SLC12A3 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Key Features

ParameterDetails
HostRabbit polyclonal
ConjugateFITC (Fluorescein Isothiocyanate)
ReactivityHuman, Mouse, Rat (predicted)
ApplicationsImmunofluorescence (IF), Immunohistochemistry (IHC), Flow Cytometry (FCM)
Dilution RecommendationsIF/IHC: 1:50–200; FCM: 1:20–100
Storage-20°C in aliquots to prevent freeze-thaw cycles

Fluorescence-Based Assays

  • Immunofluorescence (IF): Used to localize SLC12A3 in renal tissues, such as the distal convoluted tubules, where the cotransporter mediates sodium and chloride reabsorption .

  • Flow Cytometry (FCM): Quantifies SLC12A3 expression in cell suspensions, aiding studies on kidney physiology or pathologies like Gitelman syndrome .

Immunohistochemistry (IHC)

  • Tissue Staining: Detects SLC12A3 in paraffin-embedded kidney sections (e.g., rat, mouse, human), with protocols involving antigen retrieval and counterstaining with hematoxylin .

Technical Considerations and Optimization

  • Antigen Retrieval: Tris-EDTA buffer (pH 9.0) is recommended for IHC to enhance epitope accessibility .

  • Cross-Reactivity: Ensure antibodies do not cross-react with homologs (e.g., SLC12A1/A2) to avoid false positives .

  • Storage Stability: Aliquot and store at -20°C to preserve fluorescence signal integrity .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery timeframes.
Synonyms
FLJ96318 antibody; Na Cl cotransporter antibody; Na Cl symporter antibody; Na-Cl symporter antibody; NaCl electroneutral thiazide sensitive cotransporter antibody; NCC antibody; NCCT antibody; S12A3_HUMAN antibody; slc12a3 antibody; Solute carrier family 12 (sodium/chloride transporters) member 3 antibody; Solute carrier family 12 member 3 antibody; Thiazide sensitive Na Cl cotransporter antibody; Thiazide sensitive sodium chloride cotransporter antibody; Thiazide-sensitive sodium-chloride cotransporter antibody; TSC antibody
Target Names
SLC12A3
Uniprot No.

