ARSA Human, SF9

Arylsulfatase A Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Production and Purification

ARSA Human, SF9 is expressed using a Baculovirus-insect cell system, followed by proprietary chromatographic purification . The final formulation includes phosphate-buffered saline (pH 7.4) and 10% glycerol for stability . Unlike E. coli-derived ARSA (non-glycosylated, 54.3 kDa) , the Sf9-produced variant retains glycosylation, enhancing its structural and functional fidelity .

Enzyme Activity and Assays

ARSA activity is measured using synthetic substrates like p-nitrocatechol sulfate (PNCS) or 4-nitrocatechol sulfate. Key findings include:

  • Normal Activity: Healthy human neural cells exhibit 0.43–10.25 mU/mg protein .

  • MLD Patient Cells: Activity falls below detection limits (<0.25 mU/mg) .

  • Overexpression: ARSA-SF9 shows 140–1,792 mU/mg in transfected cells, surpassing physiological levels .

Table 2: ARSA Activity in Cell Models

Cell TypeARSA Activity (mU/mg protein)Source
Healthy Control (lt-NES)0.43 ± 0.05
MLD Patient (lt-NES)Undetectable
ARSA-Overexpressing (APCs)612.53 ± 15.15 (proliferating)
Recombinant ARSA (SF9)760.75 ± 15.62 (differentiated)

Disease Modeling

ARSA-SF9 is used to study MLD pathogenesis. Transplanting ARSA-overexpressing neural progenitor cells into ARSA⁻/⁻ mice reduced sulfatide deposits by 70–90% in adjacent brain regions .

Biochemical Characterization

Immortalized mesenchymal stromal cells (MSCs) from MLD patients serve as platforms to test ARSA variants. Seven novel pathogenic mutants exhibited <10% wild-type activity, confirming their role in MLD .

Therapeutic Development

Gene-corrected ARSA-SF9 demonstrates potential for enzyme replacement therapy. In vitro, transfected cells secreted up to 2.0 µg ARSA/mg protein, restoring sulfatide clearance .

Pathophysiological Relevance

ARSA deficiency causes sulfatide accumulation, leading to demyelination and neurological decline . The Sf9-produced enzyme’s high specific activity (~1,800 mU/mg post-differentiation) makes it a candidate for cross-correction strategies, where enzyme secretion by donor cells benefits neighboring host cells .

Stability and Formulation

  • Storage: Stable at -70°C for 12 months; 1 month at 4°C post-reconstitution .

  • Inhibitors: Phosphate ions covalently bind the active-site 3-oxoalanine, reversibly inhibiting activity .

Product Specs

Introduction
Arylsulfatase A, also called cerebroside-sulfatase, is an enzyme that breaks down sulfatides. Its primary target is cerebroside 3-sulfate, which it converts to cerebroside and sulfate. In humans, the ARSA gene is responsible for encoding this enzyme. Arylsulfatase A's activity can be inhibited by phosphate, which forms a covalent bond with the enzyme's active site at the 3-oxoalanine residue.
Description
Produced in Sf9 insect cells using a baculovirus expression system, ARSA is a single, glycosylated polypeptide chain. It consists of 498 amino acids (with a sequence spanning from amino acid 21 to 509) and has a molecular weight of 53.0 kDa. On SDS-PAGE, its apparent molecular size will be approximately 50-70 kDa. This ARSA protein is engineered with a 9 amino acid His tag at its C-terminus to facilitate purification, which is carried out using proprietary chromatographic methods.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The ARSA protein solution has a concentration of 0.25 mg/ml and is stored in a buffer consisting of Phosphate Buffered Saline at a pH of 7.4 with 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the ARSA protein can be stored at 4°C. For longer storage, it should be kept frozen at -20°C. Adding a carrier protein like HSA or BSA to a final concentration of 0.1% is recommended for long-term storage. Repeated freezing and thawing of the protein should be avoided.
Purity
The purity of the ARSA protein is greater than 95.0% as determined by SDS-PAGE analysis.
Biological Activity
The specific activity of this enzyme is greater than 2,500 pmol/min/µg. This activity is determined by measuring the rate at which the enzyme hydrolyzes 4-Nitrocatechol at a pH of 5.0 and a temperature of 37°C.
Synonyms

