HAO1 Mouse

Hydroxyacid Oxidase 1 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

HAO1 Mouse Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 393 amino acids (1-370) and having a molecular mass of 43.4 kDa.
HAO1 is fused to a 23 amino acid His-Tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Hydroxyacid Oxidase 1 (HAO1), a member of the alpha hydroxy acid oxidases (HAO) enzyme family, catalyzes the FMN-dependent oxidation of glycolate to glyoxylate and glyoxylate to oxalate, reducing oxygen to hydrogen peroxide in the process. Primarily found in the liver and pancreas, HAO1 exhibits highest activity with two-carbon substrates like glycolate. This enzyme plays a significant role in hyperoxaluria, a condition characterized by excessive calcium oxalate deposits leading to kidney stone formation.
Description
Recombinant Mouse HAO1, expressed in E.coli, is a non-glycosylated polypeptide chain with a molecular weight of 43.4 kDa. It consists of 393 amino acids, with residues 1-370 representing the HAO1 sequence. A 23 amino acid His-Tag is fused to the N-terminus to facilitate purification via proprietary chromatographic methods.
Physical Appearance
A clear solution, sterile-filtered for purity.
Formulation
HAO1 protein is supplied at a concentration of 1mg/ml in a solution of Phosphate Buffered Saline (pH 7.4) containing 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Biological Activity
The specific activity, a measure of the enzyme's catalytic efficiency, is greater than 1,000 pmol/min/ug. This value represents the enzyme's ability to oxidize glyoxylate at a pH of 8.0 and a temperature of 25°C.
Synonyms

(S)-2-hydroxy-acid oxidase; EC 1.1.3.15, Glycolate oxidase, GOX, GOX1MGC142227;
GOXMGC142225, HAO1, HAO-1, HAOX1, hydroxyacid oxidase (glycolate oxidase) 1, hydroxyacid oxidase 1, Hydroxyacid Oxidase1, Hydroxyacid Oxidase-1.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMLPRLVC ISDYEQHVRS VLQKSVYDYY RSGANDQETL ADNIQAFSRW KLYPRMLRNV ADIDLSTSVL GQRVSMPICV GATAMQCMAH VDGELATVRA CQTMGTGMML SSWATSSIEE VAEAGPEALR WMQLYIYKDR EISRQIVKRA EKQGYKAIFV TVDTPYLGNR IDDVRNRFKL PPQLRMKNFE TNDLAFSPKG NFGDNSGLAE YVAQAIDPSL SWDDITWLRR LTSLPIVVKG ILRGDDAKEA VKHGVDGILV SNHGARQLDG VPATIDVLPE IVEAVEGKVE VFLDGGVRKG TDVLKALALG AKAVFVGRPI IWGLAFQGEK GVQDVLEILK EEFRLAMALS GCQNVKVIDK TLVRKNPLAV SKI.

Q&A

What is HAO1 and what is its primary biological function?

HAO1 encodes glycolate oxidase, an enzyme that catalyzes the oxidation of glycolate to glyoxylate in peroxisomes. This enzyme plays a critical role in glyoxylate metabolism and is highly expressed in the liver. In the metabolic pathway of primary hyperoxaluria type 1 (PH1), HAO1 activity leads to the production of glyoxylate, which can be converted to oxalate, a compound that forms kidney stones and causes renal damage when accumulated . The inhibition or knockout of HAO1 is a therapeutic strategy to reduce oxalate production by preventing glycolate conversion to glyoxylate, thereby potentially treating PH1 .

How do HAO1 knockout mouse models compare to human HAO1 deficiency?

Human HAO1 deficiency, as observed in a rare case of a healthy adult woman with complete HAO1 knockout, results in markedly elevated plasma glycolate levels (12 times the upper limit of normal) and urinary glycolate (6 times the upper limit of normal) without clinical phenotype . HAO1 knockout mouse models similarly exhibit elevated glycolate levels but may show variances in the magnitude of elevation compared to humans. The human knockout case demonstrates that lifelong HAO1 deficiency is compatible with normal health despite metabolic alterations, providing critical translational context for interpreting mouse model findings .

What phenotypic changes are observed in HAO1 knockout mice?

