GNGT1 Human

Guanine Nucleotide Binding Protein Gamma Transducing 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

GNGT1 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 94 amino acids (1-71 a.a) and having a molecular mass of 10.6kDa.
GNGT1 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Guanine Nucleotide Binding Protein Gamma Transducing 1, also known as GNGT1, is a member of the G protein gamma family. GNGT1 is involved in various transmembrane signaling systems, acting as a modulator or transducer. Additionally, GNGT1 (beta & gamma chains) is essential for GTPase activity, the exchange of GDP for GTP, and interactions between G proteins and their effectors.
Description
Recombinant human GNGT1, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 94 amino acids (1-71 a.a). It has a molecular weight of 10.6 kDa. The protein is fused to a 23-amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
GNGT1 protein solution (1 mg/mL) in 20 mM Tris-HCl buffer (pH 8.0), 10% glycerol, 0.1 M NaCl, and 1 mM EDTA.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 95.0% purity as determined by SDS-PAGE.
Synonyms
Guanine Nucleotide Binding Protein (G Protein), Gamma Transducing Activity Polypeptide 1, Transducin Gamma Chain, Guanine Nucleotide-Binding Protein G(T) Subunit Gamma-T1, GNG1, Guanine nucleotide-binding protein G(T) subunit gamma-T1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMPVINIE DLTEKDKLKM EVDQLKKEVT LERMLVSKCC EEVRDYVEER SGEDPLVKGI PEDKNPFKEL KGGC.

Q&A

What is GNGT1 and what is its functional role in human cells?

GNGT1, also known as Transducin gamma chain, belongs to the G protein gamma family. G proteins function as modulators or transducers in various transmembrane signaling systems, playing critical roles in cellular communication pathways. The beta and gamma chains of G proteins (including GNGT1) are required for three essential functions: GTPase activity, replacement of GDP by GTP, and facilitating G protein-effector interactions . This protein serves as a key component in signal transduction cascades that regulate numerous cellular processes, forming heterotrimeric complexes with other G protein subunits to mediate downstream signaling events.

How is GNGT1 typically expressed in normal human tissues?

GNGT1 expression varies across different tissue types in humans. Based on experimental evidence from transgenic mouse models, GNGT1 expression shows tissue specificity with variable levels observed between the lung, liver, kidney, and colon . When examining GNGT1 expression at the protein level through immunohistochemistry, researchers can detect differential expression patterns between normal and diseased tissues. The protein appears to have tissue-specific functions that correspond to its variable expression patterns, which researchers should consider when designing tissue-specific studies.

What mechanisms underlie GNGT1's role in cancer pathogenesis, particularly in lung adenocarcinoma?

GNGT1 has been identified as a potential oncogenic factor in lung adenocarcinoma (LUAD), with significant implications for tumor development and progression. Research has demonstrated that GNGT1 is overexpressed in LUAD tissues compared to adjacent normal tissues, as validated across multiple datasets including TCGA and GEO (GSE30219, GSE10072) . The oncogenic properties of GNGT1 appear to function through several mechanisms:

  • Promotion of cell proliferation: GNGT1 overexpression correlates with increased expression of proliferation markers such as Ki-67 and PCNA in lung tissues .

  • Enhancement of cancer stemness: GNGT1 expression positively correlates with stemness gene expression in LUAD, suggesting it may contribute to tumor initiation and progression through cancer stem cell mechanisms .

  • Interaction with driver genes: GNGT1 shows significant correlation with common driver genes in LUAD, indicating it may be part of important oncogenic signaling networks .

  • Microenvironment remodeling: GNGT1 appears to influence the tumor microenvironment through the activation of neutrophil extracellular trap (NET) formation via the FGB-NET axis, which may contribute to tumor promotion .

These mechanisms collectively suggest that GNGT1 overexpression is not merely a biomarker but potentially an early driving event in LUAD pathogenesis.

How does GNGT1 expression correlate with clinical outcomes in cancer patients?

GNGT1 expression levels have demonstrated significant associations with clinical outcomes in cancer patients, particularly in lung adenocarcinoma. Analysis of patient data reveals:

These clinical correlations suggest that GNGT1 may serve as both a prognostic biomarker and a potential therapeutic target, particularly for patients with specific molecular profiles.

What is the relationship between GNGT1 and immune cell infiltration in the tumor microenvironment?

GNGT1 has been found to significantly influence immune cell infiltration in the tumor microenvironment, potentially contributing to cancer progression. Research utilizing Tumor Immune Estimation Resource (TIMER) analysis has revealed correlations between GNGT1 expression and various immune cell populations:

  • GNGT1 expression correlates with the presence of specific immune cell types, including dendritic cells, macrophages, neutrophils, and T cell subpopulations in the tumor microenvironment .

