SLC51B Human

Solute Carrier Family 51 Beta Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Role in Physiology

The OSTα-OSTβ complex mediates:

  • Bile Acid Transport: Facilitates basolateral export of taurocholate and glycine-conjugated bile acids from enterocytes to portal blood .

  • Sterol Circulation: Transports estrone sulfate, dehydroepiandrosterone sulfate, and prostaglandin E2 .

  • Neurosteroid Regulation: Expressed in adrenal glands and brain steroidogenic cells, influencing neurosteroid homeostasis .

Functional studies show SLC51B is required for:

  • Trafficking SLC51A to the plasma membrane .

  • Maintaining SLC51A stability and glycosylation .

Disease Associations

ConditionMechanism/Evidence
Cholestatic DisordersMutations linked to congenital chronic diarrhea and cholestatic liver disease .
Biliary AtresiaElevated OSTα/β expression observed in primary biliary cholangitis .
MODY3 Renal DiseaseHNF1A deficiency reduces SLC51B expression, impairing estrone sulfate uptake and nephroprotection .
Nonalcoholic SteatohepatitisUpregulated hepatic OSTα/β correlates with disease progression .

Clinical Implications

  • Mouse Models: Slc51a knockout mice show abolished Ostβ expression and impaired bile acid absorption .

  • Human Studies: SLC51B-deficient patients exhibit severe bile acid malabsorption and cholestasis .

Diagnostic and Therapeutic Tools

  • Antibodies: Anti-SLC51B antibodies (e.g., ab121285) validate protein expression in IHC/WB, showing high reactivity in intestine, kidney, and testis .

  • ELISA Kits: Commercial kits (e.g., Abbexa) enable quantitative SLC51B measurement in research settings .

Expression and Tissue Distribution

SLC51B is highly expressed in:

  • Gastrointestinal Tract: Small intestine, colon .

  • Excretory Organs: Liver, kidney .

  • Steroidogenic Tissues: Adrenal gland, testis, ovary .

Immunohistochemistry reveals cytoplasmic/membranous localization in enterocytes and renal tubules .

Research Applications

  • Drug Development: Target for modulating bile acid homeostasis in cholestasis .

  • Renal Studies: Investigated for estrone sulfate uptake defects in MODY3 nephropathy .

  • Membrane Protein Studies: Purified recombinant SLC51B enables structural/functional assays .

Product Specs

Introduction
The Solute Carrier Family 51 Beta (SLC51B) gene is responsible for encoding the SLC51B protein, which functions as a subunit of the organic solute transporter SLC51. SLC51 requires two distinct proteins to form a heterodimer for transport activity. However, the specific roles of each subunit in this process are not fully understood. Research indicates that SLC51B is crucial for the proper trafficking of SLC51A and the formation of a functional transport unit. Additionally, specific amino acid residues within SLC51B have been identified as essential for these functions. SLC51B has been linked to diseases such as extrahepatic cholestasis and biliary atresia.
Description
Recombinant SLC51B protein, produced in E. coli, is available as a single, non-glycosylated polypeptide chain. This protein consists of 95 amino acids (spanning positions 57-128) and has a molecular weight of 10.7 kDa. For purification purposes, a 23 amino acid His-tag is fused to the N-terminus of the SLC51B protein, and proprietary chromatographic techniques are employed.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The SLC51B protein solution is provided at a concentration of 1 mg/ml and is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 10% glycerol, and 0.1 M NaCl.
Stability
For optimal storage, it is recommended to keep the entire vial at 4°C if it will be used within 2-4 weeks. For extended storage, the protein should be stored frozen at -20°C. To ensure long-term stability during storage, consider adding a carrier protein (0.1% HSA or BSA). It is important to avoid repeated freeze-thaw cycles.
Purity
The purity of the SLC51B protein is determined to be greater than 95.0% based on SDS-PAGE analysis.
Synonyms
OSTB, OSTBETA, Organic solute transporter subunit beta, OST-beta, Solute carrier family 51 subunit beta.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSRSIQASR KEKMQPPEKE TPEVLHLDEA KDHNSLNNLR ETLLSEKPNL AQVELELKER DVLSVFLPDV PETES

Q&A

What is SLC51B and what is its primary function?

