PARK7 Mouse

Parkinson Disease Protein 7 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Genetic Characteristics of PARK7 Mouse Models

PARK7 knockout mice are engineered through targeted deletion of exon 2–4 of the Park7 gene, resulting in complete loss of DJ-1 protein expression . Key features include:

  • Homology: 92% amino acid identity between human and mouse PARK7 .

  • Expression: Ubiquitous, with high levels in the brain, kidneys, and testes .

  • Phenotype: Age-dependent motor deficits, dopaminergic neuron vulnerability, and increased sensitivity to oxidative stress .

Metabolic Regulation

PARK7 prevents damage from glycolytic intermediates:

MechanismOutcomeModelCitation
Degradation of 1,3-BPGPrevents glycerate/phosphoglycerate adducts on proteins/metabolitesPark7 −/− mice, human cell lines
Antioxidant activityStabilizes Nrf2, upregulates SOD1/3Mouse brain, Drosophila

Loss of PARK7 leads to accumulation of damaged metabolites (e.g., N-glyceroyl-glutamate) in brain and peripheral tissues .

Neurodegenerative Phenotypes

  • Motor deficits: Hypokinesia and impaired coordination manifest by 8–12 months .

  • Dopaminergic system: Reduced tyrosine hydroxylase expression in substantia nigra (40% loss by 18 months) .

  • Pathology: No Lewy body formation, but increased α-synuclein aggregation susceptibility .

Sex-Specific Transcriptomic Changes

A 2024 study revealed sex-dependent effects in Park7 −/− mice:

ParameterMale MiceFemale Mice
Differential genes (8 months)1,214 upregulated, 987 downregulatedNo significant changes
Key pathwaysNRF2 signaling, estrogen metabolism, EMTUnaffected
Astrocyte dysfunctionCYP1B1 downregulation (linked to estrogen/retinoic acid metabolism)Normal CYP1B1 levels

Antibodies

  • Clone 4H4: Detects human PARK7 (21 kDa) but not mouse orthologs .

  • AF3668: Cross-reactive with human/mouse PARK7 (23–30 kDa) .

Chemical Probes

  • JYQ-92: Cyanimide inhibitor covalently targets Cys106; used for activity-based profiling .

  • SulfoCy5 probe: Enables PARK7 inhibition assays in cell lysates .

Therapeutic Insights

  • Metabolic intervention: Targeting 1,3-BPG metabolism may benefit PD patients with PARK7 mutations .

  • NRF2 activation: Rescues antioxidant deficits in male Park7 −/− astrocytes .

Limitations and Controversies

  • Variable phenotypes: Some studies report mild motor deficits, while others show progressive neurodegeneration .

  • Methylglyoxal role: PARK7’s deglycase activity toward methylglyoxal adducts remains debated .

Product Specs

Introduction
PARK7 is a protein found throughout the body that plays a role in many cell functions. These include the production of sperm and fertilization, cancer development, interaction with RNA, signaling of the androgen receptor, and managing oxidative stress. Changes in the PARK7 gene can lead to a type of Parkinson's disease that starts early in life, known as autosomal recessive early-onset Parkinson's disease 7 (Park7).
Description
Recombinant PARK7 Mouse protein is produced in E. coli bacteria. It is a single chain of 212 amino acids (specifically amino acids 1-189), with a molecular weight of 22.4 kDa. The protein is not glycosylated, meaning it does not have sugars attached to it. A 23 amino acid His-tag is attached to the beginning (N-terminus) of the PARK7 protein to aid in purification, which is carried out using specialized chromatographic techniques.
Physical Appearance
The product is a clear and colorless solution that has been sterilized by filtration.
Formulation
The PARK7 protein is supplied in a solution with a concentration of 0.5 mg/ml. The solution also contains Phosphate Buffered Saline (PBS) at a pH of 7.4, 20% glycerol to protect the protein during freezing, and 1mM DTT to prevent the formation of unwanted bonds within the protein.
Stability
For short-term storage (2-4 weeks), the product can be kept at refrigerated at 4°C. For longer storage, it is recommended to freeze the product at -20°C. To further enhance long-term stability, consider adding a carrier protein such as HSA or BSA at a concentration of 0.1%. Repeated freezing and thawing of the product should be avoided.
Purity
Analysis by SDS-PAGE indicates that the purity of the PARK7 protein is greater than 90%.
Synonyms
Protein deglycase DJ-1, Parkinson disease protein 7 homolog.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMASKRAL VILAKGAEEM ETVIPVDVMR RAGIKVTVAG LAGKDPVQCS RDVMICPDTS LEDAKTQGPY DVVVLPGGNL GAQNLSESPM VKEILKEQES RKGLIAAICA GPTALLAHEV GFGCKVTTHP LAKDKMMNGS HYSYSESRVE KDGLILTSRG PGTSFEFALA IVEALVGKDM ANQVKAPLVL KD.