  • GNGT1 appears to modulate NET (neutrophil extracellular trap) formation, which has been implicated in creating a favorable environment for tumor growth .

  • Expression of NET-related genes (PADI3, PADI4, and ELANE) as well as inflammatory markers (IL-6, CXCL4, and CXCL15) is altered in GNGT1-overexpressing models .

  • GNGT1 may influence molecules associated with tumor matrix remodeling (HMGB-1 and MMP9), further supporting its role in reshaping the tumor microenvironment .

These findings suggest that GNGT1 may be an important modulator of tumor-immune interactions, potentially serving as a target for immunotherapeutic approaches.

What are the recommended experimental models for studying GNGT1 function in vivo?

For studying GNGT1 function in vivo, researchers have successfully employed several experimental models, with the lung-specific GNGT1 transgenic mouse model representing a particularly valuable system:

  • GNGT1fl/+ transgenic mice: This model involves conditional overexpression of GNGT1 specifically in lung tissue. The construction typically follows a pattern as described in the literature, involving targeted gene insertion and verification .

  • Validation approaches for GNGT1 overexpression models should include:

    • Analysis of GNGT1 mRNA expression across multiple organs (lung, liver, kidney, colon) to confirm tissue-specific expression

    • Histological examination of different organs to assess morphologic structure

    • Immunohistochemical analysis to confirm GNGT1 protein expression

    • Evaluation of cell proliferation markers (Ki-67, PCNA) and apoptosis markers (caspase-3)

  • Phenotypic characterization should include assessment of:

    • Tumor formation and progression

    • Stemness gene expression (using RT-qPCR and immunofluorescence)

    • Expression of driver genes and other key molecular markers

This methodology enables researchers to directly assess the oncogenic properties of GNGT1 in a physiologically relevant context.

What techniques are most effective for analyzing GNGT1 expression in clinical samples?

For accurate and comprehensive analysis of GNGT1 expression in clinical samples, researchers should employ multiple complementary techniques:

  • RNA-level analysis:

    • Real-time quantitative PCR (RT-qPCR) for GNGT1 mRNA expression in frozen tissue samples

    • Transcriptomic analysis using microarray or RNA-sequencing, with validation across multiple datasets (e.g., GEO datasets GSE7670, GSE81089, GSE118370, TCGA)

  • Protein-level analysis:

    • Immunohistochemistry (IHC) for GNGT1 protein expression in formalin-fixed tissue samples

    • Western blotting for quantitative protein analysis

    • Tissue microarray analysis for high-throughput screening

  • Data analysis approaches:

    • Differential expression analysis using cutoff criteria of log fold change >1 and P<0.05

    • Correlation analysis between GNGT1 expression and clinical parameters

    • Survival analysis to assess prognostic significance

    • Fisher exact test to evaluate relationships between GNGT1 expression and clinicopathological characteristics

This multi-modal approach provides robust verification of GNGT1 expression patterns and their clinical significance.

How can researchers effectively identify and validate GNGT1-associated pathways and networks?

Identifying and validating GNGT1-associated pathways requires a comprehensive bioinformatic and experimental approach:

  • Bioinformatic analysis:

    • Gene Ontology (GO) analysis to describe gene products in terms of molecular function, biological process, and cellular component

    • Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis to identify enriched signaling pathways

    • Network analysis using tools like Cytoscape with the cytohubba app to calculate GNGT1-related hub genes

    • Correlation analysis between GNGT1 and potential key genes using heatmaps and scatter plots

  • Experimental validation:

    • RT-qPCR validation of identified hub genes and pathway components in appropriate model systems

    • Protein-protein interaction studies using co-immunoprecipitation or proximity ligation assays

    • Functional assays to assess the impact of GNGT1 modulation on identified pathways

  • Integration of multi-omics data:

    • Combined analysis of transcriptomic, proteomic, and epigenomic data

    • Correlation of GNGT1 expression with mutations in common driver genes (e.g., TP53)

    • Assessment of stemness gene expression in relation to GNGT1 levels

This systematic approach enables researchers to comprehensively map the molecular networks through which GNGT1 exerts its biological effects.

How can GNGT1 be utilized as a biomarker for early diagnosis of lung adenocarcinoma?

GNGT1 has demonstrated significant potential as a biomarker for early diagnosis of lung adenocarcinoma (LUAD), with several key characteristics supporting its clinical utility:

  • Consistent overexpression: GNGT1 shows significantly increased expression in LUAD tissues compared to adjacent normal tissues, as validated across multiple independent datasets (TCGA, GSE30219, GSE10072) and through experimental verification in patient samples using both mRNA and protein analysis .

  • Early occurrence: Evidence suggests that GNGT1 overexpression may constitute an early driving event in LUAD pathogenesis, making it potentially valuable for detecting disease at pre-symptomatic stages .

