A recombinant version produced in E. coli is used for research:
G3BP1 initiates stress granule (SG) formation under cellular stress (e.g., oxidative or viral stress), sequestering untranslated mRNAs to halt protein synthesis .
Stabilization: Binds and stabilizes transcripts like Ctnnbl and CDK7 to regulate cardiac hypertrophy and neuronal differentiation .
Degradation: Promotes decay of CDK9 and PMP22 mRNAs, influencing cell proliferation .
G3BP1 interacts with Ras-GTPase-activating protein (Ras-GAP) via its SH3 domain, modulating Ras-mediated cell proliferation and differentiation .
Antiviral Role: Activates RIG-I and cGAS to detect RNA/DNA viruses .
Viral Hijacking: SARS-CoV-2 and CHIKV manipulate G3BP1 to evade immune responses .
Embryonic Lethality: G3bp1 knockout in mice causes neuronal apoptosis in the hippocampus and cortex .
Tau Regulation: Stabilizes Tau mRNA via IMP-1 interactions, affecting neuronal differentiation .
To investigate G3BP1-mediated stress granule assembly, researchers should combine live-cell imaging with immunofluorescence using validated antibodies against G3BP1 and co-markers like TIAR or PABP1 . Stress induction protocols (e.g., sodium arsenite treatment at 0.5 mM for 30–60 minutes) should be standardized across cell lines. Quantitative analysis of granule size and number can be performed using automated image analysis tools (e.g., CellProfiler). For dynamic assembly/disassembly studies, fluorescence recovery after photobleaching (FRAP) on GFP-tagged G3BP1 constructs is recommended .
Electrophoretic mobility shift assays (EMSAs) using purified G3BP1 protein and fluorescently labeled RNA probes (e.g., AU-rich elements) are foundational. Include competition assays with unlabeled RNA to confirm specificity. For higher throughput, RNA-protein crosslinking immunoprecipitation (CLIP-seq) in cellular models identifies endogenous RNA targets . Structural validation requires nuclear magnetic resonance (NMR) or cryo-EM to resolve interactions between G3BP1’s RRM/RGG domains and RNA substrates .
Include rescue experiments with wild-type G3BP1 transfection in knockout models to confirm phenotype specificity. Use multiple guide RNAs for CRISPR-Cas9-mediated knockout to rule off-target effects. Parallel analysis of stress granule markers (e.g., TIA-1) and polysome profiling ensures observed effects are G3BP1-specific . For aggregation-prone models (e.g., Huntington’s disease), quantify both soluble and insoluble protein fractions via filter trap assays .
Early studies proposed direct binding via G3BP1’s SH3 domain, but recent work challenges this . To reconcile discrepancies:
Perform co-immunoprecipitation (co-IP) under varying lysis conditions (e.g., high-salt buffers to disrupt weak interactions).
Use proximity ligation assays (PLA) to visualize intracellular interaction spatiality.
Employ isothermal titration calorimetry (ITC) to measure binding affinity in vitro.
Validate findings across cell types (e.g., HEK293 vs. neuronal cells) to assess context dependency .
Use pooled siRNA libraries with ≥3 independent sequences targeting distinct G3BP1 exons.
Combine knockdown with pharmacological inhibition (e.g., with the G3BP1 inhibitor C108) to confirm phenotype reproducibility .
Perform RNA sequencing post-knockdown to identify unintended transcriptome-wide effects.
Cross-validate results in inducible knockout models to separate acute vs. chronic effects .
G3BP1’s intrinsically disordered region 1 (IDR1) undergoes phosphorylation at S149/S232 under basal conditions, which suppresses RNA binding. To study this:
Generate phosphomimetic (S149D/S232D) and phospho-null (S149A/S232A) mutants.
Compare their phase separation propensity using in vitro droplet assays with recombinant proteins and RNA.
Monitor stress granule dynamics in live cells expressing these mutants under oxidative stress .
Patient-derived iPSCs: Differentiate into striatal neurons for Huntington’s disease studies; assess mutant HTT aggregation via immunofluorescence and FRET-based biosensors .
C. elegans models: Use strains expressing polyQ-expanded HTT in neurons; perform RNAi knockdown of gtbp-1 (ortholog of G3BP1) to quantify aggregation and motility defects .