Q&A

What is the PARK7 Gene and How Does It Relate to Parkinson's Disease?

The PARK7 gene encodes the DJ-1 protein, which functions as both a protein and nucleotide deglycase that catalyzes the deglycation of Maillard adducts formed between amino groups of proteins or nucleotides and reactive carbonyl groups of glyoxals . Loss-of-function mutations in PARK7 can lead to early-onset Parkinson's disease (PD) through multiple mechanisms .

DJ-1 serves critical protective functions including:

  • Repairing methylglyoxal and glyoxal-glycated proteins

  • Deglycating cysteine, arginine, and lysine residues in proteins

  • Preventing formation of advanced glycation end products (AGEs)

  • Repairing glycated guanine in the free nucleotide pool and in DNA/RNA

These functions highlight why DJ-1 deficiency contributes to neurodegeneration in PD, particularly through increased vulnerability to oxidative stress .

How Do PARK7 Knockout Mice Compare to Other PD Genetic Models?

PARK7 knockout mice exhibit distinct characteristics compared to other genetic PD models such as LRRK2 or Parkin knockout models. These differences are essential for researchers to consider when selecting appropriate models for specific research questions.

Model TypeKey PhenotypesAge of OnsetNotable Characteristics
PARK7 knockout- Dopaminergic deficits
- Oxidative stress sensitivity
- Sex-specific transcriptome changes
8+ months- Male-specific vulnerability
- Age-dependent progression
LRRK2 transgenic- Motor coordination abnormalities
- Striatal DA release defects
- TH+ neurite dystrophy
Variable- Responds to L-DOPA
- Often lacks DA neuron degeneration
Parkin knockout- Mild motor deficits
- Heightened startle response
- Limited progressive phenotype
Variable- Often lacks clear DA neurodegeneration
- More subtle phenotypes

The variability in phenotypes across different PARK7 knockout studies can be attributed to factors including age at assessment, sex differences, genetic background, and experimental conditions . This variability highlights the importance of standardized protocols when working with these models.

What Are the Sex-Specific Transcriptomic Changes in PARK7 Knockout Mice?

Recent research has revealed striking sex-specific transcriptomic differences in PARK7 knockout mice that provide important insights into potential mechanisms of disease vulnerability:

At 3 months of age:

  • No significant transcriptomic changes in either male or female Park7 knockout mice compared to wildtype littermates

At 8 months of age:

  • Male Park7 knockout mice show extensive gene deregulation

  • Female Park7 knockout mice maintain relatively normal transcriptomic profiles

The male-specific changes at 8 months affect genes involved in:

  • Focal adhesion pathways

  • Extracellular matrix interaction

  • Epithelial-to-mesenchymal transition (EMT)

  • Nuclear factor erythroid 2-related factor 2 (NRF2) target genes

  • Antioxidant response elements

Many of the misregulated genes are known targets of estrogen and retinoic acid signaling, which already show sex-specific expression patterns in wildtype mice. This suggests that loss of DJ-1 interferes with sex hormone-dependent gene regulation pathways, potentially explaining the male bias observed in certain aspects of PD pathology .

How Does PARK7 Deletion Affect the NRF2-CYP1B1 Axis in Astrocytes?

PARK7 deletion leads to significant disruption of the NRF2-CYP1B1 axis specifically in male astrocytes, creating a potential mechanism for sex-specific vulnerability to oxidative stress . This relationship can be characterized as follows:

  • NRF2 pathway disruption:

    • DJ-1 normally stabilizes NRF2 (Nuclear factor erythroid 2-related factor 2)

    • PARK7 deletion leads to altered NRF2 target gene expression

    • Male astrocytes show greater sensitivity to this disruption

  • CYP1B1 downregulation:

    • CYP1B1 (cytochrome P450 family 1 subfamily B member 1) is significantly downregulated in male DJ-1 deficient astrocytes

    • CYP1B1 is involved in estrogen and retinoic acid metabolism

    • Knockdown of CYP1B1 alone can induce gene expression changes similar to DJ-1 depletion

These findings suggest that DJ-1's role extends beyond direct antioxidant functions to include regulation of sex hormone metabolism through the NRF2-CYP1B1 axis, which may explain the higher male susceptibility to certain aspects of PD pathology .

What Methodological Approaches Are Recommended for Studying Dopaminergic Deficits in PARK7 Mouse Models?