HAO1 knockout mice typically exhibit biochemical alterations similar to those observed in the identified human knockout case, including elevated glycolate levels in plasma and urine. Metabolomic analysis in the human case revealed 18 markedly elevated biochemicals (>5 standard deviations above controls), suggesting HAO1 has effects beyond glycolate oxidation that may also be present in mouse models . Despite these biochemical changes, both human and mouse HAO1 knockouts generally lack overt clinical phenotypes under normal conditions, supporting the safety of HAO1 inhibition as a therapeutic approach .

What are the recommended methods for genotyping HAO1 knockout or modified mice?

Standard genotyping for HAO1 modified mice should include PCR-based methods using primers flanking the modified region. For precise mutations like the c.997delC identified in the human HAO1 knockout case, sequence verification is essential . When establishing HAO1 knockout mouse colonies, it's recommended to perform both genomic DNA analysis and functional validation through biochemical assays of glycolate levels in plasma and urine. Expression analysis at the protein level (Western blot) is also advised to confirm complete protein knockout, as frameshift mutations like p.Leu333SerfsTer4 can lead to protein degradation or mislocalization rather than merely truncated proteins .

What biomarkers should be monitored when studying HAO1 deficiency in mice?

Based on human HAO1 knockout data, the following biomarkers should be monitored:

BiomarkerExpected ChangeSampling MethodAnalytical Technique
Plasma glycolate12× increaseNon-fasting blood sample, Na Hep collectionLC-MS/MS
Urinary glycolate6× increase24-hour urine collectionLC-MS/MS
OxalateMinimal changeUrine and plasmaIon chromatography
GlyoxylateDecreaseFlash-frozen liver tissueSpecialized enzyme assay
Additional metabolitesVariable changesPlasma metabolomicsUntargeted LC-MS

Comprehensive plasma metabolomics and lipidomics (covering ~1800 biochemicals) may reveal additional HAO1-related metabolic alterations as observed in the human knockout case .

How should tissue collection and processing be optimized for HAO1 activity assays?

For optimal HAO1 enzyme activity measurements, liver samples should be collected via rapid hepatectomy and flash-frozen in liquid nitrogen immediately. For protein localization studies, fresh liver tissue should be processed for subcellular fractionation to isolate peroxisomes. For metabolic assays, samples should be prepared according to standardized protocols as used in the human case study: blood collected in Na Hep vacutainers, inverted 10 times, centrifuged at 1500×g for 10 minutes at 4°C within 45 minutes of collection, and plasma aliquoted and stored at -80°C . For certain metabolites like glyoxylate, acidification of samples (pH 2.3-3.5) using concentrated HCl before freezing is recommended to prevent degradation .

How can single-cell multi-omic approaches enhance HAO1 mouse model studies?

Single-cell multi-omic tools can provide comprehensive molecular profiles of HAO1 expression and function at cellular resolution. These approaches enable researchers to understand cell-type-specific impacts of HAO1 deficiency across liver cell populations . By applying scRNA-seq, researchers can identify transcriptional changes in different hepatocyte populations and non-parenchymal cells, revealing potential compensatory mechanisms in HAO1 knockout mice . Additionally, scATAC-seq can uncover changes in chromatin accessibility at HAO1-regulated genes, potentially identifying upstream regulatory networks . These technologies allow for unbiased measurements across cell types isolated from HAO1 knockout mice, enabling detection of subtle phenotypic alterations that might be obscured in bulk tissue analyses .

What are the key considerations when designing HAO1 inhibition studies in mouse models?

When designing HAO1 inhibition studies, researchers should consider:

  • Inhibition strategy selection: Compare genetic knockout models with pharmacological inhibition or RNA interference approaches to distinguish developmental versus acute effects

  • Dosing regimen design: Based on the human knockout data showing complete inhibition safety, consider testing various inhibition levels from partial to complete

  • Duration of inhibition: Include long-term studies since the human case demonstrates lifelong inhibition is well-tolerated

  • Comprehensive phenotyping: Beyond primary hyperoxaluria endpoints, investigate the 18 markedly elevated biochemicals identified in the human knockout case

  • Environmental challenges: Test HAO1-inhibited mice under various metabolic stresses to identify potential context-dependent phenotypes not apparent under standard conditions

How relevant are HAO1 mouse models for studying primary hyperoxaluria type 1 (PH1)?

HAO1 mouse models provide valuable tools for studying PH1 therapeutic approaches, particularly HAO1 inhibition strategies. The human HAO1 knockout case directly supports this research direction by demonstrating that lifelong HAO1 deficiency is safe without clinical phenotype, effectively de-risking therapeutic approaches targeting HAO1 . For maximum translational relevance, researchers should design mouse studies that:

  • Compare HAO1 inhibition in wild-type versus PH1 mouse models (AGXT-deficient)

  • Measure both glycolate and oxalate levels to confirm pathway modulation

  • Assess kidney crystal formation and renal function alongside metabolic parameters

  • Consider potential compensatory mechanisms that might develop in constitutive knockout models versus acute inhibition

What insights from human HAO1 knockout cases should inform mouse model interpretation?

The rare human HAO1 knockout case provides critical contextual information for interpreting mouse model findings. The human data suggests:

  • Nearly complete loss of HAO1 activity (<2% residual activity) is compatible with normal health

  • Substantial elevations in glycolate levels (12× plasma, 6× urine) occur without clinical consequences

  • Protein mislocalization may contribute to the functional deficiency (as seen with p.Leu333SerfsTer4)

  • Beyond glycolate metabolism, HAO1 deficiency affects multiple biochemicals (18 markedly elevated metabolites)

Researchers should compare biochemical profiles between mouse models and human cases to validate translational relevance, particularly when evaluating potential therapeutic approaches targeting HAO1.

How can HAO1 mouse models be used to evaluate RNA interference therapeutics for PH1?

HAO1 mouse models are ideal platforms for evaluating RNA interference therapeutics like lumasiran for PH1. Since HAO1 silencing is the mechanism of lumasiran, an investigational RNA interference therapeutic for PH1, HAO1 knockout mice provide important insights into the target biology . When designing such studies, researchers should:

  • Compare constitutive knockout models with siRNA/RNAi-mediated transient knockdown

  • Establish dose-response relationships between HAO1 inhibition levels and metabolic outcomes

  • Measure glycolate and oxalate levels as primary pharmacodynamic endpoints

  • Monitor the 18 biochemicals identified as altered in human HAO1 deficiency to assess off-target effects

  • Evaluate both hepatic and renal outcomes in long-term studies

How can variability in HAO1 phenotypes between mouse strains be addressed?

Genetic background can significantly influence HAO1 knockout phenotypes. To address this variability:

  • Back-cross HAO1 modified mice to at least 10 generations on a uniform background (typically C57BL/6)

  • Consider using multiple strains to identify strain-specific modifier effects

  • Implement CRISPR-based modifications on pure background strains rather than hybrid backgrounds

  • Use littermate controls with appropriate genotypes for all experiments

  • Consider analyzing modifier genes through quantitative trait locus mapping if phenotypic variability persists

For translational studies, compare findings across multiple genetic backgrounds to ensure robustness of therapeutic approaches targeting HAO1.

What are the best practices for measuring glycolate and other HAO1-related metabolites in mouse models?

For accurate metabolite measurements in HAO1 mouse studies:

  • Collect blood samples via cardiac puncture or tail vein under consistent conditions (time of day, feeding status)

  • Process samples immediately according to standardized protocols (as described in 2.3)

  • For glycolate measurements, use LC-MS/MS methods with isotope-labeled internal standards

  • Include both wild-type and heterozygous controls alongside homozygous knockouts

  • Consider comprehensive metabolomic profiling beyond targeted glycolate/oxalate assays to capture the full spectrum of metabolic alterations seen in human HAO1 knockout (18 markedly elevated biochemicals)

  • For tissue measurements, normalize to tissue weight and protein content to account for sample variability

How should researchers interpret contradictory findings between HAO1 mouse models and human data?

When faced with discrepancies between mouse models and human HAO1 knockout data:

  • Consider species-specific metabolic differences that might impact glycolate/glyoxylate pathway regulation

  • Evaluate differences in genetic modification approach (complete gene deletion versus frameshift mutation)

  • Assess compensation mechanisms that might develop differently in mice versus humans

  • Examine environmental factors including diet, which significantly impacts glycolate metabolism

  • Consider age-dependent effects, as the human case demonstrates lifelong tolerance to HAO1 deficiency

  • Employ cross-species in vitro systems (mouse versus human hepatocytes) to directly compare HAO1 function and inhibition effects

What novel approaches could enhance HAO1 mouse model utility for PH1 research?

To advance HAO1 mouse models for PH1 research:

  • Develop humanized HAO1 mouse models expressing human HAO1 variants to better model therapeutic responses

  • Create dual knockout models combining HAO1 deficiency with AGXT mutations to directly model PH1 intervention

  • Implement tissue-specific inducible HAO1 expression systems to define critical windows for intervention

  • Apply single-cell multi-omic approaches to characterize cell-type-specific responses to HAO1 modulation

  • Develop in vivo imaging approaches to visualize oxalate crystal formation and clearance in real-time

  • Investigate the impact of HAO1 deficiency on the broader metabolic network using systems biology approaches

How might environmental and dietary factors interact with HAO1 deficiency in mouse models?

The interaction between diet and HAO1 deficiency warrants dedicated investigation:

  • Compare standard chow versus glycolate-enriched diets in HAO1 knockout mice

  • Evaluate vitamin B6 supplementation effects, given its role as a cofactor in glyoxylate metabolism

  • Test caloric restriction and high-fat diet impacts on glycolate/oxalate handling in HAO1-deficient mice

  • Investigate gut microbiome influences on oxalate metabolism in HAO1 knockout models

  • Examine differences between male and female HAO1 knockout mice under various dietary conditions

These studies would complement the human HAO1 knockout case data, which represents a single individual with specific environmental and genetic background factors .

What potential applications exist for HAO1 mouse models beyond primary hyperoxaluria research?

HAO1 mouse models have potential applications beyond PH1 research:

  • Investigating peroxisome biology and broader metabolic network regulation

  • Studying mechanisms of protein mislocalization and degradation (based on the p.Leu333SerfsTer4 variant)

  • Exploring metabolic adaptations to enzyme deficiencies

  • Developing and validating novel RNA interference therapeutics targeting liver-expressed enzymes

  • Investigating the 18 markedly elevated biochemicals identified in human HAO1 knockout for potential new roles of HAO1

  • Using HAO1-deficient primary cells for in vitro drug screening platforms

Product Science Overview

Introduction

Hydroxyacid Oxidase 1 (HAO1), also known as glycolate oxidase, is an enzyme that belongs to the superfamily of alpha hydroxy acid oxidases. This enzyme plays a crucial role in the oxidation of glycolate to glyoxylate and glyoxylate to oxalate, with the concomitant reduction of oxygen to hydrogen peroxide .

Structure and Expression

HAO1 is a single, non-glycosylated polypeptide chain consisting of 393 amino acids, with a molecular mass of approximately 43.4 kDa. The recombinant form of HAO1 is produced in Escherichia coli and is fused to a 23 amino acid His-Tag at the N-terminus . The enzyme is primarily expressed in the liver and pancreas, where it exhibits the highest activity on two-carbon substrates such as glycolate .

Function and Mechanism

The primary function of HAO1 is to catalyze the FMN-mediated oxidation of glycolate to glyoxylate and glyoxylate to oxalate. This reaction is significant as it helps in the metabolism of glycolate, a byproduct of various metabolic pathways. The enzyme’s activity is crucial in preventing the accumulation of glycolate and glyoxylate, which can lead to the formation of calcium oxalate crystals and kidney stones .

Biological Significance

HAO1 is of particular interest due to its role in hyperoxaluria, a disorder characterized by excessive oxalate production leading to kidney stone formation. The enzyme’s activity in the liver and pancreas is essential for maintaining oxalate homeostasis in the body .

Recombinant Production

The recombinant form of HAO1 is produced using Escherichia coli as the host organism. The production process involves the expression of the HAO1 gene in E. coli, followed by purification using proprietary chromatographic techniques. The recombinant enzyme is available in various quantities and is used for laboratory research purposes .

Stability and Storage

The recombinant HAO1 protein is formulated in phosphate-buffered saline (pH 7.4) with 10% glycerol. It is recommended to store the enzyme at 4°C if it will be used within 2-4 weeks. For long-term storage, the enzyme should be frozen at -20°C, and it is advisable to add a carrier protein to prevent degradation .

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.