ARSA Mouse, Active

Arylsulfatase A Mouse Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Introduction to ARSA Mouse, Active

Arylsulfatase A (ARSA) Mouse, Active, is a recombinant enzyme produced in Sf9 cells, a line derived from the fall armyworm Spodoptera frugiperda. This enzyme is crucial for the degradation of sulfatides, which are important components of the myelin sheath in the nervous system. ARSA deficiency leads to metachromatic leukodystrophy (MLD), a severe neurological disorder characterized by progressive demyelination and accumulation of sulfatides in the central and peripheral nervous systems .

Key Features of ARSA Mouse, Active:

  • Molecular Composition: It is a single, glycosylated polypeptide chain containing 498 amino acids (18-506) with a molecular mass of approximately 53.2 kDa .

  • Purification: The enzyme is purified using proprietary chromatographic techniques and is formulated in phosphate-buffered saline (PBS) with 10% glycerol .

  • Functionality: ARSA hydrolyzes cerebroside sulfate to cerebroside and sulfate, a critical step in the lysosomal degradation pathway .

Role in Metachromatic Leukodystrophy (MLD)

ARSA plays a pivotal role in preventing the accumulation of sulfatides, which is a hallmark of MLD. In ARSA-deficient mice, sulfatide accumulation leads to severe neurological symptoms, making these models ideal for studying MLD and testing therapeutic interventions .

Gene Therapy Approaches

Recent studies have explored gene therapy as a potential treatment for MLD. Adeno-associated virus (AAV) vectors encoding ARSA have been used to restore enzyme activity in ARSA-deficient mice. These vectors, such as AAV9 and AAVHSC15, have shown promise in correcting sulfatide accumulation and improving neurological symptoms .

Therapeutic Outcomes

  • AAV9/ARSA: Intrathecal administration of AAV9/ARSA vectors has been effective in treating MLD model mice by reducing sulfatide storage in the brain and spinal cord .

  • HSC15/ARSA: This vector has demonstrated superior efficacy compared to AAV9/ARSA in terms of ARSA activity and motor function improvement in treated mice .

Gene Therapy Outcomes

VectorAdministration RouteEfficacy
AAV9/ARSAIntrathecalEffective in reducing sulfatide storage in CNS
HSC15/ARSAIntravenousSuperior ARSA activity and motor function improvement

Product Specs

Introduction
Arylsulfatase A, also known as ARSA, plays a crucial role in breaking down cerebroside sulfate into cerebroside and sulfate. This enzyme's activity is hindered by phosphate, which forms a covalent bond with the active site's 3-oxoalanine. Genetic defects affecting the ARSA gene lead to metachromatic leukodystrophy (MLD). MLD is a progressive neurological disorder characterized by the deterioration of the myelin sheath, resulting in a range of neurological issues and ultimately death.
Description

Recombinant ARSA Mouse protein, expressed in Sf9 cells, is a single, glycosylated polypeptide chain consisting of 498 amino acids (positions 18-506), with a molecular mass of 53.2 kDa. It's important to note that on SDS-PAGE, the apparent molecular size might range from 50 to 70 kDa. The ARSA Mouse protein includes a 9-amino acid His-Tag fused at its C-terminus and is purified using proprietary chromatographic techniques.

Physical Appearance
The product appears as a clear solution that has been sterilized through filtration.
Formulation

The ARSA protein solution is provided at a concentration of 0.5 mg/ml. It is formulated in a solution of Phosphate Buffered Saline (pH 7.4) containing 10% glycerol.

