SLC35D3 (Solute Carrier Family 35 Member D3) functions as a probable UDP-glucose transmembrane transporter involved in UDP-glucose transport from the cytosol to the lumen of synaptic vesicles . This protein plays several critical roles in cellular physiology:
Functions as a molecular adapter enhancing the formation of the PI3KC3-C1/AIC/autophagy initiation complex to promote autophagy in dopaminergic neurons
Regulates plasma membrane localization of the D1A dopamine receptor (DRD1) and dopamine signaling
Promotes white adipose tissue browning to ameliorate metabolic dysfunction
The protein is specifically expressed in striatonigral MSNs (medium spiny neurons) that express D1R rather than striatopallidal MSNs expressing D2R . Physically, SLC35D3 interacts with D1R, with the N-terminal portion of SLC35D3 (1-241aa) interacting with the C-terminal region of D1R (217-446aa) .
Several validated SLC35D3 antibodies are available for research applications:
Antibody | Host/Type | Reactive Species | Applications | Immunogen |
---|---|---|---|---|
Abcam ab211520 | Rabbit Polyclonal | Human | IHC-P, WB, ICC/IF | Synthetic peptide within Human SLC35D3 |
Proteintech 18100-1-AP | Rabbit Polyclonal | Human, mouse, rat | ELISA | SLC35D3 fusion protein Ag12203 |
Boster A15258 | Rabbit Polyclonal | Human, mouse | ELISA, WB, ICC, IF | 17aa synthetic peptide near C-terminus |
These antibodies detect both known isoforms of SLC35D3 . The calculated molecular weight of SLC35D3 is approximately 44 kDa, though the observed molecular weight in Western blots is reported to be around 68 kDa, possibly due to post-translational modifications .
SLC35D3 antibodies have been validated for multiple experimental applications:
Sample preparation: Total protein extraction from striatum or other relevant tissues
Loading: 20-40 μg of total protein per lane is recommended
Detection: Total D1 receptor expression levels can be compared between wild-type and experimental samples using this method
Note: Western blotting confirms that SLC35D3 does not affect total D1R expression levels, only its cellular distribution
Sample preparation: Formalin-fixed, paraffin-embedded tissues
Antigen retrieval: Heat-mediated antigen retrieval is recommended
Working concentration: 5 μg/ml has been validated for human small intestine tissue
Visualization: DAB or fluorescent secondary antibody systems
Cell fixation: 4% paraformaldehyde is standard
Working concentration: 2.5 μg/ml has been validated in HeLa cells
Co-localization studies: Can be combined with organelle markers to determine subcellular localization (ER vs. endosomes)
SLC35D3 antibodies can be used in conjunction with D1R antibodies to investigate dopamine receptor trafficking mechanisms:
Co-immunoprecipitation: Use anti-SLC35D3 antibodies to precipitate protein complexes and detect D1R by Western blot, or vice versa, to confirm physical interaction between SLC35D3 and D1R
Immunofluorescence co-localization:
Double-stain cells/tissues with SLC35D3 and D1R antibodies
Assess co-localization using confocal microscopy
Quantify co-localization using appropriate software
Subcellular fractionation and Western blotting:
Immuno-electron microscopy:
Research into SLC35D3's role in metabolic syndrome can be approached through several experimental strategies:
Animal models:
Use of ros mutant mice (harboring SLC35D3 mutation) which display obesity and metabolic syndrome
Generation of adipocyte-specific SLC35D3 knockout mice using the adiponectin Cre-lox system (SAKO mice)
Monitoring metabolic parameters: BMI, waist circumference, blood pressure, triglycerides, cholesterol, and glucose levels
Pharmacological intervention studies:
Human mutation screening:
Adipose tissue analysis:
The molecular interaction between SLC35D3 and D1R can be investigated using several sophisticated techniques:
Domain mapping through co-immunoprecipitation:
Subcellular localization studies:
Live-cell imaging of trafficking dynamics:
Generate fluorescently tagged SLC35D3 and D1R constructs
Monitor trafficking in real-time using live-cell microscopy
Compare trafficking kinetics between wild-type and mutant proteins
Functional assays of dopamine signaling:
Several technical issues may arise when working with SLC35D3 antibodies:
Discrepancy between predicted and observed molecular weight:
Antibody storage and stability:
Cross-reactivity considerations:
Species-specific considerations:
Ensure the selected antibody is validated for your species of interest
Consider sequence homology when working with less common model organisms
When investigating SLC35D3 mutations and their functional impacts, researchers should consider:
Expression systems for mutant proteins:
Generate expression constructs containing identified mutations (e.g., ΔK404, insL201)
Use epitope tags to facilitate detection if antibody recognition might be affected by mutations
Consider both transient transfection and stable cell line generation
Subcellular localization analysis:
Functional assays:
D1R trafficking efficiency (plasma membrane vs. intracellular ratio)
D1R-mediated signaling (cAMP production, downstream effector activation)
UDP-glucose transport activity
Autophagy induction capacity
CRISPR-Cas9 genome editing:
Generate cellular or animal models carrying specific SLC35D3 mutations
Compare the effects of homozygous vs. heterozygous mutations
Assess tissue-specific phenotypes (adipose tissue, striatum)
Recent research has identified SLC35D3's role in adipose tissue biology :
Expression analysis approaches:
Adipocyte-specific knockout models:
White adipose tissue browning studies:
Analyze markers of brown/beige adipocytes in SLC35D3-deficient vs. control adipose tissue
Measure mitochondrial content and activity
Assess thermogenic capacity and energy expenditure
Mechanistic investigations:
Explore the link between SLC35D3's role in dopamine signaling and adipose tissue biology
Investigate potential interactions with adipogenesis regulators and thermogenic pathways
Based on current understanding of SLC35D3 function, several therapeutic approaches could be explored:
D1R agonist therapy:
SLC35D3 gene therapy approaches:
Delivery of functional SLC35D3 to relevant tissues (striatum, adipose tissue)
Development of mutation-specific approaches for patients with identified SLC35D3 mutations
Small molecule modifiers of SLC35D3 function:
Compounds that enhance remaining SLC35D3 activity in heterozygous mutation carriers
Molecules that promote D1R trafficking through alternative pathways
Adipose tissue browning inducers:
Targeting the SLC35D3-mediated pathway for white adipose tissue browning
Combination approaches targeting both central (striatal) and peripheral (adipose) SLC35D3 functions