SAR1B Human

GTP-Binding Protein SAR1B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to SAR1B Human

SAR1B is a small GTPase encoded by the SAR1B gene (secretion-associated Ras-related GTPase 1B) located on chromosome 5q31.1 in humans . It plays a critical role in COPII-mediated vesicle transport from the endoplasmic reticulum (ER) to the Golgi apparatus, particularly in lipid and protein secretion . SAR1B is highly conserved across mammals, sharing ~90% amino acid identity with its paralog SAR1A . While SAR1A is ubiquitously expressed, SAR1B exhibits tissue-specific roles, notably in enterocytes, hepatocytes, and muscle cells .

Functional Role of SAR1B

SAR1B is essential for the following cellular processes:

FunctionMechanismTissue-Specific RoleSource
Chylomicron transportInitiates COPII coat assembly to transport immature chylomicrons from ER to Golgi .Enterocytes (small intestine) absorb dietary fats
Lipid metabolismRegulates HDL formation via ABCA1-mediated cholesterol efflux .Hepatocytes (liver)
Calcium signalingFacilitates intracellular calcium transport in muscle cells .Cardiac and skeletal muscle

Chylomicron Retention Disease (CMRD)

Mutations in SAR1B cause CMRD, a rare autosomal recessive disorder characterized by:

  • Malabsorption of fats (e.g., triglycerides, cholesterol).

  • Deficiency of fat-soluble vitamins (A, D, E, K).

  • Hepatic steatosis and growth retardation .

Key Mutations in CMRD

Mutation TypeEffectFrequencySource
MissenseAltered GTPase activity or SEC23 binding (e.g., p.Arg19Cys) .~50% of cases
FrameshiftPremature stop codons (e.g., p.Glu83Lysfs*14) .~25% of cases
NonsenseTruncated protein (e.g., p.Tyr142*) .~15% of cases

Product Specs

Introduction
SAR1B, a small GTPase that forms homodimers, plays a crucial role in intracellular protein transport. This protein is involved in the transportation of molecules from the endoplasmic reticulum to the Golgi apparatus. Additionally, it participates in selecting protein cargo and assembling the COPII coat complex, which is essential for vesicle formation. Activation of SAR1B is mediated by the guanine nucleotide exchange factor PREB. Genetic defects in the SAR1B gene can lead to chylomicron retention disease (CMRD), also known as Anderson disease (ANDD), a condition affecting lipid absorption.
Description
This product consists of a recombinant human SAR1B protein produced in E. coli. It is a single, non-glycosylated polypeptide chain comprising 221 amino acids (with amino acids 1 to 198 being specific to SAR1B) and has a molecular weight of 24.8 kDa. A 23-amino acid His-tag is fused to the N-terminus of the protein to facilitate purification, which is carried out using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
The SAR1B protein is supplied in a solution at a concentration of 0.5 mg/ml. The solution also contains 20 mM Tris-HCl buffer (pH 8.0), 0.15 M NaCl, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein such as HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of this product is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
GTP-binding protein SAR1b, GTP-binding protein B, GTBPB, SAR1B, SARA2, SARB, ANDD, CMRD.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMSFIFDW IYSGFSSVLQ FLGLYKKTGK LVFLGLDNAG KTTLLHMLKD DRLGQHVPTL HPTSEELTIA GMTFTTFDLG GHVQARRVWK NYLPAINGIV FLVDCADHER LLESKEELDS LMTDETIANV PILILGNKID RPEAISEERL REMFGLYGQT TGKGSISLKE LNARPLEVFM CSVLKRQGYG EGFRWMAQYI D.

Q&A

What is the biological function of SAR1B in the secretory pathway?

SAR1B is a GTPase that functions as an essential component of the coat protein II (COPII) complex, initiating COPII coat assembly by recruiting other coat proteins to the endoplasmic reticulum (ER) membrane. This process facilitates the transport of proteins carrying signal peptides and/or transmembrane domains from the ER to the Golgi apparatus via COPII vesicles or tubules . SAR1B plays a particularly important role in lipid metabolism and intracellular chylomicron trafficking, with critical functions in vesicular COPII-dependent transport . The protein's GTPase activity is central to its function in the regulation of intracellular transport mechanisms, making it a key player in maintaining cellular protein trafficking homeostasis .

How do mutations in SAR1B relate to chylomicron retention disease?