Arylsulfatase A, Cerebroside-Sulfatase, ASA, Metachromatic Leucodystrophy, MLD, EC 3.1.6.8.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPRPPNIVL IFADDLGYGD LGCYGHPSST TPNLDQLAAG GLRFTDFYVP VSLCTPSRAA LLTGRLPVRM GMYPGVLVPS SRGGLPLEEVTVAEVLAARG YLTGMAGKWH LGVGPEGAFL PPHQGFHRFL GIPYSHDQGP CQNLTCFPPA TPCDGGCDQG LVPIPLLANL SVEAQPPWLP GLEARYMAFA HDLMADAQRQ DRPFFLYYAS HHTHYPQFSG QSFAERSGRG PFGDSLMELD AAVGTLMTAI GDLGLLEETL VIFTADNGPETMRMSRGGCS GLLRCGKGTT YEGGVREPAL AFWPGHIAPG VTHELASSLD LLPTLAALAG APLPNVTLDG FDLSPLLLGT GKSPRQSLFF YPSYPDEVRG VFAVRTGKYK AHFFTQGSAH SDTTADPACH ASSSLTAHEP PLLYDLSKDP GENYNLLGGV AGATPEVLQA LKQLQLLKAQLDAAVTFGPS QVARGEDPAL QICCHPGCTP RPACCHCPDP HAHHHHHH.

Q&A

What is ARSA and what is its function in human metabolism?

ARSA (Arylsulfatase A) is a lysosomal enzyme responsible for breaking down sulfatides in humans. It specifically catalyzes the hydrolysis of cerebroside 3-sulfate into cerebroside and sulfate . The enzyme is encoded by the ARSA gene, and deficiencies in this enzyme lead to Metachromatic Leukodystrophy (MLD), a devastating lysosomal storage disorder characterized by sulfatide accumulation in the central and peripheral nervous systems.

For studying ARSA's metabolic role, researchers should employ multiple analytical approaches:

  • Enzyme activity assays using p-nitrocatechol sulfate as an artificial substrate

  • Mass spectrometry analysis of sulfatide levels in tissue samples

  • Immunofluorescence staining to visualize ARSA localization in cells

  • Comparative analysis between healthy and MLD patient-derived cells

Why is the Sf9 insect cell system preferred for recombinant ARSA production?

The Sf9 insect cell-baculovirus expression vector system (IC-BEVS) offers significant advantages for ARSA production:

  • High expression yields: The baculovirus system enables robust production of complex mammalian proteins

  • Post-translational modifications: Sf9 cells perform necessary glycosylation, though with different patterns than mammalian cells

  • Time and cost efficiency: IC-BEVS is more economical than mammalian expression systems

  • Scalability: The process can be readily scaled for larger production requirements

For implementing this system, researchers should:

  • Clone the ARSA gene into a baculovirus transfer vector

  • Generate recombinant baculovirus

  • Optimize infection parameters (MOI, timing) through transcriptomic analysis

  • Monitor protein expression and activity throughout the production process

What are the structural characteristics of ARSA produced in Sf9 cells?

ARSA produced in Sf9 baculovirus cells presents the following key structural features:

  • Single glycosylated polypeptide chain containing 498 amino acids (residues 21-509 of human ARSA)

  • Molecular mass of 53.0 kDa, though appearing at 50-70 kDa on SDS-PAGE due to glycosylation

  • Often expressed with a 9 amino acid His-tag at the C-terminus for purification purposes

To characterize Sf9-produced ARSA, researchers should employ:

  • SDS-PAGE for molecular weight determination

  • Western blotting with anti-ARSA antibodies

  • Glycosylation analysis using specialized staining or mass spectrometry

  • Enzymatic activity assays to confirm functional conformation

How is ARSA activity measured in laboratory settings?

The standard protocol for measuring ARSA activity utilizes p-nitrocatechol sulfate (PNCS) as an artificial substrate. The methodological approach is as follows:

  • Prepare assay buffer (50 mM NaOAc, 0.5 M NaCl, pH 4.5)

  • Dilute ARSA to an appropriate concentration (e.g., 20 μg/mL)

  • Prepare 2 mM PNCS substrate solution

  • Mix equal volumes of enzyme and substrate (75 μL each)

  • Incubate at 37°C for 1 hour

  • Stop reaction with 0.2 M NaOH

  • Measure absorbance at 510 nm

  • Calculate enzyme activity

For reference, ARSA activity levels vary significantly between normal and disease states:

Cell TypeConditionARSA Activity (mU/mg protein)
lt-NES cellsHealthy control0.43 ± 0.05
APCsHealthy control7.37 ± 0.04
lt-NES cellsMLD patientBelow detection limit
APCsMLD patientBelow detection limit
lt-NES cellsARSA overexpressing140.19 ± 10.59
APCsARSA overexpressing612.53 ± 15.15

Data adapted from neural cell populations study

What are the challenges in ensuring proper glycosylation of ARSA in Sf9 cells?

While Sf9 cells perform glycosylation, the patterns differ from mammalian cells, presenting several research challenges:

  • Sf9 cells produce primarily high-mannose N-glycans rather than complex mammalian-type glycans

  • Lack of proper mannose-6-phosphate residues may affect lysosomal targeting

  • Glycosylation differences can impact enzyme stability, activity, and in vivo half-life

To address these challenges, researchers should:

  • Perform comparative glycan analysis of Sf9-produced versus native ARSA using mass spectrometry

  • Evaluate the functional impact through activity assays under varying conditions (pH/temperature stability)

  • Consider genetic engineering of Sf9 cells to humanize their glycosylation machinery

  • Explore in vitro glycan modification of purified enzyme if needed for specific applications

How can researchers optimize ARSA expression levels in the Sf9 baculovirus system?

Optimizing ARSA expression requires systematic experimental approaches:

ParameterOptimization StrategyAnalytical Method
Vector designUse strong promoters (polyhedrin/p10); optimize codon usageWestern blot, activity assay
MOITest range (typically 0.1-10); lower MOI often yields better qualityCell viability, expression level
Harvest timingMonitor 24-72h post-infectionTime-course analysis of expression/activity
Cell densityOptimize initial density at infectionGrowth curves, expression yield
TemperatureTest reduced temperature during expression phaseProtein quality assessment

Additionally, transcriptome analysis of infected Sf9 cells can reveal:

  • Gene expression changes following baculovirus infection

  • Cellular pathways affected during protein production

  • Potential bottlenecks in recombinant protein expression

What methodologies effectively preserve ARSA activity during purification from Sf9 cultures?

Preserving ARSA activity during purification requires careful consideration of multiple factors:

  • Harvest optimization:

    • Determine optimal time post-infection (typically when viability begins declining)

    • Use gentle centrifugation for cell collection

  • Lysis protocol:

    • Test mild detergents versus mechanical disruption

    • Include protease inhibitors to prevent degradation

    • Maintain acidic pH to mimic lysosomal environment

  • Purification strategy:

    • For His-tagged ARSA, use immobilized metal affinity chromatography (IMAC)

    • Consider size exclusion chromatography as a polishing step

    • Monitor activity at each purification stage

  • Buffer optimization:

    • Test stability in various buffer systems (acetate, phosphate, Tris)

    • Evaluate pH range (4.0-5.5 typically optimal for lysosomal enzymes)

    • Assess effects of stabilizing additives (glycerol, reducing agents)

The purified enzyme should be characterized for:

  • Specific activity (units/mg protein)

  • Purity (SDS-PAGE, HPLC)

  • Stability under storage conditions

How do researchers compare ARSA produced in Sf9 cells to native human ARSA?

Comprehensive comparison requires multi-parameter analysis:

ParameterMethodologyKey Measurements
Enzyme kineticsSubstrate titration assaysKm, Vmax, kcat, substrate specificity
StabilityDifferential scanning fluorimetryThermal denaturation profiles, pH stability
GlycosylationMass spectrometry, lectin blottingGlycan composition, occupancy of glycosylation sites
Cellular uptakeFluorescent labeling, confocal microscopyInternalization rates, receptor dependence
Intracellular traffickingCo-localization studiesLysosomal targeting efficiency
Functional rescueMLD patient-derived cell modelsReduction in sulfatide accumulation

In experimental design, researchers should:

  • Use standardized assay conditions

  • Include appropriate reference standards

  • Perform statistical analysis to determine significant differences

  • Validate findings across multiple enzyme batches and cell types

What experimental approaches reveal ARSA's substrate interactions and inhibitor mechanisms?

Multiple complementary techniques provide insights into ARSA's molecular interactions:

  • Enzyme kinetics analysis:

    • Determine kinetic parameters with natural and artificial substrates

    • Perform inhibition studies to determine inhibition constants (Ki) and mechanisms

    • Analyze data using Lineweaver-Burk plots or non-linear regression

  • Binding studies:

    • Isothermal titration calorimetry (ITC) for thermodynamic parameters

    • Surface plasmon resonance (SPR) for association/dissociation kinetics

    • Fluorescence-based binding assays for high-throughput screening

  • Structural studies:

    • X-ray crystallography of ARSA-substrate/inhibitor complexes

    • Molecular docking and simulation studies

    • Site-directed mutagenesis of active site residues

Of particular interest is phosphate inhibition, which operates by forming a covalent bond with ARSA's active site 3-oxoalanine residue . This mechanism provides a model for designing competitive inhibitors for research applications.

How can ARSA overexpression models be developed using Sf9-produced enzyme?

The development of ARSA overexpression models involves several methodological considerations:

  • In vitro cellular models:

    • Add purified Sf9-produced ARSA to culture medium of neuronal or glial cells

    • Quantify cellular uptake through activity assays and immunofluorescence

    • Measure reduction in sulfatide accumulation in MLD patient-derived cells

  • Ex vivo tissue models:

    • Apply ARSA to brain or nerve tissue slices from MLD models

    • Assess enzyme penetration, cellular uptake, and metabolic effects

    • Measure changes in sulfatide content by mass spectrometry

  • In vivo models:

    • Administer Sf9-produced ARSA to ARSA-deficient animal models

    • Evaluate biodistribution, blood-brain barrier penetration, and half-life

    • Assess therapeutic efficacy through sulfatide measurements

A relevant example comes from a study where ARSA-overexpressing neural cells were transplanted into ARSA-deficient mice, resulting in significant reduction of sulfatide deposits in the surrounding tissue . Similar approaches could be developed using purified Sf9-produced ARSA.

What potential does Sf9-produced ARSA hold for enzyme replacement therapy in MLD?

Evaluating Sf9-produced ARSA for enzyme replacement therapy requires consideration of several factors:

  • Production advantages:

    • Higher yields and lower costs compared to mammalian systems

    • Scalable production process amenable to GMP requirements

  • Pharmacological considerations:

    • Comparison of pharmacokinetic profiles with mammalian-produced ARSA

    • Assessment of brain penetration following various administration routes

    • Evaluation of immune responses to insect cell glycosylation patterns

  • Preclinical evaluation:

    • Dose-response studies in MLD animal models

    • Long-term efficacy and safety assessment

    • Comparison with existing therapeutic approaches

Data from neural cell transplantation studies show that even local delivery of ARSA can reduce sulfatide deposits significantly in surrounding brain tissue , suggesting that properly delivered ARSA could potentially provide therapeutic benefit.

ARSA SourceAdvantagesLimitationsResearch Needs
Sf9-producedHigher yield, lower costDifferent glycosylationUptake efficiency, immunogenicity
Mammalian-producedNative-like glycosylationHigher production costOptimization of M6P content
Cell-based deliveryLocal production, long-termLimited distributionCell survival, integration

What methodological approaches maximize the utility of Sf9-produced ARSA in sulfatide metabolism research?

ARSA from Sf9 cells can be utilized in various experimental paradigms:

  • Cross-correction studies:

    • Add purified ARSA to medium of ARSA-deficient cells

    • Monitor uptake through mannose-6-phosphate or alternative receptors

    • Quantify sulfatide reduction using mass spectrometry

    • Assess downstream cellular effects (morphology, function, gene expression)

  • Mechanistic investigations:

    • Explore factors affecting enzyme uptake and activity

    • Study the role of saposins and other cofactors in ARSA function

    • Investigate cellular responses to varying degrees of ARSA activity

  • Screening applications:

    • Develop cell-based assays with Sf9-produced ARSA to identify:

      • Compounds enhancing enzyme stability or activity

      • Molecules improving cellular uptake or trafficking

      • Agents modulating sulfatide metabolism independently of ARSA

ELISA measurements reveal significant differences in ARSA concentrations between normal and disease states:

Cell TypeConditionIntracellular ARSA (μg/mg protein)Secreted ARSA (μg/mg protein)
lt-NES cellsHealthy0.003 ± 0.00040.05 ± 0.03
APCsHealthy0.01 ± 0.0250.24 ± 0.008
lt-NES cellsMLD patientBelow detectionBelow detection
APCsMLD patientBelow detection0.02 ± 0.02
lt-NES cellsARSA overexpressingNot reported1.54 ± 0.21
APCsARSA overexpressingNot reported2.0 ± 0.38

Data adapted from neural cell populations study

What translational challenges must researchers address when moving from Sf9-produced ARSA to clinical applications?

The translation of Sf9-produced ARSA to clinical applications presents several challenges requiring methodological solutions:

  • Biological barriers:

    • Blood-brain barrier penetration: Explore novel delivery strategies (nanoparticles, fusion proteins)

    • Half-life optimization: Investigate modifications to reduce clearance

    • Immunogenicity management: Develop protocols to monitor and mitigate immune responses

  • Manufacturing considerations:

    • Process scale-up while maintaining enzymatic quality

    • Development of consistent purification protocols

    • Implementation of appropriate quality control measures

    • Formulation optimization for stability and administration

  • Clinical development strategies:

    • Biomarker identification for monitoring treatment efficacy

    • Patient stratification based on mutation type and disease stage

    • Timing of intervention before irreversible neural damage occurs

    • Consideration of combination therapies (gene therapy, substrate reduction)

Transplantation studies of ARSA-expressing cells provide proof-of-concept that enzyme delivery can reduce sulfatide deposits in vivo , suggesting that purified enzyme could potentially achieve similar effects if delivery challenges are overcome.

Product Science Overview

Production and Structure

The recombinant human Arylsulfatase A (ARSA) is produced in Sf9 Baculovirus cells. The enzyme is a single, glycosylated polypeptide chain containing 498 amino acids, with a molecular mass of approximately 53.0 kDa . The recombinant ARSA is expressed with a 9 amino acid His tag at the C-terminus and is purified using proprietary chromatographic techniques .

Physical and Chemical Properties

The ARSA protein solution is formulated in Phosphate Buffered Saline (pH 7.4) with 10% glycerol . It is a sterile, filtered clear solution with a purity greater than 95.0% as determined by SDS-PAGE . The specific activity of ARSA is greater than 2,500 pmol/min/µg, defined as the amount of enzyme that hydrolyzes 4-Nitrocatechol at pH 5.0 at 37°C .

Stability and Storage

For short-term storage (2-4 weeks), ARSA should be kept at 4°C. For long-term storage, it is recommended to store the enzyme at -20°C with the addition of a carrier protein (0.1% HSA or BSA) to prevent multiple freeze-thaw cycles .

Biological Activity

ARSA plays a crucial role in the degradation of sulfatides, which are essential for maintaining the normal function of the nervous system. Deficiency in ARSA activity leads to the accumulation of sulfatides, causing a lysosomal storage disorder known as Metachromatic Leukodystrophy (MLD) . Recombinant human ARSA is in clinical development for the treatment of patients with MLD .

Research and Applications

Nonclinical comparability studies have been conducted to address manufacturing process changes in recombinant human ARSA. These studies have shown that changes in the manufacturing process did not affect the pharmacodynamic, pharmacokinetic, or safety profiles of the enzyme . Additionally, ARSA has been studied as a genetic modifier in Parkinson’s disease, indicating its potential role in neurodegenerative diseases .

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.