RAB3D Human

RAB3D, Member RAS Oncogene Family Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in Cellular Processes

RAB3D regulates regulated exocytosis and vesicle transport across diverse cell types, with specialized roles in non-neuronal tissues.

Exocytosis and Secretory Pathways

  • Mechanism: Binds GTP to activate effector proteins, facilitating vesicle docking and fusion .

  • Cell-Type Specificity:

    • Mast Cells: Localized to secretory granules; translocates to plasma membrane post-degranulation .

    • Pancreatic Cells: Regulates apical transcytosis in epithelial tissues .

  • Structural Insights: GDP-bound crystal structure reveals conserved switch regions critical for conformational changes .

Cancer Progression and Metastasis

RAB3D overexpression correlates with tumor malignancy and metastasis via multiple pathways:

Pathway/ProcessMechanismCancer TypeSource
EMT InductionActivates AKT/GSK-3β/Snail signaling, downregulating E-cadherin and upregulating N-cadherinBreast, Prostate, Lung
Exosome SecretionEnhances release of pro-metastatic exosomes containing Hsp90α and EMT markersBreast, Osteosarcoma
Cell Migration/InvasionUpregulates MMPs and CDK4/6, promoting motility and cycle progressionOsteosarcoma, Skin

Experimental Evidence:

  • In Vivo Models: Rab3D-overexpressing MCF-7 cells show invasive growth in mammary fat pads and lung metastasis .

  • Clinical Correlation: High Rab3D levels in breast, prostate, lung, and colon cancers correlate with advanced TNM staging .

Clinical and Diagnostic Relevance

Disease/ConditionAssociation with RAB3DSource
Cancer MetastasisElevated expression promotes invasion; potential therapeutic target
DacryoadenitisLinked to inflammatory secretory dysfunction
Warburg Micro Syndrome 1Rare genetic disorder involving exocytosis defects
Biomarker PotentialOverexpression in multiple cancers suggests use as a broad-spectrum diagnostic

Diagnostic Utility:

  • miR-506-3p Regulation: Downregulation of miR-506-3p leads to Rab3D upregulation, driving osteosarcoma progression .

  • Therapeutic Targeting: Inhibiting Rab3D may suppress metastasis by blocking EMT and exosome secretion .

Research Gaps and Future Directions

  1. Mechanistic Insights:

    • Effector Proteins: Limited identification of Rab3D-specific effectors beyond GTPase cycling regulators .

    • Tissue-Specific Roles: Divergent functions in adipose tissue vs. neural cells warrant further study .

  2. Therapeutic Strategies:

    • Small-Molecule Inhibitors: Development of Rab3D-specific inhibitors for metastatic cancers .

    • Combination Therapies: Synergy with EMT inhibitors or exosome-targeting agents .

Product Specs

Introduction
Rab-3D (RAB3D), a member of the Ras superfamily of small Rab GTPases, plays a crucial role in endocytosis or biosynthetic protein transport. RAB3D mRNA is primarily found in white adipose tissue and increases during the differentiation of 3T3-L1 fibroblasts, similar to GLUT4. Structurally, RAB3D closely resembles Rab 3A, an isoform known for its involvement in regulated exocytosis across various cell types.
Description
Produced in E. coli, the non-glycosylated polypeptide chain of RAB3D comprises 239 amino acids (1-219 a.a) with a molecular mass of 26.4 kDa. However, it appears larger on SDS-PAGE due to the N-terminal 20 amino acid His-tag. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
RAB3D protein solution at a concentration of 0.5 mg/ml is supplied in a buffer of 20mM Tris-HCl (pH 8.0), 1mM DTT, 30% glycerol, and 0.2M NaCl.
Stability
While RAB3D Human Recombinant remains stable at 4°C for up to one week, long-term storage is recommended at temperatures below -18°C. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity greater than 90.0%.
Synonyms

Ras-related protein Rab-3D, RAB3D, GOV, RAB16, D2-2, RAD3D.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MASAGDTQAG PRDAADQNFD YMFKLLLIGN SSVGKTSFLF RYADDSFTPA FVSTVGIDFK VKTVYRHDKR IKLQIWDTAG QERYRTITTA YYRGAMGFLL MYDIANQESF AAVQDWATQI KTYSWDNAQV ILVGNKCDLE DERVVPAEDG RRLADDLGFE FFEASAKENI NVKQVFERLV DVICEKMNES LEPSSSSGSN GKGPAVGDAP APQPSSCSC

.

Q&A

What is the fundamental role of RAB3D in human cellular function?