Biallelic mutations in the SAR1B gene (formerly known as SARA2) are the genetic basis of chylomicron retention disease (CMRD), a rare recessive disorder characterized by impaired fat absorption . These mutations lead to defects in intracellular chylomicron trafficking and secretion, resulting in fat intestinal malabsorption, steatorrhea, chylomicron secretion failure, hypocholesterolemia, and hypoalphalipoproteinemia . The pathophysiology involves the retention of chylomicrons in enterocytes due to impaired COPII-dependent transport, which prevents their secretion into the lymphatic system . Mouse models with targeted defects similar to human SAR1B aberrations have been developed to gain more insight into CMRD, confirming that heterozygous mutations can recapitulate many of the gastrointestinal abnormalities observed in patients .

What experimental models are available for studying SAR1B function?

Several experimental models have been developed to study SAR1B function:

  • CRISPR-Cas9 engineered mouse models: Researchers have created mice with:

    • Deletion of 545 base pairs removing Sar1b exon 2 on chromosome 11 (Sar1b^del/+)

    • G-to-A substitution on chromosome 11 resulting in p.D137N amino acid change (Sar1b^mut/+)

    • Sar1a coding sequence replacing that of Sar1b at the endogenous Sar1b locus

  • Cell culture models: In vitro systems using cell lines like RKO (colorectal cancer cells) with SAR1B knockdown to study proliferation, apoptosis, and signaling pathways .

  • Tissue-specific knockout models: Including hepatocyte-specific deletion of Sar1b, which is compatible with survival though resulting in hypocholesterolemia .

These models provide valuable tools for understanding SAR1B function in normal physiology and disease states, though researchers should note that homozygous deletion or mutation of Sar1b in mice appears to cause embryonic lethality .

What methodologies are most effective for studying the functional overlap between SAR1A and SAR1B in vivo?

To effectively study the functional overlap between SAR1A and SAR1B paralogs in vivo, researchers should consider:

  • Gene replacement strategies: Engineering mice with the Sar1a coding sequence replacing that of Sar1b at the endogenous Sar1b locus has proven effective. This approach demonstrated that SAR1A expression can fully rescue the lethality of Sar1b deficiency, establishing functional overlap between the paralogs .

  • Complementation assays: Adenovirus-mediated overexpression of either SAR1A or SAR1B can be used to test functional rescue in tissue-specific knockouts, such as hepatocyte-specific Sar1b deletion models that exhibit hypocholesterolemia .

  • Transcriptomic analysis: Examining differential expression patterns of SAR1A and SAR1B across tissues to identify contexts of subfunctionalization .

  • Protein-protein interaction studies: Investigating whether SAR1A and SAR1B interact with the same or different subsets of COPII components and cargo proteins.

When interpreting results, researchers should consider that while these paralogs exhibit ~90% amino acid sequence identity, their distinct functions may arise from transcriptional subfunctionalization rather than protein functional divergence .

How should researchers approach the experimental design for studying SAR1B in cancer progression?

When designing experiments to study SAR1B in cancer progression, researchers should consider:

Researchers should employ multiple cell lines and validation techniques to ensure robust findings, as SAR1B functions may vary across cancer types and genetic backgrounds.

What are the methodological considerations when using CRISPR-Cas9 technology to study SAR1B function?

When using CRISPR-Cas9 technology to study SAR1B function, researchers should consider:

  • Design strategy:

    • For gene knockouts: Target early exons (e.g., exon 2) to ensure complete loss of function

    • For point mutations: Design strategies that closely mimic human disease mutations (e.g., p.D137N)

    • For gene replacements: Careful design to maintain endogenous regulatory elements

  • Genetic background considerations:

    • Test multiple genetic backgrounds as some mouse strains may be more resistant to phenotypic effects (e.g., C57BL/6 vs. 129/sv)

    • Published studies found that neither background was capable of producing viable Sar1b homozygous knockouts

  • Verification approaches:

    • Confirm mutations at DNA level by sequencing

    • Verify altered expression at mRNA level using RT-qPCR and the 2^(-ΔΔCT) calculation method

    • Confirm protein expression changes by Western blotting

  • Phenotyping timeline:

    • For SAR1B, examine embryos at multiple developmental stages (e.g., E9.5, E13.5, E18.5) to identify when lethality occurs

    • For heterozygotes, assess phenotypes under different metabolic challenges (e.g., high-fat diet)

  • Statistical analysis:

    • Use appropriate statistical approaches depending on experimental design

    • For embryonic experiments, one-way ANOVA with Tukey post hoc test

    • For adult mice with multiple variables (e.g., diet and genetic background), two-way ANOVA with Dunnett's post hoc test

What statistical approaches are most appropriate for analyzing complex SAR1B experimental designs?

