GADD45G Human

Growth Arrest and DNA-Damage-Inducible Gamma Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

Core regulatory roles:

  • Cell cycle control: Inhibits cyclin B1/Cdk1 complex without dissociating it, enforcing S/G2-M checkpoints

  • Stress response: Activates p38/JNK pathway via MTK1/MEKK4 kinase under DNA damage or hypertonic stress

  • Developmental processes:

    • Sexual differentiation: Activates SRY through p38 MAPK signaling

    • Neurodevelopment: Enhancer deletion near GADD45G increases neuronal proliferation in humans

Interaction Partners:

  • MAP3K4 (MTK1/MEKK4)

  • PCNA, p21, cdc2

  • NF-κB (upstream regulator)

Tissue Distribution

TissueExpression LevelSource
Skeletal muscleHighProtein Atlas
PlacentaHigh
LiverModerate
BrainLow
KidneyHigh

Clinical Significance in Human Diseases

Cancer Associations:

Cancer TypeGADD45G RoleClinical Impact
Hepatocellular carcinomaDownregulation promotes JAK-STAT3 activationPoor prognosis (HR=1.48, p<0.001)
Esophageal cancerPromoter methylation (62% of cases)Reduced 5-year survival
Breast cancerOverexpression inhibits metastasis73% reduction in xenograft growth
Myeloproliferative neoplasmsHaploinsufficiency drives HSC expansion4-fold increase in myeloid progenitors

Neurological Implications:

  • Alzheimer’s disease: Modulates DNA damage response in hippocampal neurons

  • Parkinson’s disease: Regulates SRY through p38-MAPK/GATA pathway

  • Depression: Hippocampal GADD45G downregulation correlates with cognitive impairment

Recent Research Breakthroughs

Therapeutic Targeting:

  • 4-Methoxydalbergione (4MOD) upregulates GADD45G in HCC, reducing tumor volume by 58% in murine models

  • Vitamin D induces GADD45G expression in prostate cancer, restoring growth arrest

Mechanistic Insights:

  • GADD45G-deficient mice show:

    • 300% increase in My-biased HSCs

    • Defective gastrulation and neural tube closure

  • Enhancer deletion (chr9: 21,773,789-21,775,389) increases cortical neurons by 40% in human forebrain organoids

Product Specs

Introduction
Belonging to the nuclear protein family, GADD45G interacts with various proteins whose transcript levels increase in response to stressful growth arrest conditions and DNA-damaging agents. GADD45G plays a role in cellular response to environmental stresses by mediating the activation of the p38/JNK pathway. This is achieved through protein binding and activation of MTK1/MEKK4 kinase, an upstream activator of both p38 and JNK MAPKs. While GADD45G acts as a novel tumor suppressor, it is frequently epigenetically inactivated in various tumors. Notably, GADD45G mRNA expression is down-regulated in hepatocellular carcinoma. When transfected into Hep-G2 cells, GADD45G induces cell cycle arrest at the G2/M transition. Androgen-induced GADD45 Gamma expression involves de novo protein synthesis. Overexpression of GADD45 Gamma in prostate cell lines leads to growth inhibition and morphological changes, indicating its role in androgen-induced differentiation.
Description
Recombinant human GADD45G, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 159 amino acids. It has a molecular weight of 17.1 kDa.
Physical Appearance
The product is a clear, colorless solution that has been sterilized by filtration.
Formulation
The GADD45G protein solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl at pH 7.
Stability
For short-term storage (up to 2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to store the product frozen at -20°C.
To ensure long-term stability, the addition of a carrier protein (either HSA or BSA) at a concentration of 0.1% is advised.
Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the GADD45G protein is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
DDIT2, GADD45gamma, GADD45G, Growth arrest and DNA-damage-inducible protein GADD45 gamma, Cytokine-responsive protein CR6, DNA-damage-inducible transcript 2, DDIT-2, CR6, GRP17.
Source
Escherichia Coli.
Amino Acid Sequence
MTLEEVRGQD TVPESTARMQ GAGKALHELL LSAQRQGCLT AGVYESAKVL NVDPDNVTFC VLAAGEEDEG DIALQIHFTL IQAFCCENDIDIVRVGDVQR LAAIVGAGEE AGAPGDLHCI LISNPNEDAW KDPALEKLSL FCEESRSVND WVPSITLPE.

Q&A

What is GADD45G and what are its fundamental roles in human cells?

GADD45G is a member of the Growth Arrest and DNA Damage (GADD45) family, known primarily for its involvement in DNA repair, cell cycle regulation, and neural development. It contains human-specific conserved deletion enhancer-sequences (hCONDEL) that have contributed to the evolution of the human cerebrum . While traditionally known as a tumor suppressor gene, GADD45G has specialized functions in neurons where it promotes neurite outgrowth by facilitating microtubule polymerization .

