SLC17A4 antibodies target the SLC17A4 protein, a Na⁺-phosphate cotransporter homologue and organic anion transporter expressed in the liver, kidney, gastrointestinal tract, and pancreas . These antibodies enable the detection and functional characterization of SLC17A4 in various experimental contexts, including:
SLC17A4 facilitates cellular uptake of T3 and T4 with high affinity (IC₅₀: 0.35 µM for T3, 0.06 µM for T4), surpassing the efficiency of other transporters like MCT8 . Functional studies in COS-1 cells demonstrated:
4-fold increase in T3/T4 uptake with SLC17A4 overexpression .
Na⁺-dependent transport kinetics, confirmed via efflux assays .
Inhibition by diisothiocyanostilbene-2,2′-disulfonic acid (DIDS) and Evans blue .
In prostate cancer (PCa), SLC17A4 upregulation correlates with poor prognosis and aggressive phenotypes :
Knockdown of SLC17A4 via siRNA reduced PCa cell proliferation (DU145, PC3) by 40–60% and inhibited invasion .
High SLC17A4 expression associates with dysregulated immune cell infiltration (e.g., B cells, CD4⁺ T cells) .
SLC17A4 exports urate and p-aminohippuric acid (PAH) in the intestines, contributing to urate homeostasis :
Specificity: Antibodies like AV43906 and HPA055564 are validated for IHC and WB in human, mouse, and rat tissues .
Glycosylation: SLC17A4 function is unaffected by N-linked glycosylation inhibitors (e.g., tunicamycin) .
Epitopes: Prestige Antibodies® target sequences like YDDPVNHPFISAGEKRYIVCSLAQQDCSPGWSLPIRA .
SLC17A4 is a potential biomarker for:
SLC17A4 (solute carrier family 17 member 4) is a membrane transport protein originally classified as a Na+/phosphate cotransporter homologue. It is predominantly expressed in the gastrointestinal tract, particularly in the intestinal brush border membrane, as well as in the liver and kidneys . Immunohistochemical studies using specific antibodies have confirmed its localization to these tissues, with particularly strong expression in the intestinal epithelial cells . SLC17A4 belongs to the SLC17 anion transporter family, which comprises nine members that transport various organic anions in membrane potential (Δψ)- and Cl−-dependent manners .
SLC17A4 facilitates the bidirectional transport of thyroid hormones across cellular membranes. In vitro studies have demonstrated that SLC17A4 induces the cellular uptake of T3 and T4 by approximately 4 times over control cells, and of reverse (r)T3 by 1.5 times . Unlike some other thyroid hormone transporters, SLC17A4-mediated transport is Na+ and Cl− independent, and is uniquely stimulated by low extracellular pH . The protein also facilitates the efflux of T3 and T4, and to a lesser extent of 3,3′-diiodothyronine (T2), indicating its role in both the import and export of thyroid hormones . The estimated IC50 values for T3 (0.35 ± 0.13 μM) and T4 (0.06 ± 0.01 μM) transport by SLC17A4 are considerably lower than those of other known thyroid hormone transporters, suggesting it has remarkably high substrate affinity .
Beyond thyroid hormone transport, SLC17A4 has been characterized as an intestinal organic anion exporter. Studies with proteoliposomes containing purified SLC17A4 protein demonstrated uptake of radiolabeled p-aminohippuric acid (PAH) in a Cl−-dependent manner at the expense of an electrochemical gradient of protons, especially membrane potential (Δψ), across the membrane . Additionally, SLC17A4 has been identified as a Na/PO4 cotransporter that may play a role in the regulation of lithium transport and its therapeutic effects . This multifunctionality suggests SLC17A4 may have diverse physiological roles depending on tissue context and substrate availability.
