GADD45A Human

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

Description

Gene and Protein Structure

  • Gene location: Chromosome 1 (1p31.2) .

  • Protein family: Part of the GADD45 family, which includes GADD45B and GADD45γ. These are small (~18 kDa), nuclear/cytoplasmic proteins with no enzymatic activity but extensive protein interaction capabilities .

  • Expression: Ubiquitously low under normal conditions but strongly induced by stressors (e.g., UV radiation, oxidative stress, chemotherapy) .

Key Interactions

Interaction PartnerFunctional RoleReference
MAP3K4/MEKK4Activates p38/JNK stress-response pathways
PCNAFacilitates DNA repair
CDK1/Cyclin B1Regulates G2/M cell cycle arrest
AURKAModulates mitotic progression

DNA Damage Response

GADD45A promotes nucleotide-excision repair (NER) and base excision repair (BER) by recruiting repair proteins like PCNA . It also induces cell cycle arrest at G2/M via CDK1 inhibition, allowing time for DNA repair .

Apoptosis and Senescence

  • Pro-apoptotic role: Mediates stress-induced apoptosis through p53-dependent and -independent pathways .

  • Senescence regulation: Sustained GADD45A expression triggers senescence, acting as a tumor suppressor .

Immune Modulation

GADD45A deficiency in dendritic cells reduces IL-12 and CD40 expression, impairing Th1 responses. Knockout mice develop autoimmune disorders (e.g., lupus-like symptoms) .

Cancer

  • Tumor suppression: Downregulated in malignancies (e.g., melanoma, ovarian cancer), enabling evasion of apoptosis and senescence .

  • Chemoresistance: In melanoma, cisplatin upregulates GADD45A, causing G2/M arrest and reducing apoptosis. Silencing GADD45A enhances cisplatin-induced DNA damage (γ-H2AX foci) and apoptosis by 2.5-fold .

Muscle Atrophy

  • Denervation response: GADD45A is rapidly induced post-denervation, delaying muscle atrophy in mice. Knockout models exhibit accelerated atrophy .

  • Mitochondrial loss: Transgenic GADD45A expression reduces oxidative capacity and glycolytic fiber mass, contributing to weakness .

Autoimmunity

GADD45A-deficient mice show spontaneous autoimmunity, including anti-dsDNA antibodies, highlighting its role in immune tolerance .

Diagnostic Applications

  • Mutagenicity testing: The GADD45a-GFP assay detects DNA damage via fluorescence in engineered cell lines .

  • Biomarker potential: Elevated GADD45A in skeletal muscle correlates with human muscle weakness .

Therapeutic Targets

ConditionInterventionOutcome
MelanomaGADD45A siRNA + cisplatinEnhanced apoptosis
Prostate cancerFucoxanthin (GADD45A inducer)G1 arrest via SAPK/JNK
Muscle atrophyGADD45A inhibitionPotential mitigation of weakness

Product Specs

Introduction
Growth arrest and DNA Damage-Inducible Protein (GADD45A) is a protein involved in DNA replication and repair. It interacts with Cdks and PCNA to perform its functions. This protein stimulates DNA excision repair in vitro and prevents cells from entering the S phase of the cell cycle. GADD45A potentially acts as a link between the p53-dependent cell cycle checkpoint and DNA repair mechanisms. The gene encoding GADD45A belongs to a group of genes whose transcription increases in response to stressful growth arrest conditions and exposure to DNA-damaging agents. GADD45A responds to environmental stresses by mediating the activation of the p38/JNK pathway through MTK1/MEKK4 kinase. Additionally, GADD45A binds to proliferating cell nuclear antigen.
Description
GADD45A, a human protein, has been produced in E. coli for research purposes. This recombinant protein is a single, non-glycosylated polypeptide chain consisting of 173 amino acids (specifically, amino acids 1 to 165 of the native protein sequence). It has a molecular mass of 19.4kDa. For purification and detection purposes, the recombinant GADD45A protein has been engineered with an 8 amino acid His Tag fused to its C-terminus. The protein has been purified using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution.
Formulation
The GADD45A protein is supplied in a solution containing 20mM Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.1M NaCl.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability, adding a carrier protein (0.1% HSA or BSA) is recommended. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the GADD45A protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
Growth arrest and DNA damage-inducible protein GADD45 alpha, DNA damage-inducible transcript 1 protein, DDIT-1, GADD45A, DDIT1, GADD45.
Source
Escherichia Coli.
Amino Acid Sequence
MTLEEFSAGE QKTERMDKVG DALEEVLSKA LSQRTITVGV YEAAKLLNVD PDNVVLCLLA ADEDDDRDVA LQIHFTLIQA FCCENDINIL RVSNPGRLAE LLLLETDAGP AASEGAEQPP DLHCVLVTNP HSSQWKDPAL SQLICFCRES RYMDQWVPVI NLPERLEHHH HHH.