GADD45G functions through multiple mechanisms, including:

  • Regulation of cell cycle progression, particularly at the G2/M checkpoint

  • Modulation of DNA methylation and demethylation processes

  • Activation of p38 MAPK signaling pathways leading to downstream effects on cellular morphology and function

  • Temporal regulation of immediate early gene (IEG) expression during learning and memory formation

These functions make GADD45G a critical molecule in both development and adult neuroplasticity.

How does GADD45G expression vary across human development stages?

GADD45G expression follows a distinct developmental pattern in the human brain. Analysis of bulk RNA-seq data from the BrainSpan Atlas of the Developing Human Brain shows that GADD45G expression is significantly higher in the fetal brain and in the brains of humans up to 10 years of age compared to older individuals .

The temporal expression pattern shows:

  • Highest expression in early embryonic period

  • Gradually decreasing expression in late embryonic period

  • Maintained expression through childhood

  • Significant reduction in expression in adulthood

This temporal expression profile aligns with periods of active neurogenesis and neural circuit formation, suggesting GADD45G's importance in these developmental processes.

What experimental approaches are used to study GADD45G expression in human tissues?

Multiple complementary approaches are used to study GADD45G expression:

  • Quantitative Real-Time PCR: Used to measure GADD45G mRNA levels, as demonstrated in studies of human nucleus pulposus cells .

  • Western Blotting: Enables detection of GADD45G protein levels and can be used to analyze changes in expression under different conditions .

  • Immunohistochemistry: Allows visualization of GADD45G expression in tissue sections, providing spatial information about expression patterns .

  • RNA-seq Analysis: Provides comprehensive transcriptome data for examining GADD45G expression across different developmental stages and brain regions .

  • Chromatin Immunoprecipitation (ChIP): Used to study GADD45G occupancy on DNA, particularly at promoter regions of genes regulated by GADD45G .

When designing experiments, researchers should consider appropriate controls and validation methods for each technique to ensure specific detection of GADD45G.

How does the GADD45G/p38 MAPK/CDC25B signaling pathway regulate neurite outgrowth?

The GADD45G/p38 MAPK/CDC25B signaling pathway represents a sophisticated mechanism for promoting neurite outgrowth in human neurons. This pathway functions through several coordinated steps:

  • GADD45G activates p38 MAPK through phosphorylation, as verified through siRNA-mediated knockdown experiments showing that GADD45G knockdown significantly reduces phospho-p38 levels .

  • Activated phospho-p38 subsequently phosphorylates CDC25B, as demonstrated by experiments using SB203580 (a p38 MAPK inhibitor) which suppressed phospho-CDC25B levels .

  • This signaling cascade ultimately promotes the dephosphorylation of CRMP2 (Collapsin Response Mediator Protein 2) .

  • Dephosphorylated CRMP2 enhances microtubule assembly, which is essential for neurite extension and growth .

This pathway provides a mechanistic explanation for how GADD45G influences neuronal morphology, highlighting its role beyond traditional DNA damage response functions in the unique context of human neuronal development.

What is the relationship between GADD45G and DNA methylation in regulating gene expression?

GADD45G plays a critical role in active DNA demethylation processes that regulate gene expression, particularly in the context of learning and memory. Research has revealed:

  • GADD45G binds to the promoter regions of immediate early genes (IEGs) at specific time points after learning events (approximately 5 hours post-stimulus) .

  • This binding coincides with a sharp decline in 5-methylcytosine (5-mC) levels, suggesting GADD45G mediates active DNA demethylation .

  • Knockdown of GADD45G prevents this demethylation, resulting in persistently high levels of 5-mC at these promoters .

  • The demethylation process controlled by GADD45G appears to be essential for the second wave of IEG expression during memory consolidation .

This creates a two-tier model where initial gene activation may be triggered by DNA double-strand breaks, while subsequent expression is regulated by GADD45G-mediated DNA demethylation. This mechanism represents a sophisticated epigenetic control system that enables precise temporal regulation of gene expression during learning and memory formation.

How does GADD45G contribute to cell cycle regulation and DNA repair?

GADD45G functions as a cell cycle regulator primarily by mediating G2/M checkpoint control. Research shows:

  • Increased expression of GADD45 arrests cells at the G2/M boundary, characterized by MPM2 immunopositivity, 4n DNA content, and in some cells, centrosome separation .