For detecting SLC17A4 protein expression in tissue samples, immunohistochemistry (IHC) and Western blotting using specific anti-SLC17A4 antibodies have proven effective. When selecting antibodies, researchers should consider those validated in multiple experiments and cited in publications . For immunoblotting studies, cellular lysates should be prepared carefully, as SLC17A4 is subject to N-linked glycosylation . Treatment with tunicamycin, a pharmacological inhibitor of N-linked glycosylation, can help distinguish between glycosylated and non-glycosylated forms of the protein . For IHC applications, optimize fixation protocols to preserve membrane proteins, and include appropriate controls to validate antibody specificity, particularly in tissues with known expression patterns such as intestinal brush border membranes .
Validating SLC17A4 antibody specificity requires multiple complementary approaches:
Overexpression systems: Compare antibody staining in cells transiently transfected with SLC17A4 expression vectors versus empty vector controls .
Competing peptide assays: Pre-incubate the antibody with the immunizing peptide to confirm signal elimination in immunoblots or IHC.
Knockout/knockdown controls: If available, use SLC17A4 knockout tissues or cells with siRNA-mediated knockdown to confirm absence of signal.
Glycosylation analysis: Since SLC17A4 undergoes N-linked glycosylation, treatment of samples with tunicamycin or PNGase F can confirm specificity by demonstrating the expected mobility shift .
Multi-antibody validation: Use multiple antibodies targeting different epitopes of SLC17A4 to confirm consistent localization patterns.
Cross-reactivity testing: Test the antibody against closely related family members (other SLC17 transporters) to ensure specificity within the protein family.
For successful immunoprecipitation (IP) of SLC17A4, consider the following protocol:
Lysis buffer optimization: Use a buffer containing 1% NP-40 or Triton X-100, 150 mM NaCl, 50 mM Tris-HCl (pH 7.5), and protease inhibitor cocktail. For membrane proteins like SLC17A4, addition of 0.1% SDS may help solubilization while maintaining antibody recognition.
Pre-clearing: Pre-clear lysates with protein A/G beads to reduce non-specific binding.
Antibody selection: Choose antibodies validated for IP applications and raised against epitopes less likely to be affected by post-translational modifications like N-glycosylation (avoid antibodies targeting the Asn66, Asn75, and Asn90 residues identified as glycosylation sites) .
Incubation conditions: Incubate lysates with antibodies overnight at 4°C with gentle rotation to maximize antigen-antibody interaction while minimizing degradation.
Washing stringency: Use a progressive washing strategy with decreasing salt concentrations to maintain specific interactions while removing non-specific binding.
Elution considerations: Since SLC17A4 is a membrane protein, consider gentle elution methods such as peptide competition or non-denaturing buffers to preserve protein integrity.
Based on published research, several cellular models have proven effective for studying SLC17A4 transport function:
COS-1 cells: These cells have been successfully used for transient overexpression of SLC17A4 to study thyroid hormone transport capacity . They provide a clean background with minimal endogenous thyroid hormone transport activity.
Proteoliposome systems: For highly controlled transport studies, purified SLC17A4 protein can be reconstituted into proteoliposomes, allowing precise characterization of transport kinetics and substrate specificity in the absence of other cellular transporters .
Intestinal epithelial cell lines: Given SLC17A4's natural expression in the intestine, cell lines like Caco-2 or T84 may provide physiologically relevant models for studying endogenous transport function once differentiated.
Transfected HEK293 cells: These cells provide a robust mammalian expression system with low background expression of thyroid hormone transporters and are suitable for creating stable cell lines for long-term studies.
When designing experiments, co-transfection with intracellular thyroid hormone-binding proteins such as mu-crystallin (CRYM) can enhance detection of transport activity by retaining transported hormones inside the cell .
Accurate measurement of SLC17A4-mediated thyroid hormone transport requires carefully designed assays:
Radiolabeled substrate uptake: Use [125I]-labeled T3 or T4 to quantify cellular uptake in SLC17A4-expressing versus control cells . Typical assay conditions include:
Incubation of cells with 1 nM [125I]-T3 or [125I]-T4 in serum-free medium
Time course experiments ranging from 2-60 minutes to determine linear uptake phase
Termination of transport by rapid washing with ice-cold buffer
Cell lysis and scintillation counting to quantify internalized hormone
Efflux studies: To measure SLC17A4-mediated efflux, preload cells with radiolabeled hormones, then measure release into hormone-free medium over time .
pH dependence analysis: Given SLC17A4's sensitivity to extracellular pH, transport assays should be conducted at multiple pH values (typically pH 5.5-8.0) to characterize this regulatory mechanism .