SLC51B, also known as OSTβ, is a 128-amino acid protein with a single transmembrane domain that forms a functional heterodimeric transporter with SLC51A (OSTα). Together, they constitute the OSTα-OSTβ complex that serves as the intestinal basolateral transporter responsible for bile acid export from enterocytes into portal blood. This complex is essential for intestinal bile acid absorption and consequently for dietary lipid absorption. The transporter is expressed at the basolateral membrane of epithelium in multiple tissues including intestine, kidney, liver, testis, and adrenal gland . Research methodologies to study this function typically involve radioactive bile acid transport assays in cell models expressing recombinant SLC51A and SLC51B proteins.

What is the protein structure of human SLC51B?

Human SLC51B is a Type Ib membrane protein with the following characteristics:

ParameterInformation
Protein NameOrganic solute transporter subunit beta
UniProt IDQ86UW2
AliasesOSTB, OSTbeta
OrganismHomo sapiens (Human)
Transmembrane Domains1
Length (amino acids)128
Amino Acid SequenceMEHSEGAPGDPAGTVVPQELLEEMLWFFRVEDASPWNHSILALAAVVVIISMVLLGRSIQASRKEKMQPPEKETPEVLHLDEAKDHNSLNNLRETLLSEKPNLAQVELELKERDVLSVFLPDVPETES

To study the protein structure, researchers typically use molecular modeling approaches based on homology, or experimental methods such as X-ray crystallography or cryo-electron microscopy, though the small size of SLC51B presents unique challenges that require specialized techniques .

What are the primary binding partners of SLC51B?

SLC51B's primary binding partner is SLC51A, with which it forms the functional heterodimeric OSTα-OSTβ complex. This partnership is essential as expression of both subunits is absolutely required for trafficking of the proteins from the endoplasmic reticulum to the plasma membrane and for bile acid transport activity . Additionally, protein interaction network analysis shows strong functional association with:

ProteinFunctionInteraction Score
SLC51AForms heterodimer for bile acid transport0.999
CYP7A1Rate-limiting enzyme in bile acid biosynthesis0.841
SLC10A2Sodium-dependent reabsorption of bile acids0.838
ABCC3Alternative route for export of bile acids0.815

Protein-protein interaction studies typically employ co-immunoprecipitation, yeast two-hybrid assays, or fluorescence resonance energy transfer (FRET) methodologies to confirm these interactions experimentally .

How does the regulation of SLC51B expression impact bile acid homeostasis?

The regulation of SLC51B expression significantly impacts bile acid homeostasis through multiple mechanisms. Research has demonstrated that both SLC51A and SLC51B subunits must be expressed for proper function of the heterodimeric complex. Studies in knockout mouse models have revealed that deletion of Slc51a leads to loss of expression for both Ostα and Ostβ proteins, resulting in impaired intestinal bile acid absorption . This disruption leads to reduced levels of hepatic bile acid synthesis due to altered FXR/FGF15 signaling in the gut-liver axis.

Methodologically, researchers investigating these regulatory mechanisms typically employ:

  • Conditional knockout mouse models specific to intestinal or hepatic tissues

  • Gene expression analysis via qPCR and RNA-sequencing

  • Chromatin immunoprecipitation (ChIP) assays to identify transcription factor binding sites

  • Bile acid measurements using HPLC electrospray tandem mass spectrometry

  • Intestinal absorption studies using radiolabeled bile acids

The effects of disrupted SLC51B expression can be assessed through measurements of serum and fecal bile acid concentrations, providing insights into the extent of impaired enterohepatic circulation .

What transcription factors regulate SLC51B expression in different tissues?

Recent research has identified HNF1A (Hepatocyte Nuclear Factor 1 Alpha) as a key transcriptional regulator of SLC51B in human kidney cells. ChIP-Sequencing (ChIP-Seq) performed on human pluripotent stem cell-derived kidney organoids has identified genome-wide putative targets of HNF1A, including SLC51B . Loss of HNF1A results in reduced SLC51B expression, which has functional consequences for estrone sulfate transport.