Target Background

Function
SLC12A3 encodes an electroneutral sodium and chloride ion cotransporter. In the kidney's distal convoluted tubules, this protein plays a critical role in sodium and chloride reabsorption. Furthermore, SLC12A3 acts as a receptor for the proinflammatory cytokine IL18, contributing to IL18-induced cytokine production, including IFNG, IL6, IL18, and CCL2. SLC12A3 can function either independently of IL18R1 or in a complex with IL18R1.
Gene References Into Functions
  1. This research identified two novel pathogenic variants of SLC12A3 and their contribution to Gitelman syndrome. PMID: 29378538
  2. The findings revealed a strong association between SLC12A3 polymorphisms and LDL-C levels. PMID: 29661184
  3. The SLC12A3-Arg913Gln variation may be linked to increased blood pressure and urinary albumin excretion rate (UAER), potentially serving as a predictive factor for the development and progression of end-stage renal disease in Chinese type 2 diabetes patients undergoing hemodialysis. PMID: 28744814
  4. Mutations in both Gitelman syndrome pedigrees were determined to be compound heterozygous mutations in SLC12A3, with a majority identified as missense mutations. PMID: 26825084
  5. Allelic and genotypic frequencies of single nucleotide polymorphism rs11643718 of solute carrier family 12 (sodium-chloride transporters), member 3 protein (SLC12A3) gene are associated with the onset of disease hypertension. PMID: 29419876
  6. A novel recessive mutation in KLHL3 (S553L) was identified in familial hyperkalemia and hypertension. Increased urinary NCC was observed in affected members (heterozygous) with dominant KLHL3 Q309R, and in affected members (homozygous) of the recessive form. PMID: 28511177
  7. Case Report: SLC12A3 gene heterozygous mutation causing Gitelman syndrome in a primary Sjogren syndrome patient. PMID: 28685938
  8. These findings have implications for providing appropriate genetic counseling to families regarding the risk associated with inbreeding and the identification of carrier/presymptomatic relatives. Additionally, it expands the known spectrum of genotypic and phenotypic characteristics of Gitelman syndrome. PMID: 28446151
  9. Two novel heterozygous mutations: c.35_36insA and c.1095+5G>A were found in transcript NM_000339.2 in the SLC12A3 gene in a patient with Gitelman syndrome. The first mutation was also found in the patient's mother, and the second in the father. Only one of the two mutations identified in the patient (c.35_36insA) was found in his sister. PMID: 26306968
  10. Sixteen novel SLC12A3 pathogenic mutations were identified in a cohort of Chinese patients with Gitelman syndrome. PMID: 27454426
  11. Two novel mutations, a S546G substitution in exon 13, and insertion of AGCCCC at c.1930 in exon 16, were found to cause Gitelman syndrome in a South African family. PMID: 28125972
  12. Report novel SLC12A3 mutations in Chinese patients with Gitelman syndrome. PMID: 27216017
  13. A novel SLC12A3 mutation was identified in a Chinese GS pedigree, resulting in the substitution of leucine by proline at codon 700 of the NCCT transporter. The proband and his elder sister had a homozygous mutation, while his mother and daughter carried one mutated allele. As only the proband exhibited bilateral lower limb weakness, it was inferred that the same genotype could manifest as diverse phenotypes. PMID: 27783806
  14. MDCKI cells can be employed to assess the activity, cellular localization, and abundance of wild-type or mutant NCC. PMID: 28515174
  15. In wild-type mice, total (tNCC) and phosphorylated (pNCC) NCC protein expressions were 1.8- and 4.6-fold higher in females compared to males, consistent with the larger response to hydrochlorothiazide. In AT1a receptor knockout mice, tNCC and pNCC increased significantly in males to levels not different from those in females. PMID: 28566500
  16. NCC1/2, NCC1-3, and pNCC1-3-T55/T60 are upregulated by hydrochlorothiazide, and the increase in NCC abundance in urinary extracellular vesicles of essential hypertensive patients correlates with the blood pressure response to hydrochlorothiazide. PMID: 28274929
  17. Data indicates that WNK lysine deficient protein kinase 3 protein (WNK3) interacts with NCC and increases the Na-Cl cotransporter (NCC) expression on the cell membrane and in the cytoplasm. PMID: 27378340
  18. Variants of the SLC12A3 gene confer susceptibility to abnormal serum LDL-c levels in the Mongolian population. PMID: 28166833
  19. A significant association of the SLC12A3 rs11643718 and ELMO1 rs741301 (Single nucleotide Polymorphism) SNPs with diabetic nephropathy in south Indians. PMID: 27699784
  20. This paper identified a novel SLC12A3 allele in Gitelman syndrome that activates a cryptic exon flanked by interspersed repeats deep in intron 12. PMID: 27784896
  21. SLC12A3 gene homozygous mutation is associated with Gitelman syndrome. PMID: 26260218
  22. SNPs in the SLC12A3 gene confer susceptibility to hypertension in the Mongolian population. PMID: 26751802
  23. Compared with patients with 1 mutant SLC12A3 allele, patients with 2 mutant SLC12A3 alleles had more severe hypomagnesemia, but did not have more severe hypokalemia. PMID: 26770037
  24. The results of this study support that the SLC12A3 gene is a susceptibility gene for hypertension in the Mongolian population. PMID: 26345939
  25. Suggest NCC1/2 is a fully functional thiazide-sensitive NaCl-transporting protein in the kidney. PMID: 26561651
  26. Low SLC12A3 urine levels were associated with Gitelman syndrome. PMID: 25422309
  27. Association between thiazide-sensitive Na(+)-Cl(-) cotransporter mutants and human hypertension as well as Gitelman's syndrome (review). PMID: 25841442
  28. Data from two families in China suggest two homozygous mutations in SLC12A3 (Arg928Cys or Ser710X) can be associated with Gitelman syndrome; members of two families exhibit additional mutations and heterogenicity of clinical phenotype. [CASE STUDY] PMID: 25273610
  29. SLC12A3 Arg913Gln polymorphism was associated with Type 2 diabetes and diabetic nephropathy in the Malaysian cohort. The meta-analysis confirmed the protective effects of SLC12A3 913Gln allele in diabetic nephropathy. PMID: 25401745
  30. The SLC12A3 34372 AA genotype is associated with a reduced risk of diabetes. PMID: 24433479
  31. Identification of candidate mutations in the SLC12A3 gene that may induce exon skipping. PMID: 25060058
  32. Findings demonstrate a predominant role played by the SLC12A3 gene rs5804 in determining hypertension risk among northeastern Han Chinese PMID: 24430698
  33. These results suggest that WNK4 attenuates plasma membrane targeting of NCC proteins through regulation of syntaxin 13 SNARE complex formation with VAMP2 in recycling and sorting endosome. PMID: 23993962
  34. Mutation in the SLC12A3 gene is associated with Gitelman syndrome and glomerular proteinuria. PMID: 25165177
  35. Association with SNP rs999662 indicates a potential role for the region containing the solute carrier family 12 member 3 (SLC12A3) gene in transcranial Doppler vasospasm following sub-arachnoid hemorrhage. PMID: 22568564
  36. A model where NCC is constitutively cycled to the plasma membrane, and upon stimulation, it can be phosphorylated to both increase NCC activity and decrease NCC endocytosis, together increasing NaCl transport in the DCT. PMID: 24668812
  37. Highly increased excretion of total urinary NCC and phosphorylated NCC is observed in type II pseudohypoaldosteronism patients. PMID: 24026181
  38. Total urine NCC excretion is diminished in a cohort of Gitelman's syndrome patients with homozygous NCC mutations. PMID: 23833262
  39. Hsp70 and Hsp90 comprise two functionally distinct ER quality control checkpoints that sequentially monitor NCC biogenesis. PMID: 23482560
  40. The TSC gene Arg904Gln polymorphism is not associated with essential hypertension risk. PMID: 23079845
  41. Analysis of SNP databases of Japanese patients with diabetic nephropathy revealed SLC12A3 as a gene related to the above-cited diabetic complication. PMID: 23156397
  42. The NCC mutation p.Thr60Met carriers in Han populations have markedly lower blood pressure and slightly higher fasting plasma glucose compared with normal controls. PMID: 22627394
  43. Forty different SLC12A3 mutations were identified PMID: 22679066
  44. The study shows the identification of 38 novel mutations in the SLC12A3 gene and provides insight into the mechanisms that regulate the thiazide-sensitive NaCl cotransporter. PMID: 22009145
  45. Mutation leads to a failure of the thiazide-sensitive sodium-2-chloride-cotransporter, the so called Gitelman syndrome, which presents similar to a chronic thiazide therapy PMID: 21161146
  46. NCC exhibits distinct ERAD requirements, which correlate with its transmembrane topology and distinguish it from other clients PMID: 22027832
  47. Data show that K1169E lost its inhibitory effect on NCC surface expression compared to wild-type WNK4 when expressed in HEK293 cells, while it did not change NCC total protein expression. PMID: 21196779
  48. Findings suggest that rs7204044 of TSC is a genetic factor for essential hypertension (EH) in Mongolian and Han populations and that rs13306673 is a genetic factor for EH in the Han population PMID: 21644207
  49. There was no significant association between the SLC12A3 R904Q variant and the ClC-Kb-T481S variant and essential hypertension in Mongolian and Han populations in Inner Mongolia PMID: 21644212
  50. In adult patients referred for renal hypokalaemia, we confirmed the presence of mutations of the SLC12A3 gene in 80% of cases PMID: 21753071