Inhibition studies: Assess transport specificity by competition with unlabeled hormones or potential inhibitors. In particular, test various iodothyronines and metabolites, especially those containing at least three iodine moieties .
Co-expression with CRYM: To enhance sensitivity of uptake assays, co-express the intracellular thyroid hormone-binding protein mu-crystallin, which traps transported hormones inside the cell .
When investigating SLC17A4 glycosylation, several critical controls must be included:
Positive glycoprotein control: Include a well-characterized glycoprotein such as transferrin in parallel experiments to validate glycosylation detection methods.
Enzymatic deglycosylation controls:
PNGase F treatment (removes all N-linked glycans)
Endoglycosidase H treatment (removes only high-mannose and some hybrid glycans)
O-glycosidase treatment (to confirm N- vs. O-linked glycosylation)
Pharmacological inhibition: Include tunicamycin-treated samples (a pharmacological inhibitor of N-linked glycosylation) at various concentrations to demonstrate dose-dependent inhibition of glycosylation .
Site-directed mutagenesis controls: Include wild-type SLC17A4 alongside mutants in which the identified glycosylation sites (Asn66, Asn75, and Asn90) have been individually or collectively mutated to glutamine to prevent glycosylation .
Subcellular localization assessment: Monitor protein trafficking using fluorescently tagged constructs or immunofluorescence to determine how glycosylation affects membrane localization.
Functional transport assays: Compare transport activity between glycosylated and deglycosylated forms to assess the functional impact of these modifications.
SLC17A4 exhibits distinctive properties compared to other known thyroid hormone transporters:
| Transporter | T3 IC50 (μM) | T4 IC50 (μM) | Na+ dependence | pH sensitivity | Primary expression sites |
|---|---|---|---|---|---|
| SLC17A4 | 0.35 ± 0.13 | 0.06 ± 0.01 | Independent | Enhanced at low pH | Intestine, liver, kidney |
| MCT8 | 20.61 ± 1.26 | 23.22 ± 1.22 | Independent | Minimal | Brain, liver, kidney |
| OATP1C1 | ~0.9 | ~0.1 | Independent | Minimal | Brain, testis |
| LAT1 | ~8.0 | ~7.9 | Independent | Minimal | Blood-brain barrier |
| LAT2 | ~0.8 | ~7.1 | Independent | Minimal | Kidney, placenta |
The IC50 values for SLC17A4-mediated transport of T3 (0.35 ± 0.13 μM) and T4 (0.06 ± 0.01 μM) are considerably lower than those of MCT8 and many other transporters, indicating exceptionally high substrate affinity . Additionally, SLC17A4 is unique among thyroid hormone transporters in its stimulation by low extracellular pH .
Unlike most other thyroid hormone transporters, SLC17A4 transport activity is not inhibited by classical thyroid hormone transporter inhibitors, suggesting a distinct substrate binding site or transport mechanism . This combination of high affinity and unique regulatory properties positions SLC17A4 as a specialized transporter that may be particularly important in tissues with acidic microenvironments, such as the intestinal lumen.
