Researchers face several methodological challenges when measuring CBG metabolites:

Challenge CategorySpecific IssuesPotential Solutions
Analytical sensitivityLow metabolite concentrationsHigh-sensitivity LC-MS/MS techniques
Metabolite stabilityDegradation during processingSpecialized stabilization protocols
Comprehensive identificationUnknown or minor metabolitesUntargeted metabolomics approaches
Reference standardsLimited availabilityChemical synthesis of authentic standards
Individual variabilityGenetic differences in metabolismLarger sample sizes, pharmacogenetic analysis

Additional challenges include tissue-specific metabolism that may not be reflected in accessible biofluids and timing considerations relative to CBG administration .

How can human-centric approaches improve oncology research in the Clinical Genetics Branch?

Human-centric approaches can enhance Clinical Genetics Branch oncology research through:

  • Personalized risk assessment:

    • Integration of genetic, environmental, and behavioral factors

    • Development of polygenic risk scores specific to high-risk populations

    • Implementation of AI/machine learning to identify novel risk patterns

  • Shared decision-making frameworks:

    • Structured approaches to communicating uncertainty about benefits and risks

    • Methods to incorporate patient values into screening and prevention decisions

  • Patient-reported outcome integration:

    • Systematic collection of symptoms, quality of life, and functional status

    • Real-time monitoring through digital health technologies

  • Translational research acceleration:

    • Biospecimen collection linked to comprehensive clinical data

    • Patient-derived models for personalized intervention testing

These approaches can improve the relevance, acceptability, and clinical impact of oncology research while addressing the unique needs of high-risk populations.

What emerging technologies are advancing CBG research in human systems?

Emerging technologies advancing CBG research include:

  • Advanced 'omics approaches:

    • Metabolomics for comprehensive CBG metabolite profiling

    • Transcriptomics to map genome-wide responses to CBG exposure

    • Proteomics to identify affected pathways and protein interactions

    • Single-cell technologies to capture cell-specific responses

  • Organoid and microphysiological systems:

    • Human tissue-derived organoids for CBG testing

    • Organ-on-chip models incorporating multiple cell types

    • Blood-brain barrier models to assess CNS penetration

    • Gut-liver axis models to study first-pass metabolism

  • In silico modeling advances:

    • Molecular dynamics simulations of CBG-receptor interactions

    • Physiologically-based pharmacokinetic modeling

    • AI-driven prediction of drug-target interactions

  • Precision biomarker development:

    • Targeted assays for CBG-specific response indicators

    • Digital biomarkers for objective assessment of physiological effects

    • Imaging techniques to visualize receptor engagement and pathway activation

These technologies enable more precise, comprehensive research on CBG's effects in human biological systems.

What ethical considerations are unique to CBG human research?

CBG human research presents several distinct ethical considerations:

  • Regulatory classification challenges:

    • Navigating the complex regulatory status of cannabinoids

    • Establishing appropriate inclusion/exclusion criteria

    • Determining appropriate risk categorization for IRB review

  • Scientific validity requirements:

    • Ensuring rigorous study design despite evolving regulatory landscape

    • Addressing potential placebo effects and expectancy bias

    • Implementing appropriate blinding procedures

  • Participant protection concerns:

    • Monitoring for drug-drug interactions with medications

    • Special considerations for vulnerable populations

    • Clear communication about the distinction between CBG and THC

Researchers must navigate these considerations while maintaining scientific integrity and regulatory compliance.

How should researchers design SCEDs to meet regulatory requirements for evidence generation?

To design SCEDs that meet regulatory requirements:

  • Protocol development:

    • Pre-specify phase change criteria and decision rules

    • Define primary and secondary outcome measures

    • Include power calculations based on SCED-specific methods

    • Document randomization procedures for phase transitions

  • Quality control measures:

    • Implement blinded outcome assessment whenever possible

    • Establish inter-observer agreement for behavioral measures

    • Document treatment fidelity throughout the study

    • Maintain consistent measurement procedures across phases

  • Reporting standards:

    • Follow established SCED reporting guidelines

    • Include individual participant data

    • Report effect sizes with confidence intervals

    • Provide detailed protocol adherence information

These approaches align single-case methodologies with regulatory expectations for scientific rigor and evidence quality.

Product Science Overview

Gene and Protein Structure

The CGB gene is located on chromosome 19q13.3 and is part of a cluster of six highly homologous genes arranged in tandem and inverted pairs . These genes are structurally similar and are contiguous with the luteinizing hormone beta (LHB) subunit gene . The CGB gene family is distinguished by differences in the 5’ untranscribed region, which affects their regulation and expression.

The CGB protein itself is composed of 145 amino acids and forms a heterodimer with the alpha subunit of hCG. This heterodimer is essential for the biological activity of hCG, which includes stimulating the production of progesterone by the corpus luteum during early pregnancy .

Recombinant Production

Recombinant human CGB is produced using various expression systems, including glycoengineered Pichia pastoris and mammalian cell lines . These systems are designed to produce glycoproteins with human-like post-translational modifications, ensuring that the recombinant protein closely mimics the natural form found in the human body .

The recombinant production of CGB involves inserting the CGB gene into the host organism’s genome, allowing it to produce the protein. The protein is then purified and characterized to ensure its quality and functionality. This process is crucial for producing large quantities of CGB for research and therapeutic purposes.

Applications

Recombinant human CGB has several important applications:

  1. Medical Diagnostics: CGB is widely used in pregnancy tests due to its presence in the urine and blood of pregnant women. Elevated levels of hCG can also be indicative of certain types of cancers, making it a valuable biomarker in oncology.
  2. Therapeutics: Recombinant hCG is used in fertility treatments to induce ovulation and support early pregnancy. It is also used in certain cancer treatments to stimulate the production of testosterone in men with hypogonadism.
  3. Research: Recombinant CGB is used in various research applications to study its role in reproduction, cancer, and other physiological processes.

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.