When investigating dopaminergic deficits in PARK7 knockout mice, researchers should implement a multi-modal approach that accounts for the age and sex-dependent nature of the phenotypes . Based on current literature, effective methodological approaches include:

  • Transcriptomic analysis:

    • RNA-seq of midbrain sections, particularly focused on the substantia nigra

    • Age-matched cohorts (3 months vs. 8+ months)

    • Sex-separated analysis (male vs. female)

  • Neurochemical assessments:

    • High-performance liquid chromatography (HPLC) for measuring striatal dopamine levels

    • Fast-scan cyclic voltammetry to assess dopamine release dynamics

    • Microdialysis for in vivo neurotransmitter measurements

  • Behavioral assessments:

    • Fine motor coordination tests (rotarod, beam traverse)

    • Locomotor activity measurements

    • Response to dopaminergic drugs (L-DOPA, apomorphine)

  • Histological and cellular analysis:

    • Tyrosine hydroxylase (TH) immunostaining for dopaminergic neuron quantification

    • Stereological counting methods

    • Assessment of oxidative stress markers

    • Analysis of mitochondrial function

For optimal results, experimental designs should include longitudinal studies that track changes from early age (3 months) through middle age (8-12 months), with careful attention to sex as a biological variable .

What Critical Experimental Design Considerations Should Be Applied When Using PARK7 Mouse Models?

When designing experiments using PARK7 mouse models, researchers should consider several critical factors to ensure robust and reproducible results:

  • Age considerations:

    • Young mice (3 months) may not exhibit significant phenotypes

    • Middle-aged mice (8+ months) show more pronounced transcriptomic changes

    • Longitudinal designs with multiple age points are recommended

  • Sex as a biological variable:

    • Males and females should be analyzed separately

    • Male mice show stronger transcriptomic changes at 8 months

    • Pooling sexes may mask significant effects or increase variability

  • Genetic background:

    • The strain background can significantly influence phenotype severity

    • Consistent genetic background should be maintained across experiments

    • Control for genetic drift in breeding colonies

  • Environmental factors:

    • Housing conditions (enriched vs. standard)

    • Diet composition

    • Light/dark cycle consistency

    • Exposure to environmental stressors

  • Challenge paradigms:

    • PARK7 knockout phenotypes may be subtle without additional stressors

    • Consider oxidative stress challenges to reveal underlying vulnerabilities

By carefully addressing these design considerations, researchers can maximize the translational value of studies using PARK7 mouse models and improve consistency across laboratories .

How Do Human iPSC Models with PARK7 Mutations Compare to PARK7 Mouse Models?

PARK7 mouse models and human iPSC models with PARK7 mutations offer complementary insights into DJ-1-related pathology:

FeaturePARK7 Mouse ModelsHuman iPSC Models with PARK7 Mutations
Species relevanceRodent biology may differ from humanDerived from human cells with human genetic background
System complexityComplete in vivo system with intact brain circuitryLimited to cell culture or organoid systems
Temporal scaleCan study aging effects over months/yearsLimited to weeks/months in culture
Cell typesAll brain cell types in natural proportionsTypically directed to specific lineages (neurons, astrocytes)
Sex-specific effectsCan study male vs. female differencesSex-specific effects preserved in XX vs. XY lines
Key findings overlap- NRF2 pathway dysregulation
- Oxidative stress sensitivity
- Mitochondrial dysfunction
- Similar molecular pathways affected
- Similar vulnerability patterns

Recent research shows that male iPSC-derived astrocytes with PARK7 loss-of-function mutations exhibit changes in the EMT pathway and NRF2 target genes similar to those observed in male PARK7 knockout mice, suggesting conservation of key molecular mechanisms between species .

For optimal translational research, combining insights from both model systems is recommended, using mouse models for in vivo, long-term studies while employing iPSC models for human-specific molecular mechanisms and personalized medicine approaches.

What CRISPR-Based Approaches Can Be Used to Create or Modify PARK7 Mouse Models?

CRISPR technology offers versatile approaches for creating and modifying PARK7 mouse models that extend beyond traditional knockout strategies:

  • CRISPR/Cas9 knockout approaches:

    • Complete gene deletion through dual sgRNA targeting

    • Introduction of frameshift mutations mimicking human pathogenic variants

    • Precise editing to introduce specific point mutations

  • CRISPR activation (CRISPRa) applications:

    • Upregulation of endogenous PARK7 expression

    • Targeting specific PARK7 promoter or enhancer regions

    • Combined with inducible systems for temporal control

  • CRISPR interference (CRISPRi) strategies:

    • Reversible repression of PARK7 expression

    • Partial knockdown to model hypomorphic alleles

    • Cell-type specific repression using tissue-specific promoters

  • Knock-in approaches:

    • Introduction of human PARK7 variants

    • Addition of reporter tags (GFP, luciferase)

    • Creation of conditional alleles (floxed PARK7)

The OriGene mouse PARK7 activation kit exemplifies commercial tools available for CRISPR-based gene activation, containing three guide RNAs targeting the PARK7 gene along with enhancer and control vectors . These approaches extend the utility of PARK7 mouse models beyond simple knockouts to more sophisticated studies of gene dosage, cell-type specific functions, and precise modeling of human mutations.

Why Do Different PARK7 Knockout Mouse Studies Report Variable Phenotype Severity?

The variability in phenotypes observed across different PARK7 knockout studies can be attributed to several interacting factors:

  • Age-dependent penetrance:

    • Transcriptomic changes become pronounced only at 8+ months in males

    • Compensatory mechanisms may mask phenotypes in younger animals

    • Age-related decline in alternative protective pathways enhances vulnerability

  • Sex-specific mechanisms:

    • Male-specific transcriptomic changes in the NRF2-CYP1B1 axis

    • Estrogen-dependent protection in females

    • Sex hormone interactions with antioxidant pathways

  • Genetic background influences:

    • Strain-dependent differences in baseline oxidative stress management

    • Modifier genes that enhance or suppress DJ-1-related phenotypes

  • Environmental and experimental variables:

    • Housing conditions affecting baseline stress levels

    • Diet and microbiome differences

    • Experimental procedures and timing

The recent finding that DJ-1 loss leads to astrocytic alterations through the NRF2-CYP1B1 axis specifically in males helps explain some of this variability . Studies using mixed-sex cohorts or younger animals may miss these effects entirely, while studies focusing on aged male mice may detect stronger phenotypes.

What Are Optimal Methods for Assessing Oxidative Stress Responses in PARK7 Knockout Mice?

Given DJ-1's central role in oxidative stress protection, comprehensive assessment of redox status in PARK7 knockout mice requires multi-modal approaches:

  • Biochemical markers of oxidative damage:

    • Protein carbonylation (DNPH assay, Western blot)

    • Lipid peroxidation (MDA, 4-HNE, F2-isoprostanes)

    • DNA/RNA oxidation (8-OHdG, 8-OHG immunostaining)

    • Advanced glycation end products (AGEs)

  • Antioxidant system evaluation:

    • Glutathione levels (GSH/GSSG ratio)

    • Antioxidant enzyme activities (SOD, catalase, GPx)

    • NRF2 pathway activation status

    • Phase II detoxifying enzyme expression

  • Mitochondrial function assessment:

    • Membrane potential measurements

    • Respiration analysis

    • mtDNA damage quantification

    • Mitochondrial morphology analysis

  • Challenge paradigms:

    • Acute oxidative stressors (hydrogen peroxide, paraquat)

    • Neurotoxins (MPTP, 6-OHDA, rotenone)

    • Inflammatory challenges

    • Metabolic stress

For sex-specific studies, researchers should note that male PARK7 knockout mice show more pronounced deficits in antioxidant responses compared to females, particularly in the NRF2 pathway and downstream targets . Combining multiple measures across different biological scales provides the most comprehensive assessment of how DJ-1 deficiency alters oxidative stress responses.

Product Science Overview

Structure and Expression

PARK7 Mouse Recombinant is produced in E. coli as a single, non-glycosylated polypeptide chain containing 212 amino acids (1-189 a.a) and has a molecular mass of 22.4 kDa . The recombinant protein is fused to a 23 amino acid His-tag at the N-terminus, which facilitates its purification through chromatographic techniques .

Function and Mechanism

DJ-1 exhibits several types of catalytic activity, including functioning as a protein deglycase (EC 3.5.1.124) . It acts as a positive regulator of androgen receptor-dependent transcription and may function as a redox-sensitive chaperone, sensing oxidative stress and protecting neurons against oxidative stress and cell death .

In different cellular compartments, DJ-1 exhibits both catalytic and non-catalytic functions. For instance, oxidative stress promotes the dissociation of cytoplasmic DJ-1 dimers into monomers, which are then translocated to the nucleus. In the nucleus, DJ-1 acts as a coactivator of various signaling pathways, preventing cell death . In mitochondria, DJ-1 is found in the synthasome, where it interacts with the β ATP synthase subunit .

Role in Parkinson’s Disease

Mutations in the DJ-1 gene (PARK7) are associated with autosomal recessive early-onset Parkinson’s disease (PD) . These mutations often result in impaired dimerization, stability, and folding of the protein, leading to its dysfunction . Downregulation of the DJ-1 gene increases the sensitivity of cells to neurotoxins, which is a characteristic feature of PD . Introduction of recombinant DJ-1 protein has been shown to attenuate the manifestation of PD pathology in experimental models .

Applications

The recombinant PARK7 protein is used in various research applications to study its role in neuroprotection and its potential therapeutic effects in PD models. It is also utilized in biochemical assays to understand its interaction with other proteins and its function under oxidative stress conditions .

Storage and Stability

PARK7 Mouse Recombinant protein is typically stored at -20°C for long-term storage and at 4°C if it will be used within 2-4 weeks . It is recommended to avoid multiple freeze-thaw cycles to maintain its stability .

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.