Q&A

What is GADD45A and what are its primary cellular functions?

GADD45A functions as a crucial stress sensor responding to various cellular stressors, particularly DNA damage and oxidative stress. The protein plays an essential role in multiple cellular processes including cell cycle regulation, DNA repair, and apoptotic pathways.

When investigating GADD45A function, researchers should consider its dual role in:

  • DNA damage response mechanisms

  • Regulation of reactive oxygen species (ROS) levels

  • Promotion of cell differentiation

  • Protection of genomic integrity

Methodologically, GADD45A function can be studied using various approaches including promoter-reporter constructs (such as GADD45a-GFP), gene knockout models, and protein-protein interaction analyses. The GADD45a-GFP system, for example, utilizes Green Fluorescent Protein expression under the control of the GADD45a promoter to detect genotoxic potential in human cells .

How does GADD45A contribute to genomic stability?

GADD45A maintains genomic stability through several mechanistic pathways:

  • DNA repair facilitation: GADD45A interacts with components of both nucleotide excision repair and base excision repair pathways

  • Cell cycle checkpoint activation: Induces G2/M arrest following DNA damage

  • Epigenetic regulation: Influences DNA methylation patterns

  • Prevention of oncogenic transformation: Removes damaged hematopoietic stem cells through differentiation

Research demonstrates that Gadd45a-deleted mice display genomic instability, impaired DNA repair capacity, and accumulation of DNA damage and mutations in hematopoietic stem cells (HSCs) . In experimental settings, researchers can assess GADD45A's contribution to genomic stability by examining mutation rates, DNA repair kinetics, and chromosomal abnormalities in GADD45A-deficient versus wild-type cells.

What experimental systems are used to study GADD45A expression?

Several experimental systems are employed to study GADD45A expression:

Experimental SystemApplicationsAdvantagesLimitations
GADD45a-GFP assayGenotoxicity screeningHigh specificity (95%), rapid results, human cell-basedRequires fluorescence detection equipment
qRT-PCRQuantifying GADD45A mRNA expressionHighly sensitive, quantitativeDoes not assess protein function
Western blottingProtein expression analysisDetects post-translational modificationsSemi-quantitative
Single-cell sequencingCell-specific expression patternsReveals heterogeneity in expressionTechnically challenging
ChIP sequencingPromoter regulation studiesIdentifies transcription factor bindingRequires specialized expertise

The GADD45a-GFP human cell assay (also known as GreenScreen HC) has been extensively validated for genotoxicity screening and shows good concordance with other in vitro genotoxicity assays . When implementing this system, researchers should include appropriate positive and negative controls and ensure metabolic activation capability through S9 fraction incorporation when assessing compounds requiring metabolic conversion .

How does GADD45A expression change in response to cellular stress?

GADD45A expression increases rapidly following exposure to:

  • Genotoxic agents (DNA-damaging compounds)

  • Oxidative stress

  • Replication stress

  • Inflammatory cytokines

The magnitude of GADD45A induction correlates with the severity of cellular stress and serves as a biomarker for genotoxic damage. For example, in the GADD45a-GFP assay, the lowest effective concentration (LEC) that induces GADD45A expression can be used to rank the genotoxic potential of different compounds .

When designing experiments to measure stress-induced GADD45A expression, researchers should include time-course analyses, as expression typically peaks within 4-24 hours following exposure to stress, depending on the stressor and cell type.

What are the key molecular interactions of GADD45A?