  • This GADD45-mediated G2/M arrest depends on wild-type p53, as it was not observed in p53-null Li-Fraumeni fibroblasts or in normal fibroblasts co-expressed with p53 mutants .

  • Mechanistically, GADD45G appears to modulate the activity of G2-specific kinase cyclin B1/p34 cdc2, as increased expression of cyclin B1 and Cdc25C inhibited the GADD45-mediated G2/M arrest .

  • Cells with reduced GADD45 expression (via antisense technology) or knockout GADD45 show impaired G2/M checkpoint activation after exposure to genotoxic agents like ultraviolet radiation or methyl methanesulfonate .

This cell cycle regulatory function is important in the context of genomic integrity maintenance and may be particularly relevant in neural stem/progenitor cells which must carefully regulate proliferation during brain development.

What are the most effective techniques for manipulating GADD45G expression in human neural cells?

Several complementary approaches have proven effective for manipulating GADD45G expression in neural cells:

  • siRNA-mediated knockdown: This approach has been successfully employed to reduce GADD45G expression in human iPSC-derived neural stem/progenitor cells (hiPSC-NS/PCs). The effectiveness can be verified through western blot analysis of phospho-p38 levels, which are reduced following GADD45G knockdown .

  • shRNA expression: Short hairpin RNAs delivered via viral vectors allow for more stable long-term knockdown of GADD45G. This approach has been used to study the role of GADD45G in IEG expression during memory formation .

  • CRISPR-Cas9 gene editing: While not explicitly mentioned in the provided search results, this technique represents the current gold standard for generating complete gene knockouts or introducing specific mutations.

  • Overexpression systems: Introduction of GADD45G expression vectors has been used to study the effects of increased GADD45G levels, for example in cell cycle regulation studies where microinjection of GADD45 expression vectors arrested cells at the G2/M boundary .

When designing knockdown experiments, researchers should verify specificity by demonstrating that other GADD45 family members (GADD45α and GADD45β) are not affected, as has been shown in ChIP experiments where only GADD45G bound to the IEG promoters .

What cell and tissue models are most appropriate for studying GADD45G function in human neural development?

Several experimental models have proven valuable for investigating GADD45G function in neural development:

  • Human iPSC-derived neural stem/progenitor cells (hiPSC-NS/PCs): These neurospheres provide an excellent model for studying GADD45G's role in neural development. They can be treated with γ-secretase inhibitors (GSIs) to induce neuronal differentiation and GADD45G expression .

  • Human fetal brain samples: For studying in vivo expression patterns, human fetal brain tissues at different developmental stages allow examination of GADD45G's temporal expression. The BrainSpan Atlas of the Developing Human Brain provides valuable transcriptomic data for such analyses .

  • Primary human fibroblasts: While not neural cells, these have been used effectively to study GADD45G's role in cell cycle regulation and can provide insights into basic mechanisms that may apply to neural cells as well .

  • Knockout mouse models: While not human, gadd45-knockout mice (gadd45−/−) have provided important genetic evidence for GADD45's role in cell cycle checkpoints and can serve as valuable complementary models .

For neural development studies specifically, hiPSC-NS/PCs represent the most accessible and ethically appropriate human model, as they recapitulate many aspects of human neuronal development while avoiding the ethical concerns associated with human fetal tissue research.

What are the key considerations for chromatin immunoprecipitation (ChIP) experiments targeting GADD45G?

When conducting ChIP experiments to study GADD45G DNA binding:

  • Antibody specificity: Crucial to verify that the antibody specifically recognizes GADD45G and not other GADD45 family members. Control experiments should demonstrate that ChIP for GADD45α or GADD45β shows no binding at the same loci .

  • Timing considerations: GADD45G binding appears to be highly time-dependent, with peak binding observed at specific time points after stimulation (e.g., 5 hours post-fear conditioning in brain tissue) . Experimental design should include multiple time points to capture this dynamic binding.

  • Genomic region selection: Include both target promoter regions and distal regions as controls. For example, ChIP experiments showed GADD45G binding at IEG promoters but not at distal promoter regions of genes like Cyr61 .

  • Validation of binding significance: Complement ChIP with functional assays such as gene expression analysis and DNA methylation assessment to establish the functional relevance of GADD45G binding .

  • Appropriate controls: Include isotype control antibodies and input samples as technical controls. Additionally, perform ChIP in tissues/cells with GADD45G knockdown to confirm binding specificity .

Following these considerations will help ensure robust and interpretable results from ChIP experiments targeting GADD45G.

How do researchers distinguish between the functions of different GADD45 family members in humans?