RAB3D belongs to the largest family of small GTPases and primarily regulates intracellular membrane trafficking. In human cells, RAB3D associates with secretory vesicles in various exocrine and endocrine cells, where it coordinates regulated exocytosis. More recent research has expanded its known functions to include regulation of cell signaling, division, survival, and migration. These processes generally occur through recruitment of effectors and regulatory proteins that control RAB3D's membrane association and activation state. Understanding RAB3D's function is critical for investigating both normal physiological processes and disease mechanisms .

How does RAB3D differ from other RAB3 isoforms in expression patterns?

While RAB3 family includes four isoforms (RAB3A, RAB3B, RAB3C, and RAB3D), their expression varies significantly across tissues. In mast cells, RAB3D transcripts are at least 10-fold more abundant than other isoforms, and RAB3D protein is approximately 60-fold more abundant than RAB3B. Interestingly, RAB3D is more prevalent in certain secretory cells than in brain tissue, where other RAB3 isoforms predominate. This differential expression suggests specialized roles for RAB3D in secretory processes outside the central nervous system .

What is the subcellular distribution pattern of RAB3D in secretory cells?

RAB3D exhibits a distinctive subcellular localization pattern. In alveolar epithelial type II cells, RAB3D associates with approximately 24% of lamellar bodies (secretory vesicles), primarily those positioned near the apical plasma membrane where exocytosis occurs. Similarly, in mast cells, RAB3D completely colocalizes with secretory granules, displaying a concentration gradient with higher levels in peripheral granules compared to central ones. Following stimulated exocytosis, RAB3D translocates to the plasma membrane where it remains for at least 15 minutes, suggesting a role in post-fusion events .

What evidence supports RAB3D as a biomarker for cancer progression?

Multiple studies have demonstrated elevated RAB3D levels in clinical samples from diverse cancer types including breast, prostate, lung, colon, ovary, liver and skin cancers. Importantly, RAB3D expression correlates strongly with tumor TNM staging rather than tumor grade, suggesting it specifically reflects cancer progression and metastatic potential rather than differentiation status. In immunohistochemical analyses, 92 out of 100 malignant cases showed positive RAB3D expression, while all normal tissues had scores of zero, indicating strong specificity as a potential diagnostic biomarker .

Through what molecular mechanisms does RAB3D promote cancer metastasis?

RAB3D promotes metastasis through multiple interconnected mechanisms:

  • Cytoskeletal reorganization: RAB3D facilitates membrane ruffle formation and actin cytoskeleton rearrangements essential for cell motility

  • Matrix degradation: RAB3D upregulates matrix metalloproteinase-9 (MMP-9) activity, enhancing extracellular matrix degradation

  • Exosome regulation: RAB3D mediates exosome release containing pro-metastatic factors

  • Secretion control: RAB3D regulates Hsp90α secretion, a driver of epithelial-mesenchymal transition

  • EMT pathway activation: RAB3D activates the Akt/GSK-3β/Snail pathway that promotes cancer cell invasion

In vivo studies confirm that RAB3D overexpression enhances tumor invasiveness and metastatic potential, while its knockdown significantly reduces metastatic colonies in lung tissues .

What experimental approaches are most effective for studying RAB3D in cancer models?

For comprehensive investigation of RAB3D in cancer, researchers should implement a multi-faceted experimental strategy:

ApproachApplicationAdvantagesLimitations
siRNA knockdownAcute loss-of-functionRapid implementation, specificityTransient effect
Stable shRNA expressionLong-term studiesPersistent knockdown, suitable for in vivoPotential compensation
GFP-RAB3D overexpressionGain-of-function, localizationVisual tracking, functional analysisPotential artifacts from overexpression
Mutant expression (e.g., Q81L)GTP-locked active state analysisInterrogates specific activation statesMay not reflect physiological regulation
Xenograft modelsIn vivo metastasis assessmentPhysiological context, multiple endpointsSpecies differences, immunodeficiency

These approaches should be combined with functional assays measuring cell migration, invasion, ruffle formation, MMP activity, and exosome release to comprehensively characterize RAB3D's role in cancer progression .

How does RAB3D coordinate with the actin cytoskeleton during exocytosis?

RAB3D demonstrates an intimate functional relationship with the actin cytoskeleton during regulated exocytosis. Research in alveolar epithelial type II cells has identified distinct secretory vesicle subpopulations based on RAB3D and actin association: RAB3D-positive vesicles (24%), RAB3D-positive and actin-coated vesicles (2%), and actin-coated vesicles without RAB3D (9%). These likely represent sequential stages in the exocytotic pathway, suggesting RAB3D first associates with secretory vesicles positioned near the plasma membrane, then participates in actin coating, followed by RAB3D dissociation while actin remains. This sequence implies that RAB3D release and actin coating are mechanistically linked processes essential for regulated secretion .