GADD45A interacts with several key proteins and pathways:

  • p38 mitogen-activated protein kinase (MAPK) signaling - promotes cell differentiation and removal of damaged cells

  • Proliferating cell nuclear antigen (PCNA) - facilitates DNA repair

  • Cyclin-dependent kinase 1 (CDK1) - mediates cell cycle arrest

  • DNA methyltransferases - influences epigenetic regulation

  • LGR4/β-catenin pathway - regulates stem cell self-renewal

In leukemia stem cells, GADD45A acts as a downstream target of the G protein-coupled receptor (LGR)4 pathway and influences β-catenin signaling, a key regulator of self-renewal in hematopoietic development and malignancies .

Research methods to study these interactions include co-immunoprecipitation, yeast two-hybrid systems, proximity ligation assays, and FRET-based approaches.

How does GADD45A deficiency affect hematopoietic stem cell function?

GADD45A deficiency produces complex effects on hematopoietic stem cells (HSCs):

  • Increased self-renewal capacity

  • Enhanced genomic instability

  • Altered ROS regulation

  • Resistance to differentiation signals

  • Elevated risk for malignant transformation

In both murine models and patient-derived xenograft (PDX) systems, knockout of GADD45A enhances leukemia progression, increases leukemia stem cell (LSC) self-renewal, and reduces reactive oxygen species (ROS) levels . These changes contribute to therapy resistance and an increasingly aggressive phenotype upon serial transplantation.

Experimental approaches to study GADD45A deficiency in HSCs include:

  • Serial transplantation assays to assess self-renewal capacity

  • Single-cell RNA sequencing to identify altered gene expression patterns

  • Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) for simultaneous surface marker and transcriptome analysis

  • ROS measurement using flow cytometry with oxidation-sensitive dyes

  • Clonogenic assays to assess differentiation potential

What is the relationship between GADD45A and resistance to oxidative stress-induced cell death?

GADD45A loss promotes resistance to oxidative stress-induced cell death through a dual mechanism:

  • Induction of DNA damage and mutations in DNA repair genes

  • Upregulation of antioxidant pathways that maintain low ROS levels

This combination creates a paradoxical state where cells accumulate DNA damage while simultaneously gaining protection against oxidative stress-induced death mechanisms like ferroptosis.

Key antioxidant pathways upregulated with GADD45A loss include:

  • Iron storage and detoxification (e.g., FTH1)

  • Peroxiredoxin system (e.g., PRDX1)

  • Glutathione metabolism

Importantly, upregulation of these pathways correlates with unfavorable outcomes in patients with acute myeloid leukemia (AML) .

To investigate this relationship experimentally, researchers can:

  • Measure iron levels and ROS production in GADD45A-deficient cells

  • Test sensitivity to ferroptosis inducers (e.g., RSL3)

  • Perform gene expression profiling of antioxidant pathways

  • Use redox-sensitive probes to assess subcellular redox status

How does the GADD45a-GFP assay compare with other genotoxicity screening methods?

The GADD45a-GFP assay offers distinct advantages over traditional genotoxicity screening methods:

AssaySensitivitySpecificityThroughputKey AdvantagesLimitations
GADD45a-GFP75-100%95%Medium-highHuman cell-based, detects multiple genotoxic mechanismsRequires fluorescence detection
Ames/Mini-Ames58%73.9%MediumEstablished regulatory acceptanceBacterial system, limited to mutagenicity
SOS-ChromoTest45%79%MediumSimple procedurePoor efficiency for detecting clastogens
Micronucleus Test80%31%Low-mediumDetects clastogenicity and aneugenicityHigh false positive rate
Mouse Lymphoma Assay73%39%LowDetects broad range of genetic damageHigh false positive rate, low throughput

The GADD45a-GFP assay demonstrated superior performance compared to the SOS-ChromoTest in detecting genotoxic compounds. When compared with the Mini-Ames test, the GADD45a-GFP assay showed 100% sensitivity for proprietary compounds and approximately 75% sensitivity for pharmaceutical compounds in general .

While implementing the assay, researchers should consider:

  • Including metabolic activation (S9 fraction) for pro-genotoxins

  • Running appropriate positive controls (e.g., methyl methanesulfonate)

  • Testing multiple concentrations to establish dose-response relationships

  • Following standardized protocols to ensure reproducibility

What methodological considerations are important when interpreting GADD45a-GFP assay results?

Several methodological considerations are crucial when interpreting GADD45a-GFP assay results:

  • Cytotoxicity assessment: Distinguish between genotoxic effects and non-specific cytotoxicity by measuring cell viability alongside GFP expression.