To study transcription factor regulation experimentally, researchers employ:

  • Promoter analysis using luciferase reporter assays

  • ChIP-Seq to identify transcription factor binding sites

  • EMSA (Electrophoretic Mobility Shift Assays) to confirm direct binding

  • Site-directed mutagenesis of predicted binding sites

  • Gene expression analysis in tissue-specific cell models with transcription factor knockdown/overexpression

These methodological approaches have revealed tissue-specific regulation of SLC51B, with different transcription factors controlling its expression in intestine, liver, and kidney, allowing for specialized functional adaptations in each organ system .

What is the functional significance of SLC51B in estrone sulfate transport in kidney cells?

Beyond its established role in bile acid transport, research has revealed that the OSTα-OSTβ complex also mediates estrone sulfate (E1S) transport in renal proximal tubule cells. Studies using HNF1A-depleted human renal proximal tubule epithelial cells (RPTECs) and MODY3 (Maturity Onset Diabetes of the Young 3) hiPSC-derived kidney organoids have demonstrated reduced SLC51B expression and consequently impaired E1S uptake .

This finding has significant implications as E1S serves as the main storage form of nephroprotective estradiol in the human body. Reduced E1S uptake and increased urinary E1S excretion may diminish the availability of nephroprotective estradiol in kidneys, potentially contributing to renal disease development in MODY3 patients .

Research methodologies to study this function include:

  • Radiolabeled estrone sulfate uptake assays in cellular models

  • Urinary E1S measurements in patient samples

  • Gene knockdown/knockout experiments in kidney cell lines

  • Human kidney organoid models derived from patient iPSCs

  • Correlation analyses between SLC51B expression and kidney disease parameters

What phenotypes are associated with SLC51B mutations in humans?

The first patients with OSTβ deficiency due to SLC51B mutations were reported in 2018. Clinical manifestations in these patients included:

Clinical FeaturesLaboratory Findings
Chronic diarrheaElevated serum gamma-glutamyltransferase
Severe fat-soluble vitamin deficiencyAbnormal bile acid profiles
Features of cholestatic liver diseaseAltered bile acid excretion patterns

Whole exome sequencing revealed a homozygous single nucleotide deletion in codon 27 of SLC51B, resulting in a frameshift and premature termination at codon 50 (p.F27fs). Functional studies in transfected cells demonstrated that this mutation truncates the OSTβ protein and markedly impairs synthesis of the OSTα-OSTβ complex and bile acid transport activity .

Research methodologies to characterize these phenotypes include:

  • Whole exome sequencing for mutation identification

  • In vitro expression studies using mutant constructs

  • Bile acid transport assays in cell culture models

  • Detailed clinical phenotyping of affected individuals

  • Family-based segregation analysis of mutations and phenotypes

How can SLC51B function be assessed in experimental models?

Researchers employ multiple complementary approaches to assess SLC51B function:

  • In Vitro Cell-Based Assays:

    • Transfection of COS cells with wild-type or mutant SLC51B constructs

    • Co-transfection with SLC51A to form functional heterodimers

    • Radiolabeled substrate transport assays (e.g., [³H]taurocholic acid)

    • Subcellular localization studies using immunofluorescence or tagged proteins

    • Site-directed mutagenesis to assess structure-function relationships

  • Animal Models:

    • Slc51b knockout mice

    • Tissue-specific conditional knockout models

    • Humanized mouse models expressing human SLC51B variants

    • Bile acid homeostasis assessment via fecal/serum bile acid measurements

  • Patient-Derived Models:

    • iPSC-derived organoids from patients with SLC51B mutations

    • Primary cell cultures from patient biopsies

    • Ex vivo intestinal or liver slice cultures

These methodological approaches allow for comprehensive functional assessment of SLC51B under normal conditions and in disease states .

What is the role of SLC51B in liver diseases like nonalcoholic steatohepatitis and primary biliary cholangitis?

Research has shown elevated expression of OSTα/β in the liver of patients with nonalcoholic steatohepatitis (NASH) and primary biliary cholangitis (PBC) . This suggests adaptive responses in bile acid transport mechanisms during these disease states. The altered expression of SLC51B likely represents a compensatory mechanism to facilitate bile acid export from hepatocytes under cholestatic conditions.