  • Implementation approaches:

    • Analysis of GNGT1 expression in liquid biopsies (circulating tumor DNA or RNA)

    • Development of GNGT1-based imaging probes for non-invasive detection

    • Integration of GNGT1 with other biomarkers to create diagnostic panels with improved sensitivity and specificity

  • Validation metrics:

    • Researchers should assess diagnostic performance using standard metrics including sensitivity, specificity, positive predictive value, and negative predictive value

    • ROC curve analysis to determine optimal cutoff values for GNGT1 expression levels

    • Longitudinal studies to evaluate the temporal relationship between GNGT1 overexpression and clinical disease manifestation

These approaches could facilitate the development of GNGT1-based diagnostic tools for early LUAD detection, potentially improving patient outcomes through earlier intervention.

What are the specific molecular mechanisms by which GNGT1 promotes lung tumor progression?

GNGT1 promotes lung tumor progression through several interconnected molecular mechanisms:

  • Enhancement of cellular proliferation:

    • GNGT1 overexpression correlates with increased expression of proliferation markers Ki-67 and PCNA

    • Reduced apoptosis as evidenced by decreased caspase-3 expression in GNGT1-overexpressing tissues

  • Promotion of cancer stemness:

    • GNGT1 expression shows positive correlation with stemness genes in LUAD samples from TCGA

    • Experimental validation in transgenic mouse models confirms upregulation of stemness-related genes in GNGT1-overexpressing lung tissues

  • Interaction with oncogenic drivers:

    • GNGT1 expression correlates with common driver genes in LUAD, suggesting interaction with established oncogenic pathways

    • Network analysis identifies key hub genes associated with GNGT1, including FGB, ALB, CTAG2, AFP, HP, and FGF4

  • Microenvironment remodeling via the FGB-NET axis:

    • GNGT1 promotes neutrophil extracellular trap (NET) formation through upregulation of FGB

    • Increased expression of NET-related genes (PADI3, PADI4, ELANE) and inflammatory markers (IL-6, CXCL4, CXCL15)

    • Elevated levels of matrix remodeling factors (HMGB-1, MMP9) supporting tumor invasion and metastasis

These mechanisms collectively contribute to a pro-tumorigenic environment conducive to LUAD initiation, progression, and potentially metastasis.

How does GNGT1 interact with other G protein subunits in the context of cancer signaling?

While the provided search results don't directly address the specific interactions between GNGT1 and other G protein subunits in cancer, we can infer some important considerations based on general G protein biology and the available data:

  • Functional relevance of G protein complexes:

    • GNGT1 belongs to the G protein gamma family and typically forms heterotrimeric complexes with beta and alpha subunits

    • The beta and gamma chains are required for GTPase activity, GDP/GTP exchange, and effector interactions

  • Potential cancer-specific interactions:

    • In the context of LUAD, GNGT1 likely interacts with specific beta subunits to form functional dimers that activate downstream signaling

    • These dimers may interact with unique effectors in cancer cells compared to normal cells, potentially explaining the oncogenic effects of GNGT1 overexpression

  • Research approaches to investigate these interactions:

    • Co-immunoprecipitation studies to identify cancer-specific binding partners of GNGT1

    • Proximity ligation assays to visualize and quantify protein-protein interactions in situ

    • FRET/BRET analysis to study dynamic interactions between G protein subunits in living cells

    • Structural biology approaches to determine the molecular details of cancer-specific G protein complexes

  • Therapeutic implications:

    • Understanding the specific interactions between GNGT1 and other G protein subunits could reveal novel therapeutic targets

    • Disrupting cancer-specific protein-protein interactions might provide a selective approach to targeting GNGT1-driven tumors

Further research specifically addressing these interactions in cancer contexts is needed to fully elucidate the role of GNGT1 in G protein signaling networks during tumorigenesis.

What therapeutic strategies targeting GNGT1 show the most promise for cancer treatment?

Based on current understanding of GNGT1's role in cancer, several promising therapeutic strategies emerge:

  • Small molecule inhibitors:

    • Development of compounds that selectively disrupt GNGT1's interaction with other G protein subunits

    • Design of molecules targeting the specific downstream effectors activated by GNGT1 in cancer contexts

  • Gene therapy approaches:

    • RNA interference (siRNA/shRNA) targeting GNGT1 mRNA

    • CRISPR/Cas9-mediated gene editing to correct aberrant GNGT1 expression

    • Antisense oligonucleotides to inhibit GNGT1 translation

  • Immunotherapeutic strategies:

    • Given GNGT1's role in immune cell infiltration and microenvironment remodeling , combination approaches with existing immunotherapies could be explored

    • Development of therapies targeting the FGB-NET axis identified as part of GNGT1's mechanism of action

  • Combination therapies:

    • Integration of GNGT1-targeted agents with conventional chemotherapy or radiation

    • Co-targeting of GNGT1 along with correlated driver genes identified through network analysis

  • Personalized medicine approaches:

    • Stratification of patients based on GNGT1 expression levels and mutation profiles

    • Development of companion diagnostics to identify patients most likely to benefit from GNGT1-targeted therapies

These therapeutic strategies represent promising avenues for translational research, though they require further development and validation in preclinical and clinical settings.