  • Metabolic activation: For compounds requiring metabolic conversion, incorporate S9 fraction or other metabolic activation systems.

  • Confounding factors: The following factors may affect assay results:

    • Antibiotics can yield ambiguous results due to their secondary pharmacological properties

    • DNA synthesis inhibitors may produce false positives

    • Aneugens may not be detected efficiently

    • Apoptosis inducers can confound interpretation

  • Result classification:

    • The lowest effective concentration (LEC) should be determined

    • Dose-response relationships should be established

    • Results should be compared with benchmark compounds

  • In vivo correlation: Positive GADD45a-GFP results are not always indicative of in vivo genotoxicity, as demonstrated with proprietary compounds F and V which were genotoxic in the assay but non-genotoxic in vivo .

For accurate interpretation, it's recommended to use the assay as part of a battery of tests rather than as a standalone method for definitive genotoxicity assessment.

How does GADD45A influence leukemia stem cell maintenance and therapy resistance?

GADD45A loss enhances leukemia stem cell (LSC) maintenance and therapy resistance through several mechanisms:

  • Enhanced self-renewal: Deletion of GADD45A prevents the reduction of β-catenin induced by LGR4 depletion, maintaining a key self-renewal pathway

  • Genomic instability: GADD45A loss causes mutations in DNA repair genes and increases replication stress

  • Iron and ROS regulation: GADD45A deletion suppresses iron and ROS accumulation, contributing to ferroptosis resistance

  • Antioxidant pathway upregulation: Genes involved in iron detoxification (FTH1) and peroxide reduction (PRDX1) are upregulated

  • Differentiation block: Downregulation of GADD45A via FLT3-ITD mutation contributes to the myeloid differentiation block in AML

These mechanisms collectively promote leukemia-initiating activity and confer resistance to ROS-associated genotoxic agents, leading to an increasingly aggressive disease phenotype .

Experimental approaches to study this phenomenon include:

  • Serial transplantation assays in immunodeficient mice

  • Single-cell analysis of patient-derived xenografts

  • Ferroptosis sensitivity testing with compounds like RSL3

  • Gene expression profiling of stemness and iron metabolism genes

What are the optimal parameters for conducting the GADD45a-GFP genotoxicity assay?

For optimal GADD45a-GFP genotoxicity assay performance, researchers should consider the following parameters:

  • Cell preparation and culture conditions:

    • Use early passage TK6 cells (p53-competent human lymphoblastoid cells)

    • Maintain cells in exponential growth phase

    • Culture in RPMI 1640 medium with appropriate supplements

    • Verify cell viability (>90%) before assay initiation

  • Test compound treatment:

    • Test at least 8-10 concentrations in half-log or quarter-log intervals

    • Include solvent controls and positive controls

    • Treatment duration: 24 hours (standard protocol)

    • Maximum recommended cytotoxicity: 50% reduction in relative population doubling

  • Metabolic activation:

    • When required, use rat liver S9 fraction (1-2%)

    • Include cofactors (NADP, glucose-6-phosphate)

    • Limit S9 exposure time to avoid cytotoxicity (typically 3 hours followed by washing)

  • GFP measurement:

    • Use flow cytometry for analysis

    • Measure minimum of 10,000 events per sample

    • Assess both percentage of GFP-positive cells and mean fluorescence intensity

    • Calculate fold induction relative to solvent control

  • Data analysis and interpretation:

    • Determine lowest effective concentration (LEC) inducing significant GFP expression

    • Generate concentration-response curves

    • Calculate EC50 values where applicable

    • Compare results with reference compounds

Following these parameters ensures high assay reproducibility and reliability, as demonstrated in interlaboratory validation studies showing consistent results across multiple testing facilities .

How can GADD45A function be studied in patient-derived samples?

Studying GADD45A function in patient-derived samples requires specialized techniques:

These approaches were successfully employed to demonstrate that loss of GADD45A enhances AML progression in PDX mouse models and is associated with resistance to ferroptosis through aberrant activation of antioxidant pathways .

What experimental designs are effective for studying GADD45A in genotoxicity ranking?

Effective experimental designs for studying GADD45A in genotoxicity ranking include:

  • Comparative ranking approach:

    • Test structurally related compounds in parallel

    • Determine lowest effective concentration (LEC) for each compound

    • Rank compounds based on LEC values

    • Compare with rankings from established genotoxicity assays

  • Benchmark dose modeling:

    • Generate full dose-response curves

    • Calculate benchmark dose (BMD) values

    • Determine point of departure for risk assessment

    • Compare BMD values across compounds for relative potency ranking

  • Reference compound calibration:

    • Include well-characterized reference compounds

    • Create standard curves for different genotoxic mechanisms

    • Position test compounds relative to references

    • Account for different modes of action

  • Multiplexed endpoint analysis:

    • Measure GADD45A-GFP induction alongside other endpoints

    • Include cytotoxicity, cell cycle analysis, and γ-H2AX staining

    • Develop integrated scoring systems

    • Improve mechanistic understanding of genotoxic response

This approach was validated with hydroquinone derivatives, where the GADD45a-GFP assay successfully ranked compounds by genotoxic potential, finding the lowest LEC for hydroquinone, followed by mequinol and lastly resorcinol, in agreement with results from mammalian gene mutation tests and cytogenetic tests .

How can researchers effectively generate and validate GADD45A knockout models?

Generating and validating GADD45A knockout models requires careful methodological approaches:

  • CRISPR-Cas9 gene editing strategy:

    • Design multiple sgRNAs targeting early exons of GADD45A

    • Screen for high editing efficiency using T7 endonuclease assay

    • Select cell clones with frameshift mutations

    • Verify knockout at protein level using Western blot

  • Validation of knockout phenotype:

    • Confirm loss of GADD45A induction following genotoxic stress

    • Assess DNA damage response (γ-H2AX foci formation)

    • Measure cell cycle checkpoint activation

    • Evaluate ROS levels and sensitivity to oxidative stress

  • Functional rescue experiments:

    • Re-express wild-type GADD45A in knockout cells

    • Use inducible expression systems for temporal control

    • Include domain mutants to identify critical functional regions

    • Quantify reversal of knockout phenotypes

  • In vivo model validation:

    • Compare engraftment potential of wild-type vs. knockout cells

    • Assess leukemia development in serial transplantation

    • Monitor therapy responses, particularly to genotoxic agents

    • Perform limiting dilution assays to quantify stem cell frequency

  • Molecular profiling:

    • Perform RNA-seq to identify differentially expressed genes

    • Conduct ChIP-seq to assess epigenetic changes

    • Use proteomics to identify altered protein interactions

    • Map genomic instability using whole genome sequencing

These approaches were successfully employed to study the role of GADD45A loss in promoting leukemia-initiating activity and therapy resistance in LGR4/HOXA9-dependent AML .

What methods are used to study GADD45A's role in oxidative stress response?

Several specialized methods are used to study GADD45A's role in oxidative stress response:

  • ROS measurement techniques:

    • Flow cytometry with oxidation-sensitive dyes (DCFDA, CellROX)

    • Live-cell imaging with genetically encoded redox sensors (roGFP, HyPer)

    • Electron paramagnetic resonance (EPR) spectroscopy for specific ROS species

    • HPLC-based detection of oxidized biomolecules

  • Iron metabolism assessment:

    • Labile iron pool measurement using fluorescent probes (Calcein-AM)

    • Total cellular iron quantification using colorimetric assays

    • Ferritin and transferrin receptor expression analysis

    • Perl's Prussian blue staining for tissue iron deposits

  • Ferroptosis sensitivity testing:

    • Treatment with ferroptosis inducers (RSL3, erastin)

    • Co-treatment with ferroptosis inhibitors (ferrostatin-1, liproxstatin-1)

    • Time-lapse imaging of cell death morphology

    • Lipid peroxidation measurement (BODIPY-C11, MDA assay)

  • Antioxidant pathway analysis:

    • Gene expression profiling of antioxidant genes (NRF2 pathway)

    • Activity assays for key antioxidant enzymes

    • Glutathione level measurement

    • Protein-glutathione adduct detection

  • In vivo oxidative stress models:

    • Treatment with pro-oxidant compounds

    • Genetic models with altered antioxidant capacity

    • Tissue-specific oxidative damage assessment

    • Correlation with GADD45A expression levels

Using these methods, researchers demonstrated that deletion of GADD45A in leukemia stem cells reduces levels of reactive oxygen species and confers resistance to ROS-associated genotoxic agents like the ferroptosis inducer RSL3, through aberrant activation of antioxidant pathways related to iron and ROS detoxification, including FTH1 and PRDX1 .