Show More

Hide All

Database Links

HGNC: 10912

OMIM: 263800

KEGG: hsa:6559

STRING: 9606.ENSP00000402152

UniGene: Hs.669115

Involvement In Disease
Gitelman syndrome (GTLMNS)
Protein Families
SLC12A transporter family
Subcellular Location
Cell membrane; Multi-pass membrane protein. Apical cell membrane; Multi-pass membrane protein.
Tissue Specificity
Predominantly expressed in the kidney (at protein level). Localizes to the distal convoluted tubules (at protein level)(PubMed:29993276). Not detected in normal aorta, but abundantly expressed in fatty streaks and advanced atherosclerotic lesions (at prot

Q&A

What is SLC12A3 and why is it significant for kidney research?

SLC12A3 (Solute Carrier Family 12 Member 3) encodes a renal thiazide-sensitive sodium-chloride cotransporter that mediates sodium and chloride reabsorption in kidney distal convoluted tubules (DCT) . This protein is critical for electrolyte homeostasis, particularly salt reabsorption. Its significance extends beyond basic physiology as SLC12A3 dysfunction is directly linked to Gitelman syndrome, a condition characterized by hypokalemia, metabolic alkalosis, hypomagnesemia, and hypocalciuria . The protein's structure includes 12 transmembrane domains that facilitate ion transport across the cell membrane, making it an important target for understanding renal function and pathology .

What cellular localization pattern should be expected when using SLC12A3 antibodies?

When using SLC12A3 antibodies in immunolocalization studies, researchers should expect predominant staining in the distal convoluted tubule (DCT) epithelial cells of the kidney . Specifically, SLC12A3 is localized to the apical membrane of these cells, with some cytoplasmic signals depending on trafficking status. The protein contains cytoplasmic topological domains and helical transmembrane regions flanked by a short amino-terminal domain and a longer carboxyl-terminal domain . Proper immunostaining should reveal this distribution pattern with clear membrane localization in DCT segments while showing minimal background in other nephron segments such as proximal tubules or collecting ducts.

What are the optimal fixation and permeabilization methods for SLC12A3 detection in kidney tissue sections?