Studying SLC17A4's role in thyroid hormone-dependent physiological processes requires multiple complementary approaches:
Animal models:
Conditional knockout mice with tissue-specific deletion of SLC17A4 (particularly in intestine, liver, and kidney)
Humanized mouse models expressing human SLC17A4 variants
Measurement of thyroid hormone levels in tissue compartments versus circulation
Ex vivo tissue studies:
Intestinal organoids from normal and SLC17A4-deficient tissues
Precision-cut liver or kidney slices to study transport in complex tissue architectures
Ussing chamber experiments with intestinal tissue to measure directional transport
Systems biology approaches:
Integration of genomic, transcriptomic, and metabolomic data from thyroid hormone target tissues
Network analysis to identify SLC17A4-dependent pathways
Multi-tissue modeling of thyroid hormone distribution
Clinical translational studies:
Correlation of SLC17A4 genetic variants with thyroid function parameters in patient cohorts
Assessment of SLC17A4 expression in thyroid disease tissue samples
Pharmacogenomic studies to determine if SLC17A4 variants affect response to thyroid hormone replacement therapy
Drug development strategies:
High-throughput screening for selective SLC17A4 modulators
Structure-based drug design targeting SLC17A4 transport mechanism
Development of tissue-specific thyroid hormone delivery systems exploiting SLC17A4 expression patterns
Researchers may encounter several technical challenges when working with SLC17A4 antibodies:
Non-specific binding: SLC17A4 shares sequence homology with other SLC17 family members, potentially leading to cross-reactivity.
Solution: Validate antibody specificity using overexpression systems, knockout controls, and peptide competition assays.
Use multiple antibodies targeting different epitopes to confirm results.
Variable glycosylation patterns: As SLC17A4 undergoes N-linked glycosylation at multiple sites (Asn66, Asn75, and Asn90) , this can create heterogeneous banding patterns in western blots.
Solution: Include PNGase F-treated samples to collapse glycoform diversity.
Consider using antibodies targeting non-glycosylated regions of the protein.
Low signal-to-noise ratio in tissues with moderate expression:
Solution: Implement signal amplification methods such as tyramide signal amplification for IHC.
Enrich membrane fractions before immunoblotting to concentrate the target protein.
Membrane protein solubilization issues:
Solution: Optimize lysis buffers with different detergents (CHAPS, DDM, or digitonin may better preserve protein conformation than SDS).
Avoid excessive heating of samples before SDS-PAGE, as this can cause membrane protein aggregation.
Epitope masking due to protein-protein interactions:
Solution: Test multiple fixation and antigen retrieval methods for IHC.
Consider native vs. denaturing conditions for immunoprecipitation.
Designing experiments to isolate SLC17A4's specific contribution to thyroid hormone transport requires:
Selective inhibition strategies:
Expression system selection:
Choose cell models with minimal expression of other thyroid hormone transporters.
Systematically knock down known transporters to isolate SLC17A4 activity.
Exploiting unique properties:
Substrate specificity profiling:
Compare transport of various iodothyronines and metabolites between SLC17A4 and other transporters.
Identify unique substrates or substrate preferences for SLC17A4.
Kinetic analysis:
Tissue-specific contributions:
Focus on intestinal models where SLC17A4 is highly expressed and other thyroid hormone transporters may be less abundant.
Compare transport in polarized cell models to assess directional preferences.
When interpreting SLC17A4 expression data across tissues, researchers should consider:
Tissue preparation methods:
Membrane protein extraction efficiency varies between tissues.
Fixation methods can differentially affect epitope accessibility.
Cell-type specific expression:
Post-translational modifications:
Glycosylation patterns may vary between tissues, affecting antibody recognition.
Phosphorylation or other modifications may influence detection.
Splice variants:
Check if tissue-specific splice variants exist that may not be detected by all antibodies.
Design primers or use antibodies that can distinguish between variants.
Protein vs. mRNA correlation:
Do not assume protein levels directly correlate with mRNA expression.
Post-transcriptional regulation may vary between tissues.
Physiological state:
Consider how thyroid status, nutritional state, or disease may affect expression.
Control for relevant physiological variables when comparing across samples.
Species differences:
Human SLC17A4 expression patterns may differ from model organisms.
Validate antibody cross-reactivity when studying non-human tissues.