How does GADD45A expression correlate with cancer prognosis?

GADD45A expression patterns have significant prognostic implications in various cancers:

Methodologically, analyzing GADD45A expression in patient samples can involve:

  • Immunohistochemistry for protein expression in tissue sections

  • qRT-PCR for mRNA quantification

  • Methylation-specific PCR for promoter methylation status

  • Integration with other prognostic markers

Upregulation of GADD45A-regulated antioxidant pathways, such as FTH1 and PRDX1, correlates with unfavorable outcomes in patients with AML, providing additional prognostic indicators .

How can GADD45A status be used to predict therapy response?

GADD45A status can serve as a predictive biomarker for therapy response through several mechanisms:

  • Genotoxic therapy sensitivity:

    • Low GADD45A expression correlates with resistance to DNA-damaging agents

    • Impaired DNA damage response and checkpoint activation

    • Possible synthetic lethality opportunities

  • Oxidative stress-based therapies:

    • GADD45A-deficient cells show resistance to ferroptosis inducers like RSL3

    • Upregulated antioxidant pathways protect against ROS-generating treatments

    • Potential for combination strategies targeting antioxidant defenses

  • Epigenetic therapy response:

    • Methylation status of GADD45A may predict response to hypomethylating agents

    • Relationship with DNMT3A and IDH1/2 mutations

  • Patient stratification approach:

    • Integrate GADD45A status with other molecular markers

    • Develop predictive algorithms for treatment decision-making

    • Consider alternative therapies for GADD45A-deficient cases

These predictive applications are supported by findings that deletion of GADD45A enhances resistance to ROS-associated genotoxic agents and acquisition of an increasingly aggressive phenotype on serial transplantation in mice .

What therapeutic strategies could target GADD45A pathways in cancer?

Several therapeutic strategies could potentially target GADD45A pathways in cancer:

  • Restoring GADD45A expression:

    • Epigenetic modifiers (DNMT inhibitors, HDAC inhibitors)

    • Targeted demethylation technologies

    • Small molecules that enhance GADD45A transcription

  • Exploiting synthetic lethality:

    • DNA repair inhibitors (PARP inhibitors in GADD45A-deficient cells)

    • Cell cycle checkpoint inhibitors

    • Replication stress inducers

  • Targeting antioxidant dependencies:

    • System xc- inhibitors to deplete glutathione

    • Thioredoxin system inhibitors

    • NRF2 pathway modulators

  • Iron-targeted approaches:

    • Iron chelators or ionophores

    • Ferritin degradation inducers

    • Combination with ferroptosis sensitizers

  • Differentiation therapy:

    • p38 MAPK pathway activators to mimic GADD45A-induced differentiation

    • Combination with standard differentiation agents

These approaches are informed by research demonstrating that GADD45A loss promotes leukemia through dual mechanisms of promoting DNA damage while upregulating antioxidant pathways that sustain low ROS levels essential for maintaining self-renewal capacity .

How does GADD45A expression change during cancer progression?

GADD45A expression undergoes dynamic changes during cancer progression:

  • Initiation phase:

    • Initially may increase as a stress response

    • Acts as a tumor suppressor by promoting DNA repair and cell cycle arrest

    • Helps eliminate damaged cells through differentiation or apoptosis

  • Progression phase:

    • Often downregulated through epigenetic silencing

    • Loss provides survival advantage to cancer cells

    • Contributes to genomic instability and mutation accumulation

  • Advanced disease:

    • Persistent loss associated with aggressive phenotypes

    • Contributes to therapy resistance

    • Enables stem cell-like properties in cancer cells

  • Metastatic disease:

    • Altered expression affects migration and invasion

    • Influences interaction with tumor microenvironment

This progression pattern is supported by findings in leukemia models, where deletion of GADD45A enhances AML progression in murine and patient-derived xenograft models, induces substantial mutations, increases leukemia stem cell self-renewal and stemness in vivo, and leads to an increasingly aggressive phenotype on serial transplantation .

What is the relationship between GADD45A and epigenetic regulation in cancer?