What methodologies are optimal for visualizing RAB3D-vesicle interactions?

To effectively investigate RAB3D-vesicle interactions, researchers should consider multiple complementary approaches:

  • Immunofluorescence co-localization: Using antibodies against RAB3D together with vesicle-specific markers (e.g., p180 lamellar body protein, Clara Cell Secretory Protein) enables quantification of association rates

  • Immunoelectron microscopy: Provides ultrastructural resolution to precisely locate RAB3D on specific vesicle subdomains

  • Phalloidin co-staining: Combines RAB3D detection with F-actin visualization to examine RAB3D-actin relationships

  • Live-cell imaging: Using fluorescently-tagged RAB3D constructs allows real-time monitoring of trafficking dynamics

  • Subcellular fractionation: Western blot analysis of isolated vesicle fractions quantifies RAB3D association

These techniques should be applied in both resting and stimulated conditions to capture the dynamic nature of RAB3D-vesicle interactions during the secretory process .

How does RAB3D function differ between neuronal and non-neuronal secretory systems?

RAB3D exhibits important functional distinctions between neuronal and non-neuronal secretory systems. In neurons, other RAB3 isoforms (particularly RAB3A) predominate and regulate synaptic vesicle exocytosis and neurotransmitter release. In contrast, RAB3D is the principal RAB3 isoform in many non-neuronal secretory cells, including mast cells and alveolar type II cells. The total mass of RAB3 proteins in non-neuronal cells (e.g., RBL mast cells) is approximately 10-fold less than in brain tissue, suggesting different stoichiometric relationships with effector proteins. Additionally, RAB3D trafficking patterns differ between systems - in mast cells following stimulation, RAB3D translocates to the plasma membrane and remains there for extended periods, a pattern distinct from the rapid recycling observed with neuronal RAB3 isoforms .

What are the key RAB3D interaction partners and how should they be investigated?

While the complete RAB3D interactome remains to be fully characterized, several approaches are recommended for identifying and validating RAB3D interaction partners:

  • Yeast two-hybrid screening: Useful for initial identification of potential binding partners

  • Co-immunoprecipitation: Validates interactions in cellular contexts

  • Proximity labeling methods (BioID, APEX): Identifies transient or compartment-specific interactions

  • Fluorescence resonance energy transfer (FRET): Examines interactions in living cells

  • Nucleotide-state specific pulldowns: Distinguishes between GTP- and GDP-bound RAB3D interactors

Researchers should focus on proteins involved in vesicle tethering, membrane fusion, and cytoskeletal regulation, as these are likely functional partners based on RAB3D's known roles. Additionally, cancer-specific interaction partners should be investigated to understand context-dependent functions in tumor progression .

How can researchers address discrepancies between RAB3D mRNA and protein expression data?

Researchers have noted inconsistencies between RAB3D mRNA levels and protein expression in cancer samples, suggesting important post-transcriptional regulation. To address these discrepancies, implement the following methodological considerations:

  • Parallel analysis: Simultaneously assess both mRNA (RT-PCR, qPCR) and protein (Western blot, IHC) from the same samples

  • Multiple detection methods: Verify findings using independent techniques with different sensitivity profiles

  • Post-transcriptional mechanism investigation: Examine microRNA targeting, RNA-binding protein effects, and translation efficiency

  • Protein stability assessment: Measure RAB3D protein half-life in different cellular contexts using cycloheximide chase assays

  • Regulatory factor identification: Perform screens for factors that specifically affect RAB3D protein but not mRNA levels

These approaches can reveal important regulatory mechanisms that contribute to RAB3D dysregulation in disease states and potentially identify new therapeutic targets .

What are the most promising approaches for targeting RAB3D therapeutically?

Based on RAB3D's role in cancer progression and metastasis, several therapeutic approaches warrant investigation:

  • Small molecule inhibitors: Developing compounds that specifically inhibit RAB3D GTPase activity or interfere with key protein-protein interactions

  • RNA interference therapies: Using siRNA or antisense oligonucleotides for targeted RAB3D knockdown

  • Exosome pathway modulators: Targeting RAB3D-mediated exosome release mechanisms

  • Combination approaches: Pairing RAB3D inhibition with cytoskeleton-targeting drugs to synergistically block metastasis

  • EMT pathway intervention: Disrupting the RAB3D-Akt/GSK-3β/Snail axis to prevent epithelial-mesenchymal transition

When developing such approaches, researchers must consider potential off-target effects on physiological secretory processes in non-cancerous tissues, particularly in granulocytes and alveolar epithelial cells where RAB3D has important normal functions .