When analyzing complex experimental designs involving SAR1B, researchers should consider:

  • Random effects models for nested designs:

    • Many SAR1B experiments involve multiple levels of nesting or crossing (e.g., treatments, environments, genotypes)

    • Random effects models can accommodate complex experimental designs with potential interactions between variables

  • Handling potential interactions:

    • Analyze potential block-treatment interactions that may affect interpretation

    • Consider the possibility that treatment effects may differ between experimental blocks

    • For example, in heterozygous SAR1B models, effects of different diets may vary across genetic backgrounds

  • Appropriate error term selection:

    • Carefully identify appropriate error terms for each main effect and interaction

    • Models should include terms that capture how effects vary between experimental units

    • Use Satterthwaite approximations for degrees of freedom when variance components differ

  • For gene expression data:

    • Use the 2^(-ΔΔCT) calculation method for RT-qPCR analysis

    • Employ appropriate normalization strategies with stable reference genes

  • Data presentation:

    • Present results as means ± SEM with significance typically considered at P < 0.05

    • Use GraphPad Prism or similar software for statistical analyses and graphical representation

The complexity of SAR1B biology often requires sophisticated statistical approaches to properly interpret experimental results, particularly when examining interactions between genotype, environment, and treatments.

How might findings on SAR1A and SAR1B functional overlap inform therapeutic approaches for chylomicron retention disease?

Research demonstrating functional overlap between SAR1A and SAR1B suggests potential therapeutic avenues for chylomicron retention disease (CMRD):

  • Therapeutic upregulation of SAR1A: Studies in mice show that expression of SAR1A in place of SAR1B can fully rescue the lethality of Sar1b deficiency, demonstrating high functional overlap between these paralogs . This suggests that therapeutic strategies aimed at upregulating SAR1A expression in patients with CMRD could potentially compensate for SAR1B deficiency .

  • Mechanism-based approach: The observation that transcriptional subfunctionalization, rather than protein functional divergence, is the primary evolutionary force maintaining both paralogs in mammals provides a mechanistic basis for therapeutic intervention . Therapeutic approaches could focus on:

    • Enhancing SAR1A transcription in tissues where SAR1B is normally predominant

    • Modifying regulatory elements to drive SAR1A expression in SAR1B-deficient tissues

    • Developing small molecules that enhance residual SAR1B function or SAR1A activity

  • Gene therapy considerations: The rescue of hypocholesterolemia by adenovirus-mediated overexpression of either SAR1A or SAR1B in hepatocyte-specific Sar1b knockout mice suggests that hepatic gene therapy approaches could be effective for addressing certain aspects of CMRD .

These findings provide strong evidence that therapeutic approaches targeting SAR1A upregulation could be leveraged to treat CMRD, offering hope for patients with this rare genetic disorder.

What methodological challenges exist in translating findings from SAR1B mouse models to human applications?

Translating findings from SAR1B mouse models to human applications presents several methodological challenges:

  • Differences in phenotype severity:

    • Homozygous deletion or mutation of Sar1b in mice causes embryonic lethality

    • In humans, biallelic SAR1B mutations cause CMRD but are compatible with survival

    • This discrepancy suggests potential species-specific differences in SAR1B function or compensatory mechanisms

  • Tissue-specific considerations:

    • Mouse models reveal that hepatocyte-specific deletion of Sar1b is compatible with survival

    • Translating these findings requires understanding tissue-specific functions of SAR1B in humans

    • Careful consideration of tissue-specific expression patterns and functions across species is necessary

  • Metabolic differences between species:

    • Mice and humans differ in lipid metabolism pathways

    • Heterozygous Sar1b mice show altered lipid homeostasis with increased fatty acid β-oxidation and diminished lipogenesis

    • Human-relevant endpoints and biomarkers must be established for proper translation

  • Experimental design considerations:

    • Mouse experiments often use highly controlled environments and genetic backgrounds

    • Human populations have greater genetic diversity and environmental variability

    • Statistical approaches must account for these differences when extrapolating findings

  • Model validation requirements:

    • Multiple mouse models (deletion, point mutation, tissue-specific knockout) provide complementary information

    • Validation in human cellular systems is necessary before clinical translation

    • Considering embryonic development effects is crucial given the lethality in mouse models

Addressing these challenges requires integrated approaches that combine findings from multiple model systems and careful validation in human-derived experimental systems.

How can researchers reconcile conflicting data regarding SAR1B function in different contexts?

When faced with conflicting data regarding SAR1B function across different experimental contexts, researchers should:

  • Consider context-dependent functions:

    • SAR1B functions in vesicular transport but has context-specific roles in different tissues and developmental stages

    • Embryonic lethality in homozygous Sar1b knockout mice versus viable tissue-specific knockouts illustrates this context-dependency

    • In colorectal cancer, SAR1B exhibits pro-proliferative functions , while in normal intestinal cells it regulates lipid metabolism

  • Evaluate methodological differences:

    • Different knockout/knockdown strategies may have varying efficiencies and off-target effects

    • Complete gene deletion versus point mutation models may reveal different aspects of protein function

    • When comparing studies, consider whether the approach targets expression level, protein function, or specific domains

  • Analyze stage-specific effects:

    • SAR1B function may differ during embryonic development versus adult physiology

    • The presence of homozygous Sar1b-deficient embryos at multiple stages (E9.5, E13.5, E18.5) despite eventual lethality suggests stage-specific requirements

  • Use statistical approaches for complex designs:

    • Apply random effects models that account for experimental design complexity

    • Consider potential interactions between variables (e.g., genotype and environmental conditions)

    • Use appropriate error terms and statistical tests based on experimental design

  • Integrate findings across models:

    • Combine evidence from multiple experimental systems (in vitro, in vivo, different genetic backgrounds)

    • Weigh evidence based on methodological robustness and reproducibility

    • Consider evolutionary conservation and paralogue complementation (SAR1A/SAR1B)

By systematically evaluating conflicting data through these approaches, researchers can develop more comprehensive and nuanced understanding of SAR1B function.

What approaches should be used to analyze the evolutionary and functional relationship between SAR1A and SAR1B?

To analyze the evolutionary and functional relationship between SAR1A and SAR1B effectively, researchers should employ:

  • Sequence-based evolutionary analysis:

    • Comparative sequence analysis across species to determine when gene duplication occurred

    • Examination of selection pressures on SAR1A and SAR1B since duplication

    • Analysis of conserved domains versus divergent regions (despite ~90% amino acid sequence identity)

  • Expression pattern analysis:

    • Comparative tissue-specific expression profiling of SAR1A and SAR1B

    • Analysis of developmental expression patterns

    • Investigation of tissue-specific transcriptional regulation mechanisms

  • Functional complementation studies:

    • Gene replacement experiments (as demonstrated with Sar1a coding sequence replacing Sar1b)

    • Cross-species complementation to determine functional conservation

    • Domain-swapping experiments to identify regions responsible for any functional differences

  • Interaction network mapping:

    • Comparative protein-protein interaction studies for SAR1A and SAR1B

    • Analysis of differential binding partners that might explain functional differences

    • Structural studies of protein complexes formed by each paralogue

  • Integrated evolutionary framework:

    • Test the subfunctionalization hypothesis suggested by current data

    • Determine whether transcriptional regulation rather than protein function drives evolutionary maintenance of both genes

    • Analyze cis-regulatory elements that might explain expression pattern differences

The evidence that expression of SAR1A in place of SAR1B fully rescues Sar1b deficiency lethality in mice strongly supports that transcriptional subfunctionalization, rather than protein functional divergence, is the primary evolutionary force maintaining both paralogs in mammals .

What advanced imaging techniques are most suitable for studying SAR1B-mediated vesicular transport?