GADD45A has complex relationships with epigenetic regulation in cancer:

These findings underscore the importance of GADD45A in epigenetic regulation and suggest potential therapeutic vulnerabilities that could be exploited in cancer treatment .

What are emerging technologies for studying GADD45A function?

Emerging technologies providing new insights into GADD45A function include:

  • Single-cell multi-omics approaches:

    • Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq)

    • Single-cell ATAC-seq for chromatin accessibility

    • Spatial transcriptomics for tissue context

    • Integrated analysis platforms for multi-dimensional data

  • Advanced genome editing technologies:

    • Base editing for precise mutation introduction

    • Prime editing for targeted sequence changes

    • Epigenome editing to modulate GADD45A expression

    • Combinatorial CRISPR screens for pathway interactions

  • Live-cell imaging innovations:

    • CRISPR-based endogenous tagging

    • Optogenetic control of GADD45A expression

    • Super-resolution microscopy for protein localization

    • Real-time reporters for DNA damage and repair

  • Computational and AI approaches:

    • Machine learning for predicting GADD45A interactions

    • Network analysis of GADD45A-dependent pathways

    • Patient data integration for clinical correlations

    • Virtual screening for GADD45A pathway modulators

These technologies will enable more comprehensive understanding of GADD45A's role in normal and malignant cell biology, potentially revealing new therapeutic opportunities.

How might targeting GADD45A pathways improve cancer therapy?

Targeting GADD45A pathways could improve cancer therapy through several innovative approaches:

  • Synthetic lethality strategies:

    • Identify dependencies created by GADD45A loss

    • Target compensatory DNA repair mechanisms

    • Exploit vulnerabilities in stress response pathways

    • Combine with existing DNA-damaging therapies

  • Metabolic vulnerability targeting:

    • Focus on altered iron metabolism in GADD45A-deficient cells

    • Develop improved ferroptosis inducers with greater selectivity

    • Target antioxidant systems upregulated in GADD45A-deficient cells

    • Combine with metabolic inhibitors affecting redox balance

  • Stem cell targeting approaches:

    • Develop agents that selectively target leukemia stem cells with GADD45A loss

    • Disrupt self-renewal pathways dependent on GADD45A status

    • Force differentiation of therapy-resistant stem cells

    • Target the stem cell niche interactions

  • Biomarker-guided personalized therapy:

    • Stratify patients based on GADD45A expression/mutation status

    • Develop companion diagnostics for GADD45A pathway activation

    • Tailor therapy selection based on GADD45A-dependent vulnerabilities

    • Monitor changes in GADD45A status during treatment

These approaches could improve outcomes for patients with GADD45A-deficient cancers by addressing the specific mechanisms through which GADD45A loss promotes stemness, therapy resistance, and disease progression .

What are unresolved questions about GADD45A in human diseases?

Several critical questions about GADD45A in human diseases remain unresolved:

  • Tissue-specific functions:

    • How does GADD45A function differ across tissue types?

    • What determines context-dependent tumor suppressor versus oncogenic roles?

    • How do tissue-specific interaction partners influence function?

  • Therapeutic resistance mechanisms:

    • How does GADD45A loss contribute to therapy resistance at the molecular level?

    • What compensatory pathways emerge following GADD45A loss?

    • How can these resistance mechanisms be overcome?

  • Regulation of GADD45A expression:

    • What are the key transcriptional and post-transcriptional regulators?

    • How do microRNAs and long non-coding RNAs affect expression?

    • What environmental factors influence GADD45A levels?

  • Role in non-cancer diseases:

    • How does GADD45A contribute to inflammatory and degenerative conditions?

    • What is its role in aging-related pathologies?

    • How does it function in metabolic diseases?

  • Therapeutic targeting:

    • Can GADD45A expression be selectively restored in cancer cells?

    • What are the most effective approaches to target GADD45A-deficient cells?

    • How can GADD45A status be effectively monitored during treatment?

Addressing these questions will require integrated approaches combining basic research, clinical studies, and advanced technologies to fully understand GADD45A's complex roles in human health and disease.

How do GADD45A family members differ in function and regulation?

GADD45A belongs to a family of stress sensors with distinct functional characteristics:

Family MemberKey FunctionsRegulationTissue ExpressionDisease Associations
GADD45ADNA damage response, Cell cycle control, ROS regulationp53-responsive, NF-κB regulated, Methylation-sensitiveUbiquitous, high in lymphoid tissuesCancer, inflammation
GADD45BImmune response, Inflammatory signaling, mRNA stabilityNF-κB dependent, JNK pathway regulatedHigh in immune cellsAutoimmune diseases, inflammation
GADD45GDifferentiation, Growth suppression, p38 MAPK activationOften silenced in tumors, Developmentally regulatedTissue-specific expression patternDevelopmental disorders, cancer

Key differences in function and regulation include:

  • Stimulus specificity:

    • GADD45A: Primarily responsive to genotoxic stress

    • GADD45B: Predominantly induced by inflammatory cytokines

    • GADD45G: Often associated with differentiation signals

  • Pathway interactions:

    • GADD45A: Strong p53 pathway association

    • GADD45B: More prominent NF-κB pathway connections

    • GADD45G: Often linked to developmental pathways

  • Experimental approaches to distinguish functions:

    • Isoform-specific antibodies and expression constructs

    • Selective knockdown using siRNA/shRNA

    • Chromatin immunoprecipitation to identify binding sites

    • Interactome analysis for protein-protein interactions

Understanding these differences is crucial for targeting specific GADD45 family members in therapeutic approaches and for interpreting experimental results correctly.

What computational approaches can advance GADD45A research?

Advanced computational approaches are transforming GADD45A research:

  • Network biology approaches:

    • Protein-protein interaction network analysis

    • Pathway enrichment and perturbation modeling

    • Gene regulatory network inference

    • Identification of central nodes and vulnerabilities

  • Machine learning applications:

    • Prediction of GADD45A expression from multi-omics data

    • Classification of patient samples based on GADD45A pathway activity

    • Drug response prediction based on GADD45A status

    • Feature selection to identify key GADD45A-associated biomarkers

  • Structural biology and molecular modeling:

    • Protein structure prediction using AlphaFold or similar tools

    • Molecular dynamics simulations of GADD45A interactions

    • Virtual screening for compounds targeting GADD45A pathways

    • Protein-protein docking to identify interaction interfaces

  • Integrative multi-omics approaches:

    • Integration of genomic, transcriptomic, proteomic, and metabolomic data

    • Single-cell data analysis and trajectory inference

    • Temporal modeling of GADD45A pathway dynamics

    • Patient stratification based on integrated biomarker panels

  • Clinical data mining and bioinformatics:

    • Analysis of GADD45A expression across cancer databases (TCGA, ICGC)

    • Correlation with clinical outcomes and therapy responses

    • Development of prognostic and predictive models

    • Meta-analysis of published GADD45A studies

These computational approaches can accelerate discovery by generating hypotheses, identifying patterns not obvious through traditional methods, and providing integrative frameworks for understanding GADD45A's complex roles in cellular physiology and disease.

Product Science Overview

Introduction

Growth Arrest and DNA-Damage-Inducible Alpha (GADD45A) is a protein encoded by the GADD45A gene in humans. This protein is part of the GADD45 family, which includes GADD45B and GADD45G. These proteins are known for their roles in responding to environmental stresses, such as DNA damage, and are involved in various cellular processes including DNA repair, cell cycle control, and apoptosis . GADD45A is particularly notable for its ability to mediate the activation of the p38/JNK pathway via MTK1/MEKK4 kinase .

Preparation Methods

The preparation of human recombinant GADD45A involves several steps. Typically, the gene encoding GADD45A is cloned into an expression vector, which is then introduced into a suitable host cell, such as E. coli or yeast. The host cells are cultured under conditions that promote the expression of the recombinant protein. After sufficient growth, the cells are harvested and lysed to release the protein. The recombinant GADD45A is then purified using techniques such as affinity chromatography, which exploits the protein’s specific binding properties to isolate it from other cellular components .

Chemical Reactions Analysis

GADD45A plays a crucial role in various biochemical pathways. It is involved in the regulation of the cell cycle and DNA repair mechanisms. One of the key interactions of GADD45A is with the ATP synthase F1 subunit alpha (ATP5A1), where it affects the ubiquitination and degradation of ATP5A1. This interaction leads to decreased ATP synthesis and inactivation of the cAMP/PKA/LKB1 signaling pathway . Additionally, GADD45A has been shown to influence the expression of adipogenic genes, promoting intramuscular preadipocyte adipogenesis .

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.