SLC38A5 transports neutral amino acids (e.g., glutamine, serine, glycine) and is implicated in cancer metabolism, angiogenesis, and metabolic reprogramming. The SLC38A5 antibody enables precise detection of this transporter in diverse experimental models, including cancer cell lines, organoids, and tissue sections.
Facilitates amino acid uptake for cancer cell proliferation and survival .
Regulates retinal angiogenesis via glutamine uptake and VEGFR2 signaling .
Coupled with Na⁺/H⁺ exchange, promoting macropinocytosis in triple-negative breast cancer (TNBC) .
Commercially available antibodies vary in specificity, reactivity, and applications. Below is a comparative analysis of prominent SLC38A5 antibodies.
ab317685: Suitable for flow cytometry due to membrane localization detection .
ab72717: Effective in Western blotting (WB) for protein quantification .
28102-1-AP: Requires antigen retrieval (TE buffer pH 9.0 or citrate buffer pH 6.0) for IHC .
Pancreatic Ductal Adenocarcinoma (PDAC):
Triple-Negative Breast Cancer (TNBC):
Retinal Angiogenesis:
Glutamine Metabolism:
mTORC1 Signaling:
PDAC: SLC38A5 deletion reduced tumor growth in xenograft models, highlighting its potential as a therapeutic target .
TNBC: Amiloride, a Na⁺/H⁺ exchanger inhibitor, blocked SLC38A5-induced macropinocytosis, suggesting a dual therapeutic strategy .
Retinopathy: SLC38A5-deficient mice exhibited delayed vascular development and reduced VEGFR2 signaling, implicating it in pathological angiogenesis .
Wnt Signaling: SLC38A5 transcription is regulated by Wnt/β-catenin, linking genetic mutations (e.g., Lrp5 −/−, Ndp) to vascular defects .
Cross-Reactivity: Polyclonal antibodies (e.g., ab72717) may exhibit non-specific binding in complex samples, necessitating validation .
Post-Translational Modifications: Observed MW discrepancies (e.g., 45 kDa vs. 51 kDa in 28102-1-AP) suggest potential glycosylation or splicing variants requiring further study .
SLC38A5 is a neutral amino acid transporter that shuttles several amino acids across cell membranes, with particular preference for glutamine, glycine, serine, histidine, alanine, cysteine, and asparagine . It has been identified as a key component of the amino acid sensing machinery that links circulating amino acids to the control of pancreatic α cell function and mass . Additionally, SLC38A5 has been shown to be highly enriched in retinal vascular endothelium, where it functions as a metabolic regulator of angiogenesis by controlling amino acid uptake and homeostasis in endothelial cells . Recent research has also identified SLC38A5 as a tumor promoter in pancreatic ductal adenocarcinoma (PDAC), where its deletion leads to significant reduction in amino acid substrates and inactivation of oxidative phosphorylation (OXPHOS) .
SLC38A5 shows tissue-specific expression patterns that are relevant for antibody-based detection studies. The transporter is highly expressed in:
Pancreatic α cells, particularly in a subset of highly proliferative α cells
Retinal vascular endothelium, especially in pathological sprouting neovessels
Brain glial cells, where it mediates transcellular transport of amino acids
Single-cell transcriptome analyses have confirmed SLC38A5 expression primarily in vascular endothelium in both mouse and human retinal tissues, with expression patterns similar to the endothelial marker PECAM1 .
SLC38A5 functions within several important signaling networks:
mTOR pathway: SLC38A5-mediated amino acid transport, particularly glutamine, activates the mTOR pathway. Rapamycin (an mTOR inhibitor) blocks SLC38A5 mRNA expression induced by glucagon receptor antibody treatment, indicating an interconnection between SLC38A5 and mTOR signaling .
Wnt/β-catenin signaling: SLC38A5 transcription is regulated by Wnt/β-catenin signaling. It is significantly downregulated in both Lrp5−/− and Ndpy/− retinas, which are genetic models with Wnt signaling mutations .
VEGF receptor 2 signaling: Inhibition of SLC38A5 in human retinal vascular endothelial cells dampens vascular endothelial growth factor receptor 2 activity .
When designing experiments to detect SLC38A5 using antibodies, researchers should consider:
Tissue-specific localization: In pancreatic tissue, SLC38A5 is confined to the plasma membrane of α cells and does not associate with lysosomes . Proper membrane preparation protocols are essential for preserving SLC38A5 epitopes.
Co-localization studies: For pancreatic tissue, dual immunostaining with glucagon (for α cells) and insulin (for β cells) antibodies helps differentiate cell types, as SLC38A5 is detected in a subset of glucagon-positive cells but not in insulin-positive cells .
Cellular resolution: For retinal tissue, techniques such as laser capture microdissection (LCM) have been used to isolate blood vessels from retinal cross-sections, followed by mRNA expression analysis using RT-qPCR to quantify SLC38A5 expression .
Co-staining markers: In retinal tissues, isolectin B4 is used as a marker of vascular endothelium for co-localization studies with SLC38A5 .
Validation of SLC38A5 antibodies requires multiple approaches:
Genetic controls: Use tissues or cells from SLC38A5 knockout models as negative controls. For example, CRISPR/Cas9-mediated knockout of SLC38A5 has been used to validate antibody specificity .
Knockdown validation: siRNA-mediated silencing of SLC38A5 (si-SLC38A5) in relevant cell lines, such as human retinal microvascular endothelial cells (HRMECs), can provide essential validation for antibody specificity .