Distinguishing between GADD45 family members (GADD45α, GADD45β, and GADD45γ) requires multiple complementary approaches:

  • Specific gene/protein targeting: Using highly specific antibodies or nucleic acid probes that recognize only one family member. Validation through knockout or knockdown models is essential to confirm specificity .

  • Differential expression analysis: Examining the spatial and temporal expression patterns of each family member can reveal distinct roles. For example, GADD45γ is specifically expressed in neural stem/progenitor cells and immature neurons in developing human fetal brains .

  • Functional complementation studies: Determining whether one family member can rescue the phenotypes caused by deficiency of another can reveal functional redundancy or specificity.

  • Region and task specificity: Different family members may function in distinct brain regions or learning tasks. For example, while global Gadd45β knockout affected contextual but not cued fear conditioning, Gadd45γ is required in the prelimbic prefrontal cortex for cued fear conditioning .

  • Target gene analysis: ChIP experiments can reveal different DNA binding profiles for each family member. For instance, ChIP for Gadd45γ showed binding to IEG promoters, while ChIP for Gadd45α or Gadd45β revealed no binding at the same loci .

These approaches collectively help delineate the unique functions of each GADD45 family member, revealing that they likely have both overlapping and distinct roles in different cellular contexts.

How can contradictory findings on GADD45G function in different tissues be reconciled?

Reconciling apparently contradictory findings on GADD45G function requires considering several factors:

  • Tissue-specific roles: GADD45G may have distinct functions in different tissues. For example, it functions as a tumor suppressor in many cell types but plays specialized roles in neurite outgrowth in neurons .

  • Developmental timing: GADD45G function may vary across developmental stages. Its expression is higher in early embryonic periods than in late embryonic periods, suggesting stage-specific roles .

  • Interaction with different signaling networks: The molecular context matters. In neurons, GADD45G interacts with the p38 MAPK/CDC25B pathway , while in memory formation it interacts with DNA methylation machinery .

  • Experimental approach differences: Variations in experimental techniques (in vitro vs. in vivo, knockdown vs. knockout, etc.) can lead to seemingly contradictory results.

  • Spatial specificity: Even within the brain, GADD45G may have region-specific functions. Different members of the GADD45 family appear to be region-specific and thus also task-specific in learning paradigms .

A systems biology approach that integrates data across multiple experimental paradigms and tissues is necessary to develop a comprehensive understanding of GADD45G's multifaceted functions. This includes computational modeling to predict how GADD45G might function differently depending on the molecular context.

What is the evidence for GADD45G involvement in human neurological disorders?

While the provided search results don't directly address GADD45G's role in neurological disorders, the gene's functions suggest potential involvement:

  • Neurodevelopmental disorders: Given GADD45G's role in human cerebral evolution and its high expression during fetal and early childhood brain development , dysregulation could potentially contribute to neurodevelopmental disorders.

  • Learning and memory disorders: GADD45G's critical role in regulating IEG expression during memory consolidation suggests it might be involved in disorders characterized by learning and memory deficits.

  • Cell cycle regulation: GADD45G's role in the G2/M checkpoint might be relevant to conditions involving neural progenitor proliferation abnormalities.

  • DNA repair processes: As GADD45G interacts with DNA double-strand breaks and DNA methylation , it could potentially be implicated in disorders arising from defective DNA repair mechanisms in the brain.

Future research directions should include:

  • Genetic association studies examining GADD45G variants in patients with neurological disorders

  • Expression analyses in postmortem brain tissue from affected individuals

  • Functional studies using patient-derived iPSCs differentiated into neural cells

  • Animal models with brain region-specific manipulation of GADD45G expression

These approaches would help establish more direct links between GADD45G and specific neurological conditions.

What are promising new approaches for studying GADD45G's role in human brain development?

Several emerging approaches hold promise for advancing our understanding of GADD45G in human brain development:

  • Human brain organoids: These 3D cultures recapitulate aspects of human brain development and could serve as powerful models for studying GADD45G function in a more physiologically relevant context than 2D cultures.

  • Single-cell transcriptomics: This technique allows assessment of GADD45G expression at the single-cell level, enabling researchers to identify cell type-specific expression patterns and functions during development.

  • Spatiotemporal epigenomic mapping: Combining ChIP-seq with DNA methylation analysis across development stages could reveal how GADD45G dynamically regulates gene expression through epigenetic mechanisms.

  • CRISPR-based epigenome editing: This approach enables targeted modification of epigenetic marks at specific genomic loci, allowing researchers to directly test how GADD45G-mediated changes in DNA methylation affect gene expression.

  • Live imaging of GADD45G activity: Development of fluorescent reporters or biosensors for GADD45G activity could allow real-time visualization of its function during neuronal differentiation and neurite outgrowth.