Stability
For short-term storage (2-4 weeks), keep the product refrigerated at 4°C. For extended storage, freeze the product at -20°C. Adding a carrier protein such as HSA or BSA (0.1%) is advised for long-term storage. It's essential to avoid repeated freeze-thaw cycles.
Purity
The purity of the ARSA protein is determined to be greater than 95% through SDS-PAGE analysis.
Synonyms

Arylsulfatase A, Arsa, As-2, AS-A, As2, ASA, AW212749, TISP73.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

ADPSPPNILL IFADDLGYGD LGSYGHPSST TPNLDQLAEG GLRFTDFYVP VSLCTPSRAA LLTGRLPVRSGMYPGVLGPS SQGGLPLEEVTLAEVLAARG YLTGMAGKWH LGVGPEGAFL PPHQGFHRFL GIPYSHDQGP CQNLTCFPPD IPCKGGCDQG LVPIPLLANL TVEAQPPWLPGLEARYVSFS RDLMADAQRQ GRPFFLYYAS HHTHYPQFSG QSFTKRSGRG PFGDSLMELD GAVGALMTTV GDLGLLEETL VIFTADNGPELMRMSNGGCSGLLRCGKGTT FEGGVREPAL VYWPGHITPG VTHELASSLD LLPTLAALTG APLPNVTLDG VDISPLLLGT GKSPRKSVFFYPPYPDEIHG VFAVRNGKYK AHFFTQGSAH SDTTSDPACH AANRLTAHEP PLLYDLSQDP GENYNVLESI EGVSPEALQA LKHIQLLKAQYDAAMTFGPS QIAKGEDPAL QICCQPSCTP HPVCCHCPGS QSHHHHHH.

Q&A

What is the established mouse model for MLD research?

The well-established MLD mouse model was originally generated by the Gieselman laboratory and has been used in numerous investigational therapies for MLD. This model, referred to as Arsa KO (Arsa tm1Gies) mice, has been rederived on a 129S6/SvEvTac background. Genotyping is typically performed using real-time PCR through services like Transnetyx . These mice develop sulfatide accumulation and brain pathology, making them suitable for studying therapeutic interventions for MLD.

What are the typical housing and handling considerations for ARSA mouse models?

ARSA mouse models require standard housing in accordance with Institutional Animal Care and Use Committees (IACUCs). In published studies, housing and breeding were conducted in accordance with IACUCs at research institutions like Homology Medicines and Taconic Biosciences . For experimentation, all procedures should be performed in accordance with established IACUC protocols. When designing studies, it's important to include equal divisions of males and females to evaluate treatment efficacy across both genders .

What are the optimal AAV vector designs for ARSA gene therapy in mouse models?

Two key AAV vector designs have shown promise in ARSA mouse model research:

  • HSC15/ARSA: This is a recombinant version of naturally occurring AAV serotype 15 (rAAVHSC15) expressing human ARSA cDNA that is codon-optimized under the control of a ubiquitous CAG promoter, followed by the SV40 early polyadenylation signal. The vector genome (3949 bp) is packaged in the rAAVHSC15 capsid, a natural Clade F AAV variant isolated from CD34+ human peripheral blood stem cells .

  • AAVPHP.eB-hARSA-HA: This vector drives expression of human ARSA cDNA under the control of the cytomegalovirus/β-actin hybrid (CAG) promoter. The addition of an HA tag facilitates detection and tracking of the protein expression .

Both vectors have demonstrated efficacy in targeting the central nervous system, with AAVPHP.eB showing particularly strong CNS tropism after intravenous administration.

What is the optimal administration route and dosing regimen for gene therapy in ARSA mouse models?

Research has evaluated different routes of administration (ROA) including intrathecal and intravenous approaches. For intravenous delivery:

  • AAVPHP.eB-hARSA-HA: Effective dose of 5×10¹¹ vg total, administered via retro-orbital delivery. Retro-orbital administration was preferred over tail vein injection based on preliminary results demonstrating better efficiency to target CNS and decreased liver transduction .

  • HSC15/ARSA: Intravenous administration as a slow 60-second bolus injection at 5 ml/kg has shown efficacy in restoring ARSA distribution .

The injected dose should be determined according to preliminary dose-ranging studies and evaluation of transduction efficacy. Treatment timing is critical - studies have shown that administration to 6-month-old mice with established pathology can still yield significant improvements within 3 months of treatment .

How do treatment outcomes in ARSA mouse models compare between different gene therapy approaches?

Studies comparing different gene therapy approaches have yielded the following comparative data:

VectorCNS TransductionPeripheral TransductionARSA ActivityTreatment Outcome
HSC15/ARSASignificant increases in brain ARSA activity, transcript levels, and vector genomes compared to AAV9/ARSARestores endogenous murine biodistributionDurability established up to 52 weeks in adult miceCorrected disease biomarkers and ameliorated motor deficits
AAVPHP.eB-hARSA-HAHigh CNS transduction (1.2-2.7 VGC in brain regions, 1.6 VGC in spinal cord)Low peripheral transduction (<0.05 VGC)Over-activity in cortex, pons, cerebellum, and spinal cordCorrection of brain and spinal cord sulfatide storage; improvement of astrogliosis and microgliosis

Both approaches demonstrated significant therapeutic benefit, with HSC15/ARSA showing advantages in broader biodistribution and AAVrh10.AAVPHP.eB-hARSA-HA showing stronger CNS-specific targeting.

What are the most reliable methods for measuring ARSA activity in mouse tissues?

The artificial p-nitrocatechol sulfate (pNCS) substrate assay is the standard method for measuring ARSA activity in mouse tissues. The procedure involves:

  • Tissue homogenization in lysis buffer (100 mM Trizma base, 150 mM NaCl, 0.3% Triton; pH 7)

  • Incubation on ice for 30 minutes followed by centrifugation

  • Collection of supernatant for protein content determination using bicinchoninic acid (BCA) protein assay

  • ARSA activity measurement using the pNCS substrate assay

  • Results expressed as nanomoles of 4-nitrocatechol (4NC) per hour per milligram of protein

For human ARSA quantification specifically, an indirect sandwich ELISA can be used with specific antibodies, with results expressed as nanograms of hARSA per milligram of protein .

What are the optimal methods for assessing biodistribution of ARSA gene therapy vectors?

Biodistribution of ARSA gene therapy vectors can be assessed using quantitative PCR (qPCR) to measure vector genome copies in various tissues. The methodology includes:

  • DNA extraction from tissue homogenates using appropriate kits (e.g., QIAsymphony DSP DNA mini kit)

  • Amplification of vector-specific sequences by qPCR using primers and probes for the codon-optimized ARSA sequence

  • Quantification relative to a standard curve generated from known quantities of plasmid containing the ARSA sequence

  • Normalization to cell number using housekeeping genes (e.g., mouse Pah gene)

  • Expression of results as vector genome copies per cell (VGC)

For comprehensive biodistribution studies, it's important to assess multiple CNS regions (cortex, striatum, cerebellum, pons, rest of brain, spinal cord) and peripheral organs.

What tissue preparation techniques are most effective for analyzing ARSA expression and localization?

For comprehensive analysis of ARSA expression and localization, the following tissue preparation techniques are recommended:

  • Perfusion: Intracardiac perfusion with phosphate-buffered saline (PBS)

  • Tissue collection and processing:

    • For molecular analyses: Dissect brain structures, freeze in liquid nitrogen, and store at -80°C

    • For protein/DNA extraction: Crush tissue in liquid nitrogen and divide into equal parts

    • For histological analyses: Post-fix tissues overnight in 4% paraformaldehyde/PBS, rinse in PBS, and cryoprotect in 30% sucrose/PBS

  • Sectioning: Embed in OCT compound and section at appropriate thickness (14-μm for brain/spinal cord, 4-μm for peripheral tissues)

For localization studies, immunofluorescence using anti-ARSA antibodies co-stained with cellular markers (e.g., anti-Lamp1 for lysosomal localization) is effective for confirming proper targeting of the enzyme to lysosomes .

How can researchers assess functional improvement in treated ARSA mouse models?

Assessment of functional improvement in treated ARSA mouse models should employ a multi-parameter approach:

  • Biochemical markers:

    • ARSA enzyme activity in CNS and peripheral tissues

    • Sulfatide levels in affected tissues

    • Other biomarkers that correlate with disease severity

  • Histopathological analysis:

    • Reduction in sulfatide storage

    • Improvement in astrogliosis and microgliosis

    • Myelin integrity assessment

  • Behavioral testing:

    • Motor function assessment

    • Cognitive function tests when applicable

Studies have demonstrated that levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit can be defined, providing important translational metrics . Triangulating multiple endpoints increases the potential for translational success to higher species .

What criteria determine successful transduction and therapeutic efficacy in ARSA mouse models?

Successful transduction and therapeutic efficacy in ARSA mouse models can be determined by the following criteria:

  • Vector transduction:

    • Vector genome copies per cell (VGC) in target tissues

    • Expression of recombinant ARSA protein

    • Proper lysosomal localization of ARSA enzyme

  • Therapeutic efficacy:

    • Restoration of ARSA enzyme activity to levels that mimic endogenous biodistribution

    • Correction of disease-specific biomarkers

    • Amelioration of histopathological features (sulfatide accumulation, astrogliosis, microgliosis)

    • Improvement in functional outcomes (motor performance)

Research has shown that intravenous administration of HSC15/ARSA restored the endogenous murine biodistribution of the enzyme, and overexpression of ARSA corrected disease biomarkers and ameliorated motor deficits in Arsa KO mice . Similarly, AAVPHP.eB-hARSA-HA resulted in correction of brain and spinal cord sulfatide storage and improvement of astrogliosis and microgliosis within 3 months of treatment .

How does gene therapy efficacy in mouse models translate to potential human applications?

Translation from mouse models to human applications requires careful consideration of several factors:

  • Biodistribution differences:

    • Studies have shown that ARSA gene therapy vectors can cross blood-nerve, blood-spinal, and blood-brain barriers in non-human primates, suggesting potential for human translation

    • The biodistribution profile in mice should be compared with that observed in higher species

  • Dose scaling:

    • Vector doses that demonstrate efficacy in mice need to be appropriately scaled for human applications

    • Allometric scaling principles should be applied, considering differences in body weight, blood volume, and target tissue mass

  • Timing considerations:

    • In mice, treatment at 6 months of age (with established pathology) was still effective

    • This suggests potential for therapeutic benefit even after symptom onset in humans

  • Biomarker correlation:

    • Identification of biomarkers that correlate with functional improvement in mice provides translational metrics for human studies

    • The levels of ARSA activity required for clinical benefit in different tissues need to be established

Studies have demonstrated that intravenous HSC15/ARSA-mediated gene therapy restored ARSA enzyme distribution in a fashion that mimics endogenous biodistribution, which is considered ideal for addressing both CNS and peripheral manifestations of MLD .

What are the most promising next steps for optimizing ARSA gene therapy in mouse models?

The most promising directions for optimizing ARSA gene therapy in mouse models include:

  • Further refinement of vector design:

    • Exploration of novel capsid variants with enhanced CNS tropism

    • Optimization of promoter and regulatory elements for cell type-specific expression

    • Development of self-regulating expression systems to prevent potential toxicity from overexpression

  • Combination therapeutic approaches:

    • Evaluation of combined gene therapy with enzyme replacement therapy

    • Assessment of adjunctive treatments targeting inflammation or other disease mechanisms

  • Long-term efficacy and safety studies:

    • Extended follow-up periods to assess durability of treatment effect

    • Comprehensive safety assessment, particularly for vectors achieving high CNS transduction

  • Addressing treatment-resistant aspects of pathology:

    • Identification of disease features that respond poorly to current gene therapy approaches

    • Development of targeted strategies for these resistant pathological elements

Current research has already established durability of transgene expression in neonate and adult mice out to 12 and 52 weeks, respectively , providing a foundation for these future optimizations.

How can researchers better model late-stage MLD pathology in mouse models?

Modeling late-stage MLD pathology in mouse models presents challenges due to differences in disease progression between mice and humans. Approaches to address this include:

  • Development of more aggressive mouse models:

    • Generation of compound mutants that accelerate sulfatide accumulation

    • Creation of conditional knockout models allowing for temporal control of ARSA deficiency

  • Environmental or genetic acceleration of pathology:

    • Dietary interventions that increase sulfatide production

    • Secondary genetic modifications affecting myelin turnover or inflammation

  • Aging studies:

    • Extended aging of Arsa KO mice to allow for more advanced pathology development

    • Correlation of age-dependent changes with disease stages in humans

  • Ex vivo modeling:

    • Development of organoid or slice culture systems from ARSA-deficient mice that can be manipulated to accelerate pathological changes

These approaches would help to better recapitulate the rapid neurological degradation seen in the late infantile form of human MLD .

What evidence supports the translation of mouse ARSA gene therapy findings to non-human primates?

Several key findings support the translation of mouse ARSA gene therapy findings to non-human primates:

  • Cross-species vector functionality:

    • HSC15/ARSA demonstrated blood-nerve, blood-spinal, and blood-brain barrier crossing in healthy non-human primates

    • Presence of circulating ARSA enzyme activity was detected in the serum of treated non-human primates

  • Vector biodistribution similarities:

    • Intravenous administration of HSC15/ARSA demonstrated similar biodistribution patterns in mice and non-human primates

    • The ability of vectors to transduce CNS tissues after peripheral administration was conserved across species

  • ARSA protein expression and activity:

    • Recombinant human ARSA expressed in both mice and non-human primates showed appropriate subcellular localization and enzymatic activity

    • The cross-species compatibility of human ARSA suggests potential for clinical translation

  • Safety profile:

    • No adverse events were observed in mice injected with AAVPHP.eB vector, attesting to the safety of the procedure

    • Preliminary safety assessments in non-human primates support further development

These findings collectively support the potential translation of ARSA gene therapy approaches from mouse models to clinical applications for MLD patients.

What are the key differences between mouse models and human MLD that researchers must consider?

Researchers must consider several key differences between mouse models and human MLD:

  • Disease progression timeline:

    • Human late infantile MLD progresses rapidly with early onset neurologic manifestations

    • Mouse models show slower progression and less severe neurological symptoms

  • Anatomical and physiological differences:

    • Brain size, complexity, and white matter proportion differ significantly

    • Blood-brain barrier properties and cerebrospinal fluid dynamics vary between species

  • Immune system considerations:

    • Immune responses to viral vectors and transgene products may differ

    • Pre-existing immunity to AAV variants is more common in humans than in laboratory mice

  • Enzyme cross-reactivity:

    • There is 96% sequence homology at the amino acid level between human and cynomolgus monkey arylsulfatase-a

    • Mouse and human ARSA may have subtle differences in substrate specificity or activity

  • Treatment access considerations:

    • Vector distribution in the much larger human CNS presents challenges

    • Scaling up vector production for human dosing requires additional considerations

Understanding these differences is crucial for appropriate experimental design and interpretation of results when translating findings from mouse models to human applications.

How can researchers address variability in ARSA activity measurements across different tissue samples?

To address variability in ARSA activity measurements:

  • Standardized tissue collection:

    • Consistent perfusion protocols to remove blood contamination

    • Precise dissection of anatomical regions

    • Immediate flash freezing and consistent storage conditions

  • Homogenization optimization:

    • Standardized buffer composition (100 mM Trizma base, 150 mM NaCl, 0.3% Triton; pH 7)

    • Consistent tissue-to-buffer ratios

    • Uniform homogenization methods (time, speed, temperature)

  • Assay standardization:

    • Use of common reference standards across experiments

    • Performance of assays in triplicate

    • Inclusion of positive and negative controls in each assay

  • Data normalization:

    • Consistent protein determination methods (BCA assay)

    • Expression of results in standardized units (nanomoles of 4NC per hour per milligram of protein)

    • Consideration of alternative normalization strategies (per DNA content, per tissue weight)

  • Statistical approaches:

    • Accounting for biological variables (age, sex, weight)

    • Appropriate statistical tests for sample size

    • Consideration of outlier analysis methods

These approaches help minimize technical variability and improve the reliability of ARSA activity measurements across different experimental conditions.

What strategies can optimize vector delivery to ensure consistent CNS transduction in ARSA mouse models?

Strategies to optimize vector delivery for consistent CNS transduction include:

  • Injection technique refinement:

    • For retro-orbital delivery: proper anesthesia, appropriate needle gauge, correct anatomical positioning, and controlled injection speed

    • For intravenous delivery: consideration of slow bolus (60 seconds) versus continuous infusion (30 minutes)

  • Vector preparation:

    • Consistent production methods using triple plasmid transfection in HEK293 suspension cells

    • Purification via affinity chromatography followed by anion exchange chromatography

    • Formulation in isotonic, neutral pH buffer suitable for in vivo administration

    • Thorough quality control testing for each vector batch

  • Animal selection and preparation:

    • Screening for anti-AAV neutralizing antibodies prior to administration

    • Consistent age and weight of animals at time of injection

    • Standardized anesthesia protocols

  • Dose optimization:

    • Determination of optimal dose through dose-ranging studies

    • Consideration of vector dose per body weight versus total vector dose

    • Adjustment of injection volume based on animal size

  • Timing considerations:

    • Consistent time of day for injections to account for circadian variations

    • Age-appropriate treatment windows based on disease progression

Research has shown that retro-orbital administration may be preferred to tail vein injection based on better efficiency to target CNS and decreased liver transduction for some vectors .

How might single-cell analysis techniques enhance understanding of cell-type specific responses to ARSA gene therapy?

Single-cell analysis techniques offer several advantages for understanding cell-type specific responses to ARSA gene therapy:

  • Cell-type specific transduction patterns:

    • Single-cell RNA sequencing can identify which cell types are preferentially transduced by different AAV capsids

    • Spatial transcriptomics can map the distribution of vector genomes and transgene expression across brain regions

  • Differential therapeutic responses:

    • Single-cell proteomics can assess ARSA protein levels and post-translational modifications in specific cell populations

    • Single-nucleus ATAC-seq can identify epigenetic changes in response to treatment across cell types

  • Disease mechanism insights:

    • Single-cell metabolomics might detect cell-type specific changes in sulfatide metabolism

    • Integrated multi-omics approaches can correlate gene expression, protein levels, and metabolic changes at single-cell resolution

  • Therapeutic optimization opportunities:

    • Identification of resistant cell populations that maintain pathology despite treatment

    • Development of cell type-specific promoters or targeting strategies based on single-cell data

These approaches would extend beyond the current immunofluorescence co-localization studies that have demonstrated proper lysosomal localization of ARSA in treated animals .

What are the implications of long-term ARSA overexpression in the CNS of treated mice?

Long-term ARSA overexpression in the CNS raises several important considerations:

  • Potential benefits:

    • Studies have demonstrated durability of transgene expression in neonate and adult mice out to 12 and 52 weeks, respectively

    • Sustained ARSA overexpression may provide continuous protection against sulfatide accumulation

    • Higher enzyme levels may allow for cross-correction of untransduced cells through secretion and uptake mechanisms

  • Potential concerns:

    • Metabolic consequences of prolonged enzyme overexpression

    • Possible immune reactions to sustained high levels of transgene product

    • Potential disruption of lysosomal function or other cellular processes

  • Dose-dependent effects:

    • Identification of optimal therapeutic window for ARSA expression

    • Possibility of differential effects in different cell types or brain regions

  • Aging considerations:

    • Interaction between transgene expression and age-related changes in CNS

    • Long-term persistence of vector genomes with aging

Research has shown that overexpression of ARSA corrected disease biomarkers and ameliorated motor deficits in Arsa KO mice , but systematic evaluation of very long-term effects would provide important safety information for clinical translation.

What are the most critical knowledge gaps in ARSA mouse model research that need to be addressed?

Several critical knowledge gaps require focused research attention:

  • Mechanism of therapeutic action:

    • Detailed understanding of cellular and molecular events following ARSA restoration

    • Temporal sequence of repair processes in different CNS regions

    • Identification of reversible versus irreversible pathological changes

  • Predictive biomarkers:

    • Validation of biomarkers that reliably predict functional outcomes

    • Development of minimally invasive methods to monitor treatment response

    • Correlation between murine biomarkers and clinical markers in humans

  • Treatment window determination:

    • Precise definition of therapeutic windows at different disease stages

    • Comparison of early intervention versus rescue approaches

    • Strategies for treatment of advanced disease

  • Combination approaches:

    • Identification of complementary therapeutic strategies

    • Optimal timing and sequencing of combination treatments

    • Mechanistic understanding of synergistic effects

Addressing these knowledge gaps would significantly advance the field and improve the translational potential of ARSA mouse model research to human MLD treatment.

What standardization efforts would most benefit the ARSA mouse model research community?

Standardization in several key areas would significantly benefit ARSA mouse model research:

  • Animal model characterization:

    • Consistent phenotyping protocols for ARSA-deficient mice

    • Standardized age milestones for disease progression

    • Agreed-upon behavioral and functional assessment batteries

  • Treatment protocols:

    • Standardized vector production and quality control methods

    • Uniform administration procedures and dosing calculations

    • Common post-treatment evaluation timelines

  • Outcome measurements:

    • Consistent methods for ARSA activity determination

    • Standardized histopathological scoring systems

    • Validated behavioral testing protocols specific to MLD mouse models

  • Reporting standards:

    • Minimum information guidelines for publishing ARSA mouse model studies

    • Comprehensive data sharing initiatives

    • Central repositories for vector constructs and protocols

Product Science Overview

Definition and Classification

Arylsulfatase A (ARSA), also known as cerebroside-sulfatase, is an enzyme that hydrolyzes cerebroside sulfate into cerebroside and sulfate . In the context of recombinant proteins, Mouse Recombinant Arylsulfatase A refers to the ARSA enzyme produced using recombinant DNA technology in a mouse cell line .

Biological Properties and Functions

ARSA plays a crucial role in the degradation of sulfatides, which are essential components of the myelin sheath in the nervous system . The enzyme’s activity is vital 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) .

Mode of Action

The enzyme ARSA hydrolyzes the sulfate ester bond in cerebroside sulfate, converting it into cerebroside and sulfate . This reaction is essential for the catabolism of sulfatides in the lysosomes. The enzyme’s activity is measured by its ability to hydrolyze synthetic substrates such as 4-Nitrocatechol Sulfate (PNCS) .

Recombinant Production

Recombinant Mouse Arylsulfatase A is produced in mouse myeloma cell lines (NS0) or Sf9 Baculovirus cells . The recombinant protein typically includes a signal peptide and a mature chain, with a molecular mass of approximately 54 kDa . It is often tagged with a His-tag for purification purposes .

Regulatory Mechanisms

ARSA activity is regulated by various factors, including the presence of phosphate, which can inhibit the enzyme by forming a covalent bond with the active site . Additionally, the enzyme’s stability and activity can be influenced by the formulation and storage conditions .

Applications and Research

Recombinant Mouse Arylsulfatase A is used in various research applications, including studies on lysosomal storage disorders, enzyme replacement therapies, and the biochemical pathways involving sulfatides . Its activity and stability make it a valuable tool for understanding the molecular mechanisms underlying MLD and other related 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 2025 TheBiotek. All Rights Reserved.