What are the current challenges in studying GNGT1 function and how might they be addressed?

Researchers studying GNGT1 face several significant challenges that require innovative solutions:

  • Tissue specificity and contextual function:

    • GNGT1 exhibits different roles across tissue types and disease states

    • Solution: Development of tissue-specific conditional models (as exemplified by the lung-specific GNGT1 transgenic mouse ) for each tissue of interest

  • Complex signaling networks:

    • GNGT1 operates within intricate G protein signaling networks with numerous interacting partners

    • Solution: Systems biology approaches integrating multi-omics data and network modeling to comprehensively map interactions

  • Translational barriers:

    • Bridging the gap between basic research findings and clinical applications

    • Solution: Early engagement with clinical researchers and industry partners to guide research toward clinically relevant questions

  • Technical limitations:

    • Challenges in studying protein-protein interactions in native contexts

    • Solution: Advanced imaging techniques (super-resolution microscopy, live-cell imaging) and proximity-based proteomics methods

  • Reproducibility concerns:

    • Ensuring consistent results across different model systems and laboratories

    • Solution: Development of standardized protocols, reagents, and reporting guidelines specific to G protein research

Addressing these challenges requires multidisciplinary collaboration and continued technological innovation in both experimental and computational approaches.

How might GNGT1 research extend beyond lung cancer to other disease models?

While current research highlights GNGT1's significance in lung adenocarcinoma, its potential relevance extends to other disease contexts:

  • Other cancer types:

    • The pancancer expression analysis indicates that GNGT1 is overexpressed in various tumors beyond LUAD

    • Research should explore whether similar mechanisms of action apply across cancer types or if tissue-specific effects exist

  • Non-malignant diseases:

    • Given G proteins' roles in signal transduction across various physiological processes, GNGT1 may have implications in:

      • Cardiovascular disorders

      • Neurodegenerative diseases

      • Inflammatory conditions

      • Metabolic disorders

  • Research approaches for disease expansion:

    • Cross-disease transcriptomic analysis to identify conditions with altered GNGT1 expression

    • Development of conditional GNGT1 models in other tissues beyond the lung

    • Functional studies examining GNGT1's role in different cell types and physiological contexts

  • Translational considerations:

    • Development of disease-specific biomarkers based on GNGT1 expression or modification

    • Exploration of common and distinct therapeutic approaches across different GNGT1-associated diseases

This expansion of GNGT1 research could reveal unexpected disease associations and broaden the impact of targeting this signaling protein for therapeutic purposes.

Product Science Overview

Introduction

Guanine Nucleotide Binding Protein Gamma Transducing 1 (GNGT1) is a crucial component of the heterotrimeric G protein complex, which plays a significant role in various cellular signaling pathways. This protein is particularly important in the visual signal transduction pathway in rod photoreceptor cells.

Gene and Protein Structure

The GNGT1 gene encodes the gamma subunit of transducin, a guanine nucleotide-binding protein (G protein) found in rod outer segments . The transducin protein is composed of three subunits: alpha, beta, and gamma. The gamma subunit, encoded by GNGT1, is essential for the proper functioning of the transducin complex .

Function

Transducin, also known as GMPase, mediates the activation of a cyclic GTP-specific (guanosine monophosphate) phosphodiesterase by rhodopsin . This process is crucial for the conversion of light signals into electrical signals in the retina, enabling vision. The gamma subunit of transducin is involved in the GTPase activity, replacement of GDP by GTP, and interaction with G protein effectors .

Pathways and Interactions

GNGT1 is involved in several signaling pathways, including:

  • ADORA2B mediated anti-inflammatory cytokines production
  • Thromboxane signaling through TP receptor

These pathways highlight the broader role of GNGT1 beyond visual signal transduction, indicating its involvement in various physiological processes.

Clinical Significance

Mutations or dysregulation of the GNGT1 gene have been associated with several diseases, including:

  • Prostate Cancer
  • Autosomal Recessive Limb-Girdle Muscular Dystrophy Type 2B

Understanding the role of GNGT1 in these diseases can provide insights into potential therapeutic targets and treatment strategies.

Recombinant GNGT1

Human recombinant GNGT1 is produced using recombinant DNA technology, which involves inserting the GNGT1 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in various research applications to study its function, interactions, and role in disease.

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.