For optimal detection of SLC12A3 in kidney tissue sections, a balanced approach to fixation and permeabilization is critical due to the protein's membrane localization. Paraformaldehyde (4%) fixation for 10-15 minutes preserves tissue architecture while maintaining antigen accessibility. For frozen sections, acetone fixation (10 minutes at -20°C) often yields superior results by preserving epitope integrity. The permeabilization step requires careful optimization: excessive permeabilization with detergents (>0.2% Triton X-100) can disrupt membrane protein organization, potentially causing artifactual distribution patterns of SLC12A3. A gentler approach using 0.1% Triton X-100 or 0.05% saponin for 5-10 minutes typically provides adequate membrane permeabilization while preserving the native localization pattern. For challenging samples, antigen retrieval using citrate buffer (pH 6.0) at 95-100°C for 15-20 minutes may significantly enhance signal detection, particularly in formalin-fixed paraffin-embedded tissues where protein cross-linking can mask epitopes.

How can researchers validate the specificity of FITC-conjugated SLC12A3 antibodies?

Validating the specificity of FITC-conjugated SLC12A3 antibodies requires a multi-faceted approach. First, perform immunostaining in kidney tissue where SLC12A3 is known to be exclusively expressed in distal convoluted tubules, comparing staining patterns with established markers of this segment. Second, implement specific controls: (1) peptide competition assays where pre-incubation of the antibody with the immunizing peptide should abolish specific staining; (2) comparative analysis using multiple antibodies targeting different epitopes of SLC12A3; and (3) knockout/knockdown validation where tissues from SLC12A3 knockout models or cells with CRISPR-mediated SLC12A3 deletion should show absence of signal . For analyzing potential cross-reactivity, Western blotting can confirm a single band at the expected molecular weight (~120-130 kDa for glycosylated SLC12A3). Additionally, validation in disease models with known SLC12A3 dysfunction, such as Gitelman syndrome patient samples, can provide further confirmation of specificity through expected alterations in expression or localization patterns .

What is the recommended protocol for using FITC-conjugated SLC12A3 antibodies in flow cytometry when analyzing primary renal cells?

When analyzing primary renal cells by flow cytometry using FITC-conjugated SLC12A3 antibodies, the following methodology is recommended: Begin with single-cell suspensions of freshly isolated kidney cells or cultured renal cells obtained through gentle enzymatic digestion (collagenase type II, 1mg/ml) at 37°C for 30 minutes with minimal mechanical disruption. Fix cells with 2% paraformaldehyde for 10 minutes at room temperature, followed by permeabilization with 0.1% saponin in PBS containing 1% BSA for membrane proteins. For intracellular epitopes, use 0.1% Triton X-100 instead. Incubate cells with the FITC-conjugated SLC12A3 antibody at optimized concentration (typically 1-5 μg/ml) for 45-60 minutes at 4°C in the dark. Include parallel samples with isotype control antibodies conjugated to FITC at equivalent concentrations to establish background fluorescence thresholds. For multi-parameter analysis, combine with antibodies against nephron segment markers (e.g., AQP1 for proximal tubules) conjugated to spectrally distinct fluorophores like PE or APC. Implement compensation controls when using multiple fluorophores to correct for spectral overlap. Set gating strategies to first exclude debris and doublets, then identify viable cells before analyzing SLC12A3 expression within specific cell populations. This approach enables quantitative assessment of SLC12A3 expression levels across different renal cell subpopulations.

How can FITC-conjugated SLC12A3 antibodies be utilized to study trafficking defects in Gitelman syndrome models?

To study trafficking defects in Gitelman syndrome models using FITC-conjugated SLC12A3 antibodies, researchers should implement a combined live-cell imaging and pulse-chase approach. First, establish cellular models expressing wild-type SLC12A3 and disease-specific mutants (such as Thr60Met, Gly141Arg, or Cys994Tyr) identified in Gitelman syndrome patients . For live trafficking studies, use a surface biotinylation assay followed by immunoprecipitation with the FITC-conjugated SLC12A3 antibody to quantify plasma membrane versus intracellular distribution ratios. Time-course experiments can track protein movement from the endoplasmic reticulum to the plasma membrane using organelle-specific markers. Confocal microscopy with z-stack acquisition enables 3D reconstruction of SLC12A3 distribution patterns, revealing whether mutant proteins are retained in specific subcellular compartments. To distinguish between folding defects and trafficking machinery problems, combine with inhibitors of specific trafficking pathways (Brefeldin A for ER-to-Golgi transport or Bafilomycin A1 for endosomal recycling). For pulse-chase experiments, synchronized release of proteins from the ER using temperature-sensitive protocols (storage at 20°C followed by shift to 37°C) can provide temporal resolution of trafficking kinetics. Co-localization analysis with markers for specific compartments (calnexin for ER, GM130 for Golgi, Rab11 for recycling endosomes) will pinpoint exactly where mutant proteins become arrested in the secretory pathway.