Cross-validation with mRNA expression: Correlate protein detection with RT-qPCR quantification of SLC38A5 mRNA levels in the same tissue samples .
Western blot analysis: Verify antibody specificity by confirming the correct molecular weight of detected proteins and the absence of non-specific bands. SLC38A5 protein levels have been successfully measured in retinal tissues using this method .
SLC38A5 plays a critical role in regulating cell proliferation:
In pancreatic α cells: SLC38A5 is required for glucagon receptor inhibition-induced α cell proliferation. Ki67 staining for cell proliferation shows that proliferation is four times greater in SLC38A5-positive than in SLC38A5-negative α cells in mice treated with glucagon receptor antibody .
In pancreatic cancer: SLC38A5 acts as a tumor promoter in PDAC. CRISPR/Cas9-mediated knockout of SLC38A5 demonstrates its tumor-promoting role in both in vitro cell line models and subcutaneous xenograft mouse models .
In retinal vascular endothelium: Genetic deficiency of SLC38A5 in mice substantially delays retinal vascular development and suppresses pathological neovascularization in oxygen-induced retinopathy. Inhibition of SLC38A5 in human retinal vascular endothelial cells impairs endothelial cell proliferation and angiogenic function .
Researchers studying the relationship between SLC38A5 and cell proliferation should employ multiple proliferation markers (such as Ki67, BrdU incorporation, or phospho-histone H3) and correlate these with SLC38A5 expression using dual immunostaining approaches.
Based on published research approaches:
For pancreatic tissue:
Fixation: 4% paraformaldehyde is generally suitable
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0)
Blocking: 5-10% normal serum from the species in which the secondary antibody was raised
Primary antibody incubation: Overnight at 4°C with optimized dilution
Detection: Fluorescently labeled secondary antibodies for co-localization studies with other markers (e.g., glucagon, insulin)
For retinal tissue:
Sample preparation: Consider both whole-mount preparations and cross-sections
Co-staining: Use isolectin B4 as a vascular endothelium marker
Visualization: Confocal microscopy is preferred for detailed co-localization analysis
Functional assessment of SLC38A5 using antibodies can be performed through:
Amino acid uptake assays: Measure glutamine uptake using bioluminescent assays in cells after manipulating SLC38A5 expression (e.g., with siRNA). Inhibition of SLC38A5 in human retinal microvascular endothelial cells resulted in approximately 25% decrease in intracellular glutamine levels .
Proliferation assays in correlation with SLC38A5 expression:
MTT assays for cell proliferation
Ki67 immunostaining for identification of proliferating cells
BrdU incorporation assays
Angiogenesis assays for endothelial cells:
Signal pathway analysis using phospho-specific antibodies:
When investigating regulation of SLC38A5 expression:
Wnt signaling manipulation:
mTOR pathway modulation:
Amino acid availability:
Disease models:
Common challenges and solutions include:
Non-specific staining:
Use appropriate negative controls, including tissues from SLC38A5 knockout animals
Include isotype controls to identify non-specific binding
Optimize antibody concentration through titration experiments
Low signal-to-noise ratio:
For membrane proteins like SLC38A5, gentle fixation and appropriate membrane preparation protocols are critical
Consider tyramide signal amplification for low-abundance targets
Cross-reactivity with other SLC38 family members:
Verify antibody specificity against other family members
Conduct peptide competition assays with the immunizing peptide
Variability in expression across different cell subtypes:
Interpretation guidelines:
Subcellular localization: SLC38A5 is primarily localized to the plasma membrane of cells, consistent with its function as a transmembrane transporter. In α cells, SLC38A5 expression is confined to the plasma membrane and does not associate with lysosomes .
Tissue-specific patterns:
In pancreas: Focus on co-localization with glucagon-positive cells
In retina: Examine co-localization with vascular endothelial markers
Expression dynamics:
Functional correlation:
Integration strategies:
Combined approaches:
Correlate SLC38A5 protein expression (by immunoblotting/immunostaining) with mRNA expression (by RT-qPCR or RNA-seq)
Link SLC38A5 expression with metabolomic analysis of amino acid levels, particularly SLC38A5 substrates like glutamine
Single-cell multi-omics:
Use single-cell RNA sequencing data to identify cell populations expressing SLC38A5
Follow up with antibody-based approaches to validate protein expression in specific cell types
Functional validation:
After identifying SLC38A5-expressing cells by antibody staining, isolate these populations for targeted metabolomic analysis
Measure glutamine uptake and metabolism in SLC38A5-positive versus negative cells
Pathway analysis:
Combine antibody detection of SLC38A5 with assessment of downstream pathways
In PDAC models, SLC38A5 deletion leads to significant reduction in many amino acid substrates and OXPHOS inactivation
Experimental validation demonstrates inhibition of mTORC1, glycolysis, and mitochondrial respiration in SLC38A5 knockout cells
SLC38A5 antibodies show promise for several disease-related applications:
Cancer research:
Retinal vascular diseases:
Metabolic disorders:
Development of combination therapies could include:
Dual targeting strategies:
SLC38A5 inhibition combined with mTOR inhibitors (like rapamycin)
Antibodies can monitor the effectiveness of these combinations on target inhibition
Metabolic intervention monitoring:
Track changes in amino acid metabolism alongside SLC38A5 expression
Combine with glutaminase inhibitors for enhanced metabolic disruption in cancer cells
Angiogenesis inhibition:
Pair SLC38A5 targeting with VEGF pathway inhibitors
Use antibodies to monitor pathway inhibition and vascular responses
Biomarker development: