ASPA Human

Aspartoacylase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction and Overview of ASPA Human

ASPA Human refers to the human isoform of the enzyme aspartoacylase (EC 3.5.1.15), encoded by the ASPA gene. This zinc-dependent hydrolase catalyzes the cleavage of N-acetyl-L-aspartate (NAA) into aspartate and acetate, a critical metabolic process in the brain . Deficiencies in ASPA activity are directly linked to Canavan disease, a lethal neurodegenerative disorder . Recombinant forms of ASPA, such as those produced for research and therapeutic development, are also classified under this term .

Biological Function and Mechanism

ASP Human regulates NAA levels in the brain, maintaining white matter integrity. Its catalytic mechanism involves:

  1. Zinc Activation: Lowers the pKa of a water molecule, enabling nucleophilic attack on NAA .

  2. Deprotonation: Glu-178 deprotonates water, forming a tetrahedral intermediate stabilized by zinc and Arg-63 .

  3. Cleavage: Concerted breakdown of the intermediate releases acetate and aspartate .

NAA metabolism is hypothesized to supply acetate for myelin lipid synthesis and regulate osmotic balance .

Role in Canavan Disease

Over 80 mutations in ASPA cause Canavan disease, characterized by NAA accumulation and spongiform brain degeneration . Common pathogenic variants include:

MutationEffect on ActivityPopulation Prevalence
E285AReduces activity to 0.3%98% of Ashkenazi Jewish cases .
A305EReduces activity to ~10%40% of non-Jewish cases .

These mutations disrupt substrate binding or zinc coordination, impairing NAA hydrolysis . Elevated NAA levels in urine (>60× normal) serve as a diagnostic marker .

Recombinant ASPA Production and Applications

Recombinant ASPA is produced in heterologous systems for research and therapeutic studies:

Expression SystemAdvantagesChallenges
Escherichia coliHigh yield (e.g., 0.5 mg/mL) Forms inclusion bodies; requires refolding .
Pichia pastorisImproved stability; post-translational modifications .Lower expression yield .

The E. coli-derived enzyme (38.1 kDa, His-tagged) is stabilized in Tris-HCl buffer with glycerol and DTT .

Therapeutic Research and Clinical Trials

Gene therapy targeting ASPA deficiency shows promise:

  • BBP-812: An AAV9 vector delivering functional ASPA reduces NAA levels in cerebrospinal fluid (CSF), brain tissue, and urine .

    • Trial (CANaspire): Participants exhibited rapid NAA reduction post-treatment, correlating with myelination signals on MRI .

    • Safety: No treatment-related serious adverse events reported .

Product Specs

Introduction
Aspartoacylase, also known as ASPA, is a protein present in various tissues, including skeletal muscle, cerebral white matter, kidney, liver, and lungs. It functions as a homodimer, catalyzing the deacetylation of N-acetylaspartic acid (NAA) to produce L-aspartate and acetate.
Description
Recombinant human ASPA, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 313 amino acids (1-313) with a molecular weight of 35.7 kDa. The protein is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The ASPA solution is provided at a concentration of 1 mg/ml in a buffer containing 10% glycerol and phosphate-buffered saline (pH 7.4).
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein like 0.1% HSA or BSA is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of ASPA is greater than 90.0% as determined by SDS-PAGE analysis.
Synonyms

Aspartoacylase, Aminoacylase-2, ACY-2, ASPA, ACY2, ASP

Source
E.coli.
Amino Acid Sequence

MTSCHIAEEH IQKVAIFGGT HGNELTGVFL VKHWLENGAE IQRTGLEVKP FITNPRAVKK CTRYIDCDLN RIFDLENLGK KMSEDLPYEV RRAQEINHLF GPKDSEDSYD IIFDLHNTTS NMGCTLILED SRNNFLIQMF HYIKTSLAPL PCYVYLIEHP SLKYATTRSI AKYPVGIEVG PQPQGVLRAD ILDQMRKMIK HALDFIHHFN EGKEFPPCAI EVYKIIEKVD YPRDENGEIA AIIHPNLQDQ DWKPLHPGDP MFLTLDGKTI PLGGDCTVYP VFVNEAAYYE KKEAFAKTTK LTLNAKSIRC CLH

Q&A

What is the ASPA gene and what is its function in humans?

The ASPA gene encodes the aspartoacylase enzyme, which facilitates the hydrolysis of N-acetyl L-aspartate (NAA) into aspartic acid and acetate. This enzyme plays a crucial role in brain metabolism and myelin maintenance. The ASPA gene has been mapped to chromosome 17p13-ter and consists of six exons and five introns spanning approximately 29 kilobases . The enzyme functions as a zinc-dependent hydrolase with specific active site residues essential for catalytic activity. Proper ASPA function is critical for normal brain development and neurological function, as it contributes to myelin formation and white matter integrity.

What is the molecular structure of the human ASPA enzyme?

The human ASPA protein comprises 313 amino acids with an approximate molecular weight of 36 kilodaltons. The enzyme forms a dimer with zinc at the catalytic site . Homology modeling based on zinc-dependent carboxypeptidase A has revealed critical structural elements of ASPA. The key catalytic and substrate-binding residues include:

  • His21, Glu24, and His116: Coordinate zinc binding

  • Glu178: Functions as the general proton donor essential for substrate affinity and catalytic activity

  • Arg63 and Arg71: Involved in transition state stabilization and substrate carboxyl binding, respectively

These structural elements work together to enable the enzyme's function in hydrolyzing NAA, which is essential for normal neurological development.

How do mutations in the ASPA gene cause Canavan disease?

Canavan disease results from mutations in the ASPA gene that lead to loss or reduced activity of the aspartoacylase enzyme. Over 70 different mutations have been reported in the ASPA gene . These mutations disrupt the enzyme's ability to hydrolyze NAA, resulting in NAA accumulation in the brain, which leads to demyelination and spongy degeneration of white matter.

The disease severity typically correlates with the extent of enzyme dysfunction, with more severe mutations causing complete loss of enzyme activity resulting in typical (early-onset) Canavan disease. Less severe mutations that allow for residual enzyme activity may result in atypical (late-onset) forms of the disease with a more variable clinical course .

How do specific mutations affect ASPA enzyme structure and function?

Experimental studies have demonstrated that mutations in critical residues can profoundly impact ASPA function. Mutations affecting zinc-binding residues (H21G, E24D/G, H116G), the general proton donor (E178A), and substrate binding sites (R63N, R71N) result in enzyme proteins with undetectable ASPA activity despite normal protein expression levels .

For example, the novel homozygous ASPA variant c.532G>A causing a p.(Glu178Lys) substitution described in a recent case report targets the critical active site of the enzyme. Glu178 is essential for substrate affinity and catalytic activity, and its mutation to lysine represents the first reported variant at this specific active site, likely causing significant disruption of substrate interaction .

Even subtle modifications, such as shortening the zinc ligand by approximately 1.5Å through an E24D mutation, completely abolishes catalytic activity, demonstrating the precise structural requirements for proper ASPA function .

What experimental models are currently used in ASPA research?

Multiple experimental models have been developed to study ASPA function and Canavan disease pathophysiology:

What are the cutting-edge therapeutic approaches for Canavan disease?

Gene therapy has emerged as a promising approach for treating Canavan disease. A recent clinical trial by Myrtelle Inc. employed a recombinant adeno-associated virus (rAAV) vector-based gene therapy (rAAV-Olig001-ASPA) that directly targets oligodendrocytes, the brain cells responsible for producing myelin .

Six-month post-treatment data from this first-in-human clinical trial revealed:

  • Statistically significant increases in myelin, white matter, grey matter, and total brain volume

  • Reduction in cerebrospinal fluid volume

  • Improvements in motor and cognitive function measured by validated assessment scales

  • No serious drug-related adverse events

These findings contrast with the natural history of untreated Canavan disease, which typically shows progressive deterioration. The encouraging results support further development of this gene therapy approach as a potential treatment for children with Canavan disease.

What are the best practices for experimental design in ASPA-related studies?

A systematic survey of biomedical research involving laboratory animals identified several key principles for rigorous experimental design that apply to ASPA research:

  • Clear hypothesis formulation: Only 59% of surveyed studies clearly stated their hypothesis or objective, highlighting the need for more explicit framing of research questions .

  • Randomization: 87% of publications failed to use randomization in animal selection, potentially introducing selection bias .

  • Blinding: 86% of studies did not implement blinding procedures for outcome assessment, risking observer bias .

  • Statistical methodology: 30% of publications using statistical methods failed to adequately describe their methods or present results with appropriate measures of error or variability .

Researchers should address these issues by:

  • Clearly stating research hypotheses

  • Implementing randomization and blinding protocols

  • Using appropriate statistical methods and reporting complete results

  • Providing comprehensive details on experimental animals and procedures

How can researchers accurately assess phenotypic outcomes in Canavan disease models?

Multiple complementary approaches are recommended for comprehensive phenotypic assessment:

  • Neuroimaging: Magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) provide quantitative measurements of:

    • Myelin content

    • White and grey matter volumes

    • Total brain volume

    • Cerebrospinal fluid volume

  • Functional assessments: Validated clinical scales provide standardized measures of neurological function:

    • Gross Motor Function Measure (GMFM) for motor abilities

    • Mullen Scales of Early Learning (MSEL) for cognitive development

  • Biochemical assays: Measurement of NAA levels and ASPA enzyme activity provides direct biochemical indicators of disease severity and treatment efficacy.

  • Molecular analyses: Genetic testing to confirm specific ASPA mutations and expression studies to assess transcript and protein levels offer insight into molecular disease mechanisms.

What approaches can be used to establish genotype-phenotype correlations in Canavan disease?

Establishing genotype-phenotype correlations requires systematic comparison of:

  • Mutation characteristics: Analysis of mutation type (missense, nonsense, frameshift, splice-site) and location within functional domains of the ASPA protein.

  • In vitro enzyme activity: Quantification of residual ASPA activity associated with specific mutations using radiometric assays .

  • Structural modeling: Homology modeling to predict how specific mutations affect protein structure and function, as demonstrated by the modeling of ASPA based on zinc-dependent carboxypeptidase A .

  • Clinical severity scales: Standardized assessment of disease progression and severity using validated clinical measures.

  • Natural history data: Comparison of disease course in patients with different genotypes to identify patterns in disease onset, progression, and response to interventions.

What emerging technologies show promise for advancing ASPA research?

Several cutting-edge technologies are poised to accelerate progress in understanding and treating Canavan disease:

  • CRISPR-Cas9 gene editing: Enables precise correction of ASPA mutations in cellular and animal models, offering both research tools and potential therapeutic approaches.

  • Single-cell transcriptomics: Provides detailed characterization of cell-type-specific responses to ASPA deficiency, helping to elucidate disease mechanisms at cellular resolution.

  • Hypoimmunogenic iPSC-derived oligodendrocyte progenitor cells: Development of these cells offers potential for off-the-shelf cell therapy approaches for myelin disorders including Canavan disease .

  • Advanced neuroimaging techniques: Novel MRI sequences with improved sensitivity for detecting white matter changes enable more precise monitoring of disease progression and treatment effects.

How can researchers develop more predictive preclinical models of Canavan disease?

Improving preclinical models requires multi-faceted approaches:

  • Humanized mouse models: Generating mice expressing human ASPA variants to better recapitulate human disease features.

  • Brain organoids: Developing three-dimensional brain organoids from patient-derived iPSCs to model complex cellular interactions in a human genetic background.

  • Rigorous study design: Implementing randomization, blinding, and appropriate controls in preclinical studies to improve translational relevance .

  • Standardized outcome measures: Establishing consistent methodologies for assessing treatment efficacy across different research groups to facilitate comparison of results.

What are the key considerations for designing clinical trials for Canavan disease therapies?

Designing effective clinical trials for rare diseases like Canavan disease presents unique challenges and considerations:

  • Natural history studies: Establishing robust natural history data to serve as comparators for intervention trials, as illustrated by Myrtelle's approach of comparing treatment outcomes to age-matched untreated patients .

  • Biomarker development: Identifying and validating surrogate endpoints that correlate with clinical outcomes but can be measured more rapidly or with greater precision.

  • Adaptive trial designs: Implementing flexible protocols that can be modified based on interim results, maximizing the information gained from small patient populations.

  • Multi-domain outcome assessment: Using complementary measures spanning imaging, functional, and biochemical domains to comprehensively evaluate treatment effects, as demonstrated in the rAAV-Olig001-ASPA gene therapy trial .

Product Science Overview

Introduction

Aspartoacylase, also known as ASPA, is a hydrolytic enzyme that plays a crucial role in the metabolism of N-acetyl-L-aspartic acid (NAA). This enzyme is encoded by the ASPA gene in humans and is primarily found in the brain. Aspartoacylase catalyzes the deacylation of NAA to produce aspartate and acetate .

Structure and Function

Aspartoacylase is a zinc-dependent hydrolase that promotes the deprotonation of water to use as a nucleophile, a mechanism similar to many other zinc-dependent hydrolases . The enzyme is a dimer composed of two identical monomers, each consisting of 313 amino acids. Each monomer has two distinct domains: the N-terminal domain (residues 1-212) and the C-terminal domain (residues 213-313) .

The primary function of aspartoacylase is to regulate the levels of NAA in the brain. NAA is abundant in the brain and is involved in maintaining white matter integrity. The hydrolysis of NAA by aspartoacylase is essential for the proper functioning of the central nervous system .

Clinical Significance

Mutations in the ASPA gene can lead to a rare autosomal recessive neurodegenerative disorder known as Canavan disease. This condition is characterized by the accumulation of NAA in the brain, leading to spongy degeneration of the white matter. Canavan disease results in severe developmental delays, motor dysfunction, and early mortality .

Recombinant Aspartoacylase

Human recombinant aspartoacylase is produced using recombinant DNA technology, which involves inserting the ASPA gene into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the enzyme for research and therapeutic purposes. Recombinant aspartoacylase is used in studies to understand the enzyme’s structure, function, and role in diseases like Canavan disease .

Research and Applications

Research on aspartoacylase has provided valuable insights into its role in the brain and its involvement in neurological disorders. Studies have shown that aspartoacylase activity is crucial for the maintenance of white matter and the proper functioning of oligodendrocytes, the cells responsible for myelination in the central nervous system .

In addition to its role in the brain, aspartoacylase has been implicated in the metabolic reprogramming of cancer cells. Recent studies suggest that ASPA may play a role in tumor development and progression, making it a potential target for cancer therapy .

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.