What are the considerations for designing co-localization studies between SLC12A3 and potential interacting proteins using FITC-conjugated antibodies?

When designing co-localization studies between FITC-conjugated SLC12A3 antibodies and potential interacting proteins, researchers must address several technical and biological considerations. First, select secondary fluorophores with minimal spectral overlap with FITC (e.g., Alexa Fluor 594 or 647) for the interacting protein to prevent bleed-through. Implement rigorous controls including single-labeled samples to establish proper imaging parameters. For advanced analysis, use super-resolution microscopy techniques such as Stimulated Emission Depletion (STED) or Stochastic Optical Reconstruction Microscopy (STORM) to overcome the diffraction limit, allowing resolution of proteins within 20-50 nm proximity. Biologically, consider the dynamic nature of protein interactions by incorporating live-cell imaging with photobleaching techniques like Fluorescence Recovery After Photobleaching (FRAP) or Fluorescence Loss in Photobleaching (FLIP) to assess kinetics of protein associations. For quantitative assessment, implement intensity correlation analysis methods like Pearson's correlation coefficient or Manders' overlap coefficient, but interpret these with caution as they do not necessarily indicate direct physical interaction. Complementary approaches such as Proximity Ligation Assay (PLA) or Förster Resonance Energy Transfer (FRET) should be used to confirm suspected interactions within the 10 nm range. When studying interactions with regulatory proteins like WNK kinases or SPAK/OSR1, consider examining both basal and stimulated conditions (e.g., low chloride or altered osmolarity) to capture regulated interactions.

How can FITC-conjugated SLC12A3 antibodies be utilized in CRISPR-Cas9 gene editing experiments to validate successful mutation correction?

FITC-conjugated SLC12A3 antibodies serve as critical validation tools in CRISPR-Cas9 gene editing experiments targeting SLC12A3 mutations. Following the approach documented in previous SLC12A3 correction studies , researchers should implement a multi-step validation strategy. First, after CRISPR-Cas9 editing of patient-derived induced pluripotent stem cells (hiPSCs) carrying SLC12A3 mutations, differentiate the cells toward renal lineages following established protocols. Then use FITC-conjugated SLC12A3 antibodies in immunofluorescence microscopy to compare protein expression patterns between wild-type, mutant, and corrected cells. Successfully corrected cells should demonstrate restoration of proper membrane localization and expression levels similar to wild-type controls.

Quantitatively assess correction efficiency by flow cytometry using the FITC-conjugated antibodies to measure the percentage of cells showing normal SLC12A3 expression levels and compare mean fluorescence intensity across populations. For functional validation, implement electrophysiological measurements of sodium-chloride cotransport activity in parallel with antibody-based detection to correlate protein expression with functional recovery. Creating a quantitative table of expression metrics (membrane-to-cytoplasmic ratios, colocalization with membrane markers, and total expression levels) across wild-type, mutant, and various corrected clones provides robust documentation of gene editing outcomes. This approach not only validates the genetic correction but also confirms proper protein folding, trafficking, and functional integration into the membrane – aspects that genomic sequencing alone cannot verify.

What are common causes of non-specific binding when using FITC-conjugated SLC12A3 antibodies and how can they be mitigated?

Non-specific binding with FITC-conjugated SLC12A3 antibodies can arise from multiple sources that require specific mitigation strategies. Excessive antibody concentration often leads to high background; perform titration experiments (typically testing 0.1-10 μg/ml) to determine the minimum concentration yielding specific signals. Inadequate blocking represents another common issue; use 5-10% normal serum from the species unrelated to the antibody source (typically goat or donkey) with 1% BSA for 1-2 hours at room temperature. Autofluorescence from tissues, particularly kidney samples with high metabolic activity, can be reduced by treating sections with 0.1% Sudan Black B in 70% ethanol for 20 minutes or using specialized quenching reagents like TrueBlack®.

When working with formalin-fixed tissues, endogenous biotin can cause background; block with avidin/biotin blocking kits before antibody incubation. Cross-reactivity with other sodium-chloride cotransporters in the SLC12 family may occur due to conserved epitopes; validate specificity using control tissues where SLC12A3 is absent but related transporters are present. In flow cytometry applications, dead cells often bind antibodies non-specifically; use viability dyes like propidium iodide or fixable viability dyes to exclude these populations. Finally, FITC itself can participate in non-specific interactions through hydrophobic or ionic mechanisms; adding 0.05% Tween-20 to wash buffers and maintaining physiological salt concentrations (150 mM NaCl) can minimize these interactions. For persistent problems, consider using Fab fragments instead of whole IgG antibodies to reduce Fc-mediated binding to Fc receptors on immune cells that may be present in kidney tissues.

How can researchers distinguish between true SLC12A3 signal reduction and technical artifacts when studying disease models?