Transgenic mice: Cross G3BP1 haploinsufficient mice with HD models (e.g., R6/2) to evaluate disease progression.
Utilize G3BP1-ΔRBP mutants lacking RNA-binding domains, which cannot form stress granules but retain protein interaction capacity .
Compare proteasomal degradation rates of aggregation-prone proteins (e.g., mutant HTT) in cells expressing wild-type vs. ΔRBP G3BP1 under stress .
Perform ubiquitination assays to determine if G3BP1 enhances mutant HTT’s ubiquitin-proteasome targeting independently of granules .
This dichotomy reflects cellular adaptation vs. exhaustion. Acute stress transiently upregulates G3BP1 to promote stress granule assembly, as shown by cycloheximide chase assays . Chronic stress (e.g., in HD) leads to G3BP1 sequestration into persistent granules, reducing soluble pools available for proteostasis. Quantify nuclear vs. cytoplasmic G3BP1 via subcellular fractionation in longitudinal studies .
Cell panel screening: Compare G3BP1 interactomes (via AP-MS) in neuronal (SH-SY5Y), epithelial (HeLa), and immortalized (HEK293) lines.
Tissue-specific knockouts: Use Cre-lox systems to delete G3BP1 in murine CNS vs. peripheral tissues.
Single-cell RNA-seq: Profile G3BP1 expression heterogeneity in human postmortem brain regions .
Crosslinking and isolation by pull-down (CLIP) combined with next-generation sequencing identifies target transcripts.
Single-molecule fluorescence in situ hybridization (smFISH) quantifies co-localization of G3BP1 with specific mRNAs in granules.
HDX-MS (hydrogen-deuterium exchange mass spectrometry) maps RNA-induced conformational changes in G3BP1 .
Purify full-length G3BP1 with an N-terminal His tag using size-exclusion chromatography.
Induce phase separation in buffer containing 150 mM NaCl, 10% PEG-8000, and 1 mg/mL total RNA.
Quantify droplet formation via differential interference contrast (DIC) microscopy and turbidity assays at 600 nm .
While G3BP1 is widely used, its absence does not preclude granule formation in certain cell types. Always co-stain with ≥2 additional markers (e.g., TIA-1, eIF4E). Note that G3BP1 antibodies may cross-react with paralogs (G3BP2); validate using knockout cell lysates in Western blots .
GTPase Activating Protein (SH3 domain) Binding Protein 1, commonly referred to as G3BP1, is a multifunctional protein that plays a crucial role in various cellular processes. It is encoded by the G3BP1 gene located on chromosome 5q33.1 in humans . G3BP1 is known for its involvement in stress granule assembly, RNA binding, and regulation of microRNA processing.
G3BP1 is a member of the heterogeneous nuclear RNA-binding proteins and is involved in the Ras signal transduction pathway . It has several key domains, including the SH3 domain, which allows it to interact with other proteins and regulate their activity. G3BP1 can bind to partially unwound DNA and RNA substrates, and it exhibits endonuclease activity .
One of the primary functions of G3BP1 is its role in stress granule formation. Stress granules are cytoplasmic aggregates of proteins and RNAs that form in response to cellular stress. G3BP1 is a key component of these granules and helps in their assembly by interacting with other proteins .
G3BP1 has been shown to regulate the processing of microRNA-1 (miR-1) during cardiac hypertrophy . During cardiac hypertrophy, G3BP1 is upregulated and binds to the consensus sequence in the pre-miR-1-2 stem-loop, restricting the processing of miR-1. This results in a decrease in mature miR-1 levels and an increase in the expression of its target genes, which are essential for transcription and translation .
G3BP1 interacts with various proteins to regulate different cellular processes. For example, it interacts with USP10, a deubiquitinating enzyme, and SND1, a component of the RNA-induced silencing complex . These interactions are crucial for the regulation of stress granule assembly and other cellular functions.
The dysregulation of G3BP1 has been implicated in several diseases, including cancer and cardiac hypertrophy. Its role in stress granule formation and microRNA processing makes it a potential target for therapeutic interventions. Understanding the function and regulation of G3BP1 can provide insights into the development of novel treatments for these conditions.