Methodological approaches to study this role include:

  • Analysis of liver biopsies from patients with NASH and PBC for SLC51B expression

  • Correlation of SLC51B expression levels with disease severity metrics

  • In vitro studies using hepatocyte models exposed to inflammatory or cholestatic stimuli

  • Animal models of NASH and PBC with assessment of SLC51B expression

  • Therapeutic interventions targeting SLC51B expression or function

Understanding these changes in SLC51B expression may provide insights into disease pathogenesis and potential therapeutic targets for these challenging liver diseases .

What are the optimal methods for assessing SLC51B-mediated transport in vitro?

Researchers studying SLC51B-mediated transport employ several methodological approaches:

  • Cell Model Selection:

    • COS cells (commonly used for heterologous expression)

    • HEK293 cells (suitable for stable expression)

    • Cell lines derived from relevant tissues (intestinal, renal, hepatic)

  • Expression System:

    • Co-transfection of SLC51A and SLC51B expression vectors

    • Creation of stable cell lines using lentiviral systems

    • Inducible expression systems for temporal control

  • Transport Assay Protocol:

    • Cells are typically grown to confluence in 24-well plates

    • Preincubation in transport buffer (modified HBSS with appropriate ion composition)

    • Incubation with radiolabeled substrate (e.g., [³H]taurocholic acid) at 37°C

    • Rapid washing to remove unbound substrate

    • Cell lysis and measurement of internalized radioactivity

    • Normalization to protein content

  • Data Analysis:

    • Calculation of transport kinetics (Km, Vmax)

    • Inhibition studies to determine specificity

    • Comparison of wild-type vs. mutant SLC51B function

This methodological framework allows for robust assessment of SLC51B-mediated transport function and can be adapted for various substrates beyond bile acids .

How can researchers effectively study SLC51B regulation in kidney organoid models?

The study of SLC51B regulation in kidney organoid models requires specialized methodologies:

  • Generation of Kidney Organoids:

    • Differentiation of human pluripotent stem cells into kidney organoids

    • Patient-derived iPSCs can be used to model genetic variants

    • Verification of proximal tubule development via marker expression

  • Gene Expression Analysis:

    • qPCR for temporal expression patterns during differentiation

    • RNA-sequencing for genome-wide expression changes

    • Single-cell RNA-seq to identify cell-type specific expression

  • Transcriptional Regulation:

    • ChIP-Seq to identify transcription factor binding sites

    • Generation of reporter constructs with SLC51B promoter elements

    • CRISPR-mediated knockdown of potential regulators (e.g., HNF1A)

  • Functional Assessments:

    • Transport assays using fluorescent or radiolabeled substrates

    • Live imaging of transport using fluorescent bile acid analogues

    • Correlation of transport function with gene expression

These approaches have successfully identified HNF1A as a key regulator of SLC51B in kidney cells, with direct implications for understanding renal disease in MODY3 patients .

What analytical methods are used to quantify bile acids and other SLC51B substrates in biological samples?

Accurate quantification of bile acids and other SLC51B substrates is essential for functional studies. Current state-of-the-art methods include:

  • HPLC Electrospray Tandem Mass Spectrometry:

    • Gold standard for bile acid profiling in plasma, urine, and tissues

    • Allows for separation and quantification of individual bile acid species

    • Can detect conjugated and unconjugated forms

    • Requires careful sample preparation and internal standards

  • Radioimmunoassay (RIA):

    • Used for specific bile acids with available antibodies

    • Less comprehensive than mass spectrometry approaches

    • Useful for high-throughput screening applications

  • Enzymatic Assays:

    • Suitable for total bile acid measurements

    • Less specific for individual bile acid species

    • Typically employed for clinical diagnostic applications

  • UPLC-MS/MS for Non-Bile Acid Substrates:

    • Optimized for estrone sulfate, digoxin, and other SLC51B substrates

    • Requires specific extraction procedures for each substrate type

    • Enables comprehensive profiling of transport activity

These analytical methods are essential for characterizing SLC51B function in various physiological and pathological contexts, and for assessing the consequences of genetic variants or pharmacological interventions .