Distinguishing between genuine SLC12A3 signal reduction and technical artifacts in disease models requires implementation of multiple internal controls and standardized protocols. First, include calibration standards in each experiment by using reference tissues or cells with known SLC12A3 expression levels processed alongside experimental samples. When working with patient-derived samples, process age/sex-matched control samples simultaneously using identical protocols to normalize for technical variation. For signal quantification, implement ratio-metric approaches by co-staining with a stable reference protein (e.g., Na+/K+-ATPase) that shouldn't change in the disease model, then calculate the SLC12A3:reference ratio to control for processing variations.

To account for epitope masking that might occur in disease states, use multiple antibodies targeting different SLC12A3 epitopes; true expression changes should be consistently detected across antibodies. Complement immunofluorescence data with orthogonal methods such as quantitative RT-PCR for mRNA expression and Western blotting for total protein levels to distinguish between transcriptional, translational, or localization changes. For cases where protein trafficking defects are suspected, as in many Gitelman syndrome mutations , use subcellular fractionation to quantify SLC12A3 in membrane versus cytosolic fractions, which can reveal whether the protein is produced but fails to reach the membrane. Finally, create a comprehensive table documenting all technical parameters (fixation time, antibody lot, incubation temperature, imaging settings) across experiments to identify potential sources of variability that might be misinterpreted as biological differences.

How should researchers interpret changes in SLC12A3 localization patterns when comparing wild-type and mutant proteins?

When interpreting changes in SLC12A3 localization patterns between wild-type and mutant proteins, researchers should implement a systematic analytical framework that distinguishes pathological alterations from normal variations. First, establish a quantitative baseline of normal SLC12A3 distribution using wild-type controls, measuring parameters such as membrane-to-cytoplasmic fluorescence ratio, colocalization with membrane markers (e.g., E-cadherin), and distribution along the apical-basolateral axis. For mutant proteins, particularly those associated with Gitelman syndrome like Gly141Arg or Thr235Lys , analyze whether the observed mislocalization follows established patterns for protein trafficking defects: ER retention would show colocalization with calnexin, Golgi accumulation with GM130, or endosomal trapping with Rab7/Rab11 markers.

Create a classification system for mutation effects based on localization patterns:

  • Class 1: Complete intracellular retention (>90% non-membrane signal)

  • Class 2: Partial trafficking defect (30-90% non-membrane signal)

  • Class 3: Normal trafficking but functional impairment

  • Class 4: Accelerated membrane removal/degradation

This classification helps correlate genotype with cellular phenotype. Additionally, analyze the effects of temperature manipulation (reduced temperature cultivation at 27°C) or chemical chaperones (4-phenylbutyrate) on mutant protein localization, as trafficking defects that can be rescued by these interventions often indicate protein folding issues rather than targeting signal disruption. For comprehensive analysis, compile data into a quantitative table showing the subcellular distribution metrics for each mutation, correlating these patterns with the specific domain affected (transmembrane, cytoplasmic loop, etc.) to identify structure-function relationships in SLC12A3 trafficking.

What are the relative advantages of FITC-conjugated SLC12A3 antibodies compared to other fluorophore conjugates for multiplex imaging?

FITC-conjugated SLC12A3 antibodies offer several distinct advantages in multiplex imaging contexts, balanced by important limitations that inform experimental design. FITC emits in the green spectrum (519 nm), positioning it strategically in multiplexing panels between blue (DAPI for nuclei) and red/far-red fluorophores. This allows for clear channel separation when combined with fluorophores like Texas Red, Cy5, or Alexa Fluor 647. FITC has a relatively high quantum yield (~0.93) compared to some other fluorophores, providing good signal intensity with moderate exposure times. Financially, FITC conjugates are typically more affordable than newer proprietary fluorophores, offering cost advantages for large-scale studies.

How do fluorescence-based detection methods using FITC-conjugated antibodies compare with mass spectrometry approaches for quantifying SLC12A3 expression?

Mass spectrometry approaches, particularly targeted methods like multiple reaction monitoring (MRM) or parallel reaction monitoring (PRM), offer absolute quantification of SLC12A3 protein abundance without epitope accessibility concerns. These methods can simultaneously quantify multiple proteoforms and post-translational modifications of SLC12A3, providing insights into regulatory mechanisms through comprehensive modification profiles. Unlike antibody methods, mass spectrometry can distinguish between highly homologous proteins by detecting peptides unique to SLC12A3 versus related transporters.

What methodological considerations are important when transitioning from conventional immunohistochemistry to quantitative high-content imaging with FITC-conjugated SLC12A3 antibodies?