Advanced imaging techniques for studying SAR1B-mediated vesicular transport include:

  • Live-cell imaging approaches:

    • Fluorescently tagged SAR1B (GFP, mCherry) for real-time visualization of dynamics

    • Photoactivatable or photoconvertible tags to track specific vesicle populations

    • FRAP (Fluorescence Recovery After Photobleaching) to measure protein mobility and exchange rates

  • Super-resolution microscopy:

    • STED (Stimulated Emission Depletion) microscopy to resolve individual COPII vesicles beyond the diffraction limit

    • STORM/PALM techniques for nanoscale localization of SAR1B and interacting partners

    • SIM (Structured Illumination Microscopy) for improved resolution of vesicular structures

  • Correlative light and electron microscopy (CLEM):

    • Combines fluorescence imaging of SAR1B with ultrastructural analysis by electron microscopy

    • Particularly valuable for studying the morphology of vesicles and their cargo in relation to SAR1B localization

    • Can reveal structural defects in vesicle formation in disease models

  • Functional imaging approaches:

    • FRET/FLIM to study SAR1B interactions with other COPII components

    • Cargo-specific transport assays using spectral variants of fluorescent proteins

    • Optogenetic approaches to spatiotemporally control SAR1B activity

  • Intravital imaging in model organisms:

    • Two-photon microscopy for deeper tissue imaging in live animals

    • Light-sheet microscopy for rapid volumetric imaging with reduced phototoxicity

    • Particularly useful for studying dynamics in developing embryos, where SAR1B function appears critical

These techniques can be combined with genetic manipulation strategies (CRISPR-Cas9) to study both wild-type and mutant forms of SAR1B in appropriate cellular contexts.

What biochemical assays can best characterize SAR1B GTPase activity and its relevance to disease states?

To characterize SAR1B GTPase activity and its relevance to disease states, researchers should consider:

  • GTP binding and hydrolysis assays:

    • Radioactive [γ-³²P]GTP or [α-³²P]GTP assays to measure GTP binding and hydrolysis rates

    • Fluorescence-based assays using mant-GTP (N-methylanthraniloyl-GTP) for real-time monitoring

    • Comparison of wild-type SAR1B versus disease-associated mutants (e.g., D137N)

  • Structural and conformational studies:

    • X-ray crystallography to determine structural changes upon GTP/GDP binding

    • Hydrogen-deuterium exchange mass spectrometry to analyze conformational dynamics

    • NMR spectroscopy to characterize protein-nucleotide interactions

  • Membrane interaction assays:

    • Liposome binding assays to measure SAR1B association with membranes

    • Surface plasmon resonance to quantify binding kinetics

    • Microscale thermophoresis to study interactions with membrane lipids and proteins

  • COPII assembly assays:

    • In vitro reconstitution of COPII vesicle budding with purified components

    • Quantification of SAR1B-dependent recruitment of Sec23/24 and Sec13/31

    • Electron microscopy to visualize COPII coat assembly structures

  • Disease-relevant functional readouts:

    • Chylomicron secretion assays in intestinal cell models

    • Lipoprotein particle analysis by gradient ultracentrifugation

    • Apolipoprotein B trafficking assays in cellular models of CMRD

These biochemical approaches should be combined with cellular assays to establish the relationship between biochemical properties and physiological functions, particularly in the context of disease-associated mutations.

Product Science Overview

Introduction

GTP-Binding Protein SAR1B, also known as Secretion Associated Ras Related GTPase 1B, is a small GTPase that plays a crucial role in the transport of proteins from the endoplasmic reticulum (ER) to the Golgi apparatus. This protein is part of the COPII coat complex, which is essential for vesicle-mediated transport within cells.

Gene and Protein Structure

The SAR1B gene is located on chromosome 5 at the cytoband q31.1 and spans approximately 48,122 base pairs. It encodes a protein that consists of 198 amino acids with a molecular weight of around 22 kDa . The protein functions as a homodimer and is activated by the guanine nucleotide exchange factor PREB .

Function

SAR1B cycles between an active GTP-bound state and an inactive GDP-bound state. In its active form, SAR1B inserts into the ER membrane, where it recruits other components of the COPII coat complex . This complex is responsible for the sorting of cargo proteins and the deformation and budding of membranes into vesicles destined for the Golgi apparatus .

Biological Significance

The proper functioning of SAR1B is critical for maintaining cellular homeostasis and efficient protein trafficking. Defects in the SAR1B gene can lead to Chylomicron Retention Disease (CMRD), also known as Anderson Disease. This condition is characterized by the inability to properly secrete chylomicrons, leading to fat malabsorption and other related symptoms .

Pathways and Interactions

SAR1B is involved in several biological pathways, including the SARS-CoV-2 infection pathway and the trafficking of the cystic fibrosis transmembrane conductance regulator (CFTR) protein . It also synergizes with the cargo receptor SURF4 to mediate the export of lipoproteins from the ER, thereby regulating lipid homeostasis .

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.