What are the emerging therapeutic implications of targeting SLC51B?

Understanding SLC51B function opens potential therapeutic avenues that researchers are actively exploring:

  • Bile Acid Disorders:

    • Development of SLC51B modulators for cholestatic conditions

    • Compensatory upregulation strategies for patients with partial SLC51B deficiency

    • Gene therapy approaches for complete loss-of-function mutations

  • Kidney Protection:

    • Targeted interventions to enhance SLC51B-mediated estrone sulfate uptake

    • Development of nephroprotective strategies for MODY3 patients

    • Screening for compounds that can stabilize HNF1A-SLC51B interactions

  • Metabolic Disease:

    • Exploration of SLC51B's role in energy homeostasis

    • Potential interventions for diabetes and metabolic syndrome

    • Investigation of triglyceride and glucose metabolism connections

Methodological approaches would include:

  • High-throughput screening for SLC51B modulators

  • Structure-based drug design targeting the OSTα-OSTβ interface

  • Preclinical testing in relevant disease models

  • Biomarker development for patient stratification

How do age-related changes in SLC51B expression impact bile acid homeostasis?

This remains an understudied area with significant implications for aging populations. Research methodologies to address this question would include:

  • Age-stratified expression analysis in human and animal tissues

  • Functional transport studies in primary cells isolated from donors of different ages

  • Correlation of SLC51B expression with age-related changes in bile acid profiles

  • Exploration of epigenetic mechanisms regulating SLC51B expression during aging

  • Investigation of potential interventions to maintain optimal SLC51B function with aging

What are the research challenges in developing tissue-specific models for SLC51B function?

Researchers face several methodological challenges when developing tissue-specific models:

  • Technical Challenges:

    • Achieving physiologically relevant expression levels of both OSTα and OSTβ

    • Recreating tissue-specific regulatory mechanisms

    • Maintaining appropriate polarization of epithelial cell models

    • Development of assays that reflect in vivo transport kinetics

  • Biological Complexity:

    • Tissue-specific post-translational modifications

    • Interaction with tissue-specific partner proteins

    • Influence of local microenvironment on transport function

    • Differential substrate preferences in various tissues

  • Experimental Approaches:

    • Tissue-specific conditional knockout animals

    • Advanced organoid models incorporating multiple cell types

    • Microfluidic systems to study vectorial transport

    • In silico modeling of tissue-specific functions

Addressing these challenges will enhance our understanding of SLC51B's diverse roles across different tissues and potentially reveal novel functions beyond the currently established roles in bile acid and estrone sulfate transport.

Product Science Overview

Gene and Protein Structure

The SLC51B gene is located on chromosome 15 in humans and encodes the beta subunit of the organic solute transporter (OST) complex. The OST complex is a heterodimer composed of two subunits: OST-alpha (SLC51A) and OST-beta (SLC51B). The beta subunit is essential for the proper functioning of the OST complex, as it modulates the glycosylation, membrane trafficking, and stability of the alpha subunit .

Function and Mechanism

The primary function of the OST complex is to facilitate the export of bile acids from enterocytes (intestinal cells) into the portal blood, which is a critical step in the enterohepatic circulation of bile acids. The OST complex efficiently transports major bile acid species, such as taurocholate, and can also transport steroids like estrone 3-sulfate and dehydroepiandrosterone 3-sulfate. Additionally, it plays a role in the transport of eicosanoids, such as prostaglandin E2 .

Biological Significance

The proper functioning of the SLC51B gene is vital for maintaining bile acid homeostasis and overall metabolic health. Mutations or dysregulation of this gene can lead to various disorders, including bile acid malabsorption and visceral myopathy. The OST complex’s ability to transport a wide range of substrates highlights its importance in multiple physiological processes, including lipid metabolism and steroid hormone regulation .

Recombinant SLC51B

Recombinant human SLC51B is produced using advanced biotechnological methods to study its structure, function, and potential therapeutic applications. The recombinant protein is typically expressed in suitable host cells, purified, and used in various research applications, including functional assays, structural studies, and drug development .

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.