Transitioning from conventional immunohistochemistry to quantitative high-content imaging with FITC-conjugated SLC12A3 antibodies requires methodological adjustments across sample preparation, image acquisition, and data analysis phases. For sample preparation, conventional IHC protocols must be optimized for fluorescence detection: minimize fixation time to reduce autofluorescence (12-24 hours maximum in 4% PFA), implement autofluorescence quenching steps using sodium borohydride (0.1% for 5 minutes) or specialized quenching reagents, and use mounting media with anti-fade agents to prevent photobleaching during extended imaging sessions. Antibody concentration requires careful titration specifically for fluorescence applications, typically using lower concentrations (0.5-2 μg/ml) than chromogenic IHC to optimize signal-to-noise ratios.

For image acquisition, implement standardized microscopy settings across all specimens, including exposure time, gain, and offset, ideally using automation to eliminate operator variability. Capture multiple random fields per sample (minimum 10-15) to account for tissue heterogeneity, and acquire z-stacks (0.5 μm steps) to ensure complete capture of membrane signals. Establish rigorous quality control parameters such as minimum signal-to-background ratios and maximum coefficient of variation between replicate fields to exclude suboptimal images from analysis.

Data analysis requires transition from qualitative assessment to quantitative image analysis: implement automated segmentation algorithms to identify cell boundaries, subcellular compartments, and membrane regions using machine learning approaches for complex tissue architecture. For SLC12A3 quantification, define specific measurements including membrane-to-cytoplasm ratio, total fluorescence intensity, and colocalization metrics with membrane markers. Normalize measurements using internal reference standards (fluorescent beads or invariant structural proteins) to enable cross-sample and cross-experimental comparisons. Finally, implement statistical validation including power analysis to determine minimum sample sizes and appropriate statistical tests for specific hypotheses regarding SLC12A3 expression or localization changes in experimental conditions.

What are the necessary validation steps when developing custom FITC-conjugated SLC12A3 antibodies?

Developing custom FITC-conjugated SLC12A3 antibodies requires a comprehensive validation pipeline to ensure specificity, sensitivity, and reproducibility. Initial validation begins with confirming antibody binding to the target epitope through ELISA using both the immunizing peptide and recombinant full-length SLC12A3 protein. Western blotting against kidney tissue lysates must demonstrate a single band at the expected molecular weight (~120-130 kDa), with additional validation using SLC12A3-transfected versus non-transfected cell lines. The FITC conjugation process itself requires optimization of the fluorophore-to-antibody ratio (typically 3-5 FITC molecules per antibody) through spectrophotometric analysis, balancing maximum fluorescence signal against potential interference with antigen binding.

Critical specificity controls include testing the antibody in SLC12A3 knockout tissue or CRISPR-edited cells with SLC12A3 deletion , which should show complete absence of signal. Cross-reactivity assessment against related SLC family transporters (particularly SLC12A1 and SLC12A2) is essential due to sequence homology. For immunolocalization validation, the antibody must demonstrate the known exclusive expression pattern in distal convoluted tubule cells of kidney tissue , with no detection in other nephron segments. Performance consistency requires lot-to-lot validation using reference standards and stability testing under various storage conditions (4°C, -20°C, -80°C) and freeze-thaw cycles. Finally, application-specific validation is necessary for each intended use (immunofluorescence, flow cytometry, immunoprecipitation) as FITC conjugation may differentially affect performance across applications. Documenting these validation steps in a comprehensive table with quantitative metrics for each parameter provides essential quality assurance for subsequent experimental applications.

How can FITC-conjugated SLC12A3 antibodies be utilized to study phosphorylation-dependent regulation of the protein?

Studying phosphorylation-dependent regulation of SLC12A3 using FITC-conjugated antibodies requires a sophisticated experimental approach integrating multiple techniques. SLC12A3 function is regulated through phosphorylation at multiple sites, particularly by WNK/SPAK/OSR1 kinase pathways, with key phosphorylation sites including Thr60, which is directly implicated in Gitelman syndrome when mutated . Begin by using phospho-specific antibodies against known SLC12A3 phosphorylation sites in combination with the FITC-conjugated pan-SLC12A3 antibody in dual immunofluorescence experiments. This allows visualization of how phosphorylation status correlates with subcellular localization and membrane insertion patterns.

For dynamic studies, implement live-cell imaging using kidney cell models expressing fluorescently-tagged SLC12A3 constructs with either phospho-mimetic (serine/threonine to glutamate) or phospho-resistant (serine/threonine to alanine) mutations at key regulatory sites. Compare trafficking patterns between these mutants using FITC-conjugated SLC12A3 antibodies that recognize surface-exposed epitopes in non-permeabilized cells to quantify membrane insertion rates. For comprehensive phosphorylation profiling, combine immunoprecipitation using the FITC-conjugated SLC12A3 antibody followed by mass spectrometry analysis to identify phosphorylation sites and their relative occupancy under various stimuli (hypotonic stress, low chloride conditions, aldosterone treatment).