These advanced techniques would provide deeper insights into the molecular mechanisms through which GADD45G influences human brain development and potentially identify new therapeutic targets for neurodevelopmental disorders.

How might understanding GADD45G signaling contribute to regenerative medicine approaches for neurological conditions?

GADD45G's roles in neurite outgrowth and neural development suggest several potential applications in regenerative medicine:

  • Neural regeneration promotion: Since GADD45G enhances neurite outgrowth by facilitating microtubule polymerization through the GADD45G/p38 MAPK/CDC25B pathway , modulating this pathway could potentially enhance neural regeneration after injury.

  • Stem cell differentiation control: GADD45G's expression in neural stem/progenitor cells suggests it may regulate their differentiation . Manipulating GADD45G levels could help direct stem cells toward specific neural lineages for transplantation therapies.

  • Epigenetic reprogramming: GADD45G's role in active DNA demethylation could be harnessed to epigenetically reprogram cells for regenerative purposes, potentially enabling conversion of non-neuronal cells into neurons.

  • Cell cycle regulation for neural progenitors: GADD45G's function in G2/M checkpoint control might be utilized to expand neural progenitor populations while maintaining genomic integrity.

  • Memory enhancement or restoration: Given GADD45G's role in memory consolidation , targeted manipulation of this pathway could potentially enhance learning or restore memory function in conditions like Alzheimer's disease.

Translational research should focus on developing small molecule modulators of the GADD45G pathway or gene therapy approaches that could selectively activate or inhibit GADD45G function in specific neural cell populations.

What statistical approaches are most appropriate for analyzing temporal dynamics of GADD45G activity?

Analyzing the temporal dynamics of GADD45G activity requires sophisticated statistical approaches:

  • Time series analysis: Methods such as functional data analysis or growth curve modeling are well-suited for capturing the temporal patterns of GADD45G expression, which shows distinct waves of activity during processes like memory formation .

  • Change-point detection algorithms: These can identify critical time points when GADD45G binding or expression significantly changes, helping to define key transition points in biological processes.

  • Mixed-effects models: These account for both fixed effects (e.g., experimental conditions) and random effects (e.g., biological variability between samples), making them appropriate for analyzing repeated measures of GADD45G activity across time.

  • Clustering approaches for temporal patterns: Methods like dynamic time warping can identify similar temporal patterns of GADD45G activity across different genes or experimental conditions.

  • Causal inference methods: Granger causality or structural equation modeling can help establish causal relationships between GADD45G activity and downstream molecular events across time.

  • Network analysis of temporal data: Time-varying network models can capture how GADD45G's interactions with other molecules change over time, providing insights into the dynamic nature of its regulatory functions.

When designing experiments to study GADD45G temporal dynamics, researchers should include multiple time points with sufficient resolution to capture the biphasic patterns observed in processes like memory consolidation, where GADD45G shows specific binding at the 5-hour time point .

Product Science Overview

Gene and Protein Structure

The GADD45G gene is located on chromosome 9 in humans . It is also known by several aliases, including CR6, DDIT2, GRP17, and OIG37 . The protein encoded by this gene is involved in several processes, including sexual development, human-specific brain development, tumor suppression, and the cellular stress response .

Function and Mechanism

GADD45G plays a crucial role in the regulation of growth and apoptosis. It mediates the activation of stress-responsive MTK1/MEKK4 MAPKKK pathways . This protein responds to environmental stresses by activating the p38/JNK pathway via MTK1/MEKK4 kinase . The activation of these pathways leads to various cellular responses, including DNA repair, cell cycle arrest, and apoptosis.

Expression and Regulation

The expression of GADD45G is highly induced under stressful growth arrest conditions and treatment with DNA-damaging agents . It is highly expressed in the placenta and other tissues . The protein interacts with several other proteins involved in DNA repair, cell cycle control, apoptosis, and senescence .

Clinical Significance

Low expression of GADD45G has been associated with various types of cancer . Its role as a tumor suppressor was discovered in 1999 . The protein’s ability to mediate stress responses and regulate cell cycle arrest makes it a critical component in maintaining cellular integrity and preventing tumorigenesis.

Historical Background

GADD45G was originally cloned by Beadling under the name CR6 in 1993 . It was identified as an immediate early response gene in T lymphocytes induced by IL-2 . Its role as a tumor suppressor and its regulation by Oncostatin M were later discovered, leading to its current understanding as a critical stress response protein .

Research and Applications

Research on GADD45G continues to explore its role in various cellular processes and its potential therapeutic applications. The recombinant form of this protein is used in various research studies to understand its function and mechanism in greater detail.

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.