What statistical approaches should be applied when analyzing RAB3D expression in clinical samples?

When analyzing RAB3D expression in clinical contexts, researchers should implement rigorous statistical methodologies:

  • Expression correlation analysis:

    • Spearman rank correlation for non-parametric association with clinical parameters

    • Multiple regression models to account for confounding variables

    • ROC curve analysis to determine optimal diagnostic cutoff values

  • Survival analysis:

    • Kaplan-Meier curves stratified by RAB3D expression levels

    • Cox proportional hazards regression for multivariate survival analysis

    • Time-dependent AUC analysis for prognostic performance evaluation

  • Expression pattern analysis:

    • Hierarchical clustering to identify patient subgroups with similar expression profiles

    • Principal component analysis to reduce dimensionality when analyzing multiple markers

    • Random forest algorithms for identifying predictive biomarker combinations

These analyses should incorporate appropriate sample size calculations, multiple testing corrections, and validation in independent cohorts to ensure robust and clinically meaningful results .

How should researchers design experiments to distinguish RAB3D-specific effects from general exocytosis perturbations?

Distinguishing RAB3D-specific effects from general secretory pathway disruption requires careful experimental design:

  • Isoform specificity controls:

    • Include parallel manipulations of other RAB3 isoforms (RAB3A/B/C)

    • Use RAB proteins from different subfamilies (e.g., RAB27) as comparative controls

  • Rescue experiments:

    • Complement RAB3D knockdown with wild-type or mutant RAB3D expression

    • Test whether specific RAB3D domains are necessary and sufficient for observed phenotypes

  • Targeted functional readouts:

    • Measure multiple secretory parameters beyond bulk exocytosis

    • Examine vesicle subpopulation dynamics, particularly those associated with RAB3D

  • Temporal analysis:

    • Use acute manipulation systems (e.g., chemical-genetic approaches) to minimize compensatory adaptations

    • Implement time-course studies to identify primary versus secondary effects

  • Spatial resolution:

    • Apply super-resolution microscopy to precisely locate RAB3D during secretory events

    • Implement optogenetic tools for spatiotemporally controlled manipulation

These approaches can help delineate RAB3D's specific contributions to complex cellular processes beyond general secretory functions .

Product Science Overview

Overview

RAB3D, a member of the RAS oncogene family, is a protein-coding gene that plays a crucial role in various cellular processes. It is part of the Rab family of small GTPases, which are involved in the regulation of intracellular membrane trafficking. The RAB3D gene is located on chromosome 19 and encodes a protein that is primarily involved in the regulation of exocytosis and protein transport .

Gene and Protein Structure

The RAB3D gene is composed of multiple exons and introns, and its expression results in the production of a protein that consists of 219 amino acids. The protein has a molecular mass of approximately 26.9 kDa and contains a GTP-binding domain, which is essential for its function as a molecular switch in intracellular signaling pathways .

Function

RAB3D is primarily involved in the regulation of the secretory pathway. It plays a significant role in the exocytosis of secretory vesicles, which is a process crucial for the release of various substances, including hormones, neurotransmitters, and enzymes, from cells. Additionally, RAB3D is involved in bone resorption, a process where bone tissue is broken down to release minerals into the bloodstream .

Expression and Localization

RAB3D is expressed in various tissues, including the lymphoid tissue, bone marrow, testis, and skeletal muscle. It is localized in the cytoplasmic microtubules and secretory vesicles, where it interacts with other proteins to facilitate its role in exocytosis and protein transport .

Clinical Significance

Mutations or dysregulation of the RAB3D gene have been associated with several diseases. Notably, it has been linked to neurodevelopmental disorders characterized by brain abnormalities, poor growth, and dysmorphic facial features. Additionally, RAB3D has been implicated in beta-thalassemia major, a blood disorder that reduces the production of hemoglobin .

Research and Applications

Human recombinant RAB3D protein is used in various research applications to study its function and role in disease. Recombinant proteins are produced through genetic engineering techniques, where the RAB3D gene is inserted into a host organism, such as E. coli, to produce the protein in large quantities. This recombinant protein can then be purified and used in biochemical assays, structural studies, and therapeutic research .

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.