Create a quantitative experimental matrix documenting the relationship between specific phosphorylation events and functional outcomes by correlating phosphorylation status (detected by phospho-specific antibodies) with membrane localization (quantified using surface biotinylation) and transport activity (measured by radioactive ion flux assays or intracellular ion-sensitive fluorescent indicators). This approach provides mechanistic insights into how post-translational modifications regulate SLC12A3 function in both physiological and pathological contexts.

What considerations are important when using FITC-conjugated SLC12A3 antibodies for diagnostic applications in kidney diseases?

When using FITC-conjugated SLC12A3 antibodies for diagnostic applications in kidney diseases, particularly Gitelman syndrome, several critical considerations must be addressed to ensure accurate interpretation. First, establish standardized protocols with precise fixation parameters, antibody concentrations, and imaging settings to enable reliable quantitative comparisons across patient samples. Create normative reference ranges for SLC12A3 expression and localization patterns using healthy control kidney samples stratified by age, sex, and genetic background, as expression levels may vary physiologically. For diagnostic accuracy, implement dual staining with markers of distal convoluted tubules (e.g., parvalbumin or NCC) to correctly identify the nephron segment expressing SLC12A3, preventing false interpretations due to tubular atrophy or segmental transitions.

In suspected Gitelman syndrome cases, employ pattern recognition approaches to classify SLC12A3 abnormalities: complete absence, reduced expression, or abnormal localization . Correlate these patterns with specific mutation types documented in genetic testing, as different mutations may produce distinct protein expression patterns. For example, mutations affecting the Thr60 phosphorylation site show normal synthesis but defective membrane trafficking . Implement quantitative image analysis using machine learning algorithms to detect subtle changes in distribution patterns that might not be evident through visual inspection alone.

Establish clear criteria distinguishing primary SLC12A3 defects from secondary changes due to other kidney diseases or medication effects, particularly diuretics. Document limitations of antibody-based diagnostics, including the inability to detect functional defects when protein expression and localization appear normal. Finally, correlate antibody-based findings with clinical parameters including serum electrolyte levels and urinary electrolyte excretion to establish diagnostic algorithms integrating molecular, histological, and clinical data for comprehensive patient evaluation.

Table 1: Comparison of Detection Methods for SLC12A3 in Research Applications

MethodSpatial ResolutionSensitivityQuantification CapabilityMain AdvantagesMain Limitations
FITC-conjugated antibody (IF)Subcellular (200-300 nm)500-1000 molecules/cellSemi-quantitativeSpatial information on trafficking and localizationPhotobleaching, epitope accessibility
Alexa Fluor 488-conjugated antibody (IF)Subcellular (200-300 nm)300-800 molecules/cellSemi-quantitativeHigher photostability, pH insensitivityHigher cost, narrower stokes shift
Western blottingNone10-50 ng total proteinSemi-quantitativeDifferent glycoforms distinguishableLoss of spatial information
Targeted mass spectrometryNone5-10 ng total proteinAbsolute quantitativePTM identification, isoform-specific quantificationComplex sample preparation, no spatial info
Flow cytometry (FITC-conjugated)Cellular200-500 molecules/cellRelative quantitativeSingle-cell analysis, high throughputLoss of tissue context and architecture
Super-resolution microscopy (FITC)Subcellular (50-100 nm)100-300 molecules/cellSemi-quantitativeNanoscale distribution patterns visibleTechnical complexity, specialized equipment

Table 2: Common SLC12A3 Mutations and Their Effects on Protein Expression and Function

MutationProtein DomainEffect on Protein ExpressionEffect on Membrane LocalizationAssociated Clinical PhenotypeDetectable by FITC-antibodyReference
Thr60MetN-terminal cytoplasmicNormal synthesisDecreased membrane expressionClassical GS, moderate hypokalemiaYes, shows ER retention
Gly141ArgTransmembrane domain 1Normal synthesisSevere trafficking defectGS with early onset, severeYes, shows perinuclear accumulation
Leu170GlnTransmembrane domain 2Reduced stabilityPartial membrane expressionMild GS phenotypeYes, diffuse staining pattern
Thr235LysExtracellular loop 2Normal synthesisModerate trafficking defectGS with metabolic syndromeYes, partial membrane localization
Arg913GlnC-terminal cytoplasmicNormal synthesis and traffickingFunctional defect despite normal localizationGS with association to diabetic kidney diseaseYes, normal localization pattern
Cys994TyrC-terminal cytoplasmicReduced stabilityDecreased membrane expressionClassical GS phenotypeYes, reduced intensity, diffuse pattern

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 2025 TheBiotek. All Rights Reserved.