Emerging evidence suggests SLC17A4 may play important roles in thyroid-related pathologies:
Thyroid dysfunction: Genome-wide association studies have identified SLC17A4 genetic variants associated with thyroid function parameters, including circulating free T4 levels and the T3/T4 ratio . This suggests SLC17A4 variants may contribute to risk for both overt and subclinical thyroid disease.
Intestinal thyroid hormone absorption: Given SLC17A4's expression in intestinal brush border membranes and its ability to transport thyroid hormones, it may affect the bioavailability of orally administered thyroid hormone replacement therapy . Genetic variants could potentially explain inter-individual variability in treatment response.
Thyroid hormone metabolism: By influencing the tissue distribution of thyroid hormones, especially in the liver and kidneys where metabolizing enzymes are abundant, SLC17A4 may indirectly affect systemic thyroid hormone metabolism and clearance .
Autoimmune thyroid disease: Some genetic variants identified in genome-wide studies were associated with thyroid peroxidase antibody (TPOAb) positivity or Graves' disease, suggesting potential links between SLC17A4 and autoimmune thyroid pathologies .
Future research should investigate these connections through clinical studies correlating SLC17A4 variants with treatment outcomes in hypothyroidism, functional studies of variant effects on transport activity, and targeted interventions to modulate SLC17A4 function in relevant disease models.
Research on SLC17A4 opens several promising avenues for therapeutic development:
Targeted drug delivery: SLC17A4's high expression in intestinal brush border membranes makes it a potential target for enhancing oral bioavailability of thyroid hormone mimetics or other drugs. Prodrugs designed as SLC17A4 substrates could improve intestinal absorption .
Precision medicine approaches: Genetic screening for SLC17A4 variants could identify patients who might require adjusted dosing of thyroid hormone replacement therapy based on their transporter efficiency .
Tissue-selective thyroid hormone action: Given SLC17A4's tissue-specific expression pattern, compounds that modulate its transport activity could potentially direct thyroid hormone action to specific tissues while sparing others, reducing side effects.
Novel treatment strategies for thyroid disorders: Understanding how SLC17A4 contributes to thyroid hormone homeostasis may reveal new therapeutic targets for conditions like thyroid hormone resistance or thyrotoxicosis.
Biomarker development: SLC17A4 expression levels or genetic variants could potentially serve as biomarkers for predicting thyroid disease risk or treatment response.
Research approaches should include high-throughput screening for selective SLC17A4 modulators, structure-function studies to guide rational drug design, clinical studies correlating genetic variants with treatment outcomes, and development of tissue-specific delivery systems leveraging SLC17A4 expression patterns.
Several technological advances would significantly enhance our understanding of SLC17A4:
Structural biology approaches:
Cryo-electron microscopy to determine the three-dimensional structure of SLC17A4, particularly in different conformational states during the transport cycle
X-ray crystallography of SLC17A4 in complex with substrates to elucidate binding sites
Hydrogen-deuterium exchange mass spectrometry to map dynamic regions involved in transport
Advanced imaging techniques:
Super-resolution microscopy to visualize SLC17A4 distribution and dynamics in native membranes
Live-cell imaging with fluorescent thyroid hormone analogs to track transport in real-time
Correlative light and electron microscopy to connect function with ultrastructural localization
Computational approaches:
Molecular dynamics simulations to model transport mechanisms and substrate interactions
Machine learning algorithms to predict structure-function relationships and identify potential inhibitors
Systems biology models integrating SLC17A4 into thyroid hormone homeostasis networks
Genetic engineering tools:
CRISPR-based approaches for precise genome editing to study SLC17A4 variants in isogenic cell lines
Inducible and tissue-specific knockout models to assess physiological roles
Base editing to introduce specific mutations without disrupting the entire gene
Single-molecule techniques:
Patch-clamp fluorometry to correlate structural dynamics with transport function
Single-molecule FRET to monitor conformational changes during substrate binding and transport
Nanobody-based probes to track specific conformational states
These technological advances would provide unprecedented insights into how SLC17A4 functions at the molecular level, enabling more targeted therapeutic approaches and a deeper understanding of its physiological roles.