Recombinant Mouse Putative tyrosine-protein phosphatase auxilin (Dnajc6), partial

Shipped with Ice Packs
In Stock

Description

General Information

DnaJ heat shock protein family (Hsp40) member C6, also known as DNAJC6, belongs to the evolutionarily conserved DNAJ/HSP40 family of proteins, which regulate molecular chaperone activity by stimulating ATPase activity of HSP70 proteins . DNAJC6 acts as an auxilin protein and has been linked to early-onset Parkinson's disease (PD) .

Role in Parkinson's Disease

Mutations in the DNAJC6 gene, which encodes for the HSP40 auxilin, have been identified in patients with early-onset Parkinson's disease (PD) . Studies using human embryonic stem cells with CRISPR-Cas9-mediated gene editing have demonstrated that DNAJC6 mutations cause key PD pathologic features, including:

  • Midbrain-type dopamine (mDA) neuron degeneration

  • Pathologic α-synuclein aggregation

  • Increase of intrinsic neuronal firing frequency

  • Mitochondrial and lysosomal dysfunctions in human midbrain-like organoids (hMLOs)

In addition, neurodevelopmental defects were also manifested in hMLOs carrying the mutations . Transcriptomic analyses revealed that defects in DNAJC6-mediated endocytosis impair the WNT-LMX1A signal during mDA neuron development, and reduced LMX1A expression during development caused the generation of vulnerable mDA neurons with the pathologic manifestations .

DNAJC6 and Autolysosomal Degradation

DNAJC6 may have a role in autolysosomal degradation of α-syn in an LMX1A-independent manner . Functional lysosomes are regenerated by an autophagic lysosome reformation process, the terminal step of autophagy, which involves the clathrin function necessary for proto-lysosome budding from reformation tubules . Golgi-to-lysosome protein transport is also mediated by clathrin-mediated vesicular trafficking . In DNAJC6 mutant cells, the number of free clathrin is expected to decrease due to a defect in the clathrin uncoating process, which might explain why the autophagy-lysosomal process is impaired .

Impact on Vesicular Transport

The loss of DNAJC6 function can impair the vesicular transport process of lysosomal proteins, ultimately decreasing lysosomal enzyme activities . Specifically, autolysosome formation caused by reduced lysosome number and vesicular transport of lysosomal enzymes were defected in DNAJC6 mutant neuronal cells .

Association with Dopamine Homeostasis

DNAJC6 mutations may affect D2R protein levels and normal postsynaptic function, and it is likely that presynaptic D2R autoreceptor function will also be affected .

Role in Neurodevelopment

DNAJC6 mutations can cause neurodevelopmental defects . Exogenous DNAJC6 expression in mutant NSC cultures could almost completely rescue all PD phenotypes in differentiated mDA neuronal cells .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which may serve as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The tag type is determined during production. If a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
Dnajc6; Kiaa0473; Putative tyrosine-protein phosphatase auxilin; EC 3.1.3.48; DnaJ homolog subfamily C member 6
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function

This protein recruits HSPA8/HSC70 to clathrin-coated vesicles and facilitates their uncoating. It plays a significant role in clathrin-mediated endocytosis within neurons.

Database Links

Q&A

What is the primary function of auxilin (Dnajc6) in neuronal cells?

Auxilin (encoded by the DNAJC6 gene) functions as a neuron-specific co-chaperone that recruits heat shock cognate protein 70 (HSC70) to clathrin-coated vesicles for disassembly. This process is crucial for clathrin-mediated endocytosis (CME), particularly at presynaptic terminals. Auxilin plays an essential role in the uncoating of clathrin-coated vesicles following endocytosis, allowing for proper synaptic vesicle recycling. The protein contains domains that interact with both clathrin and the HSC70 chaperone, facilitating the ATP-dependent removal of the clathrin coat from newly formed vesicles. This process is critical for maintaining normal synaptic transmission and neuronal communication through efficient recycling of synaptic vesicles .

How can stem cell-derived models be optimized for studying Dnajc6 function?

Optimizing stem cell-derived models for Dnajc6 research requires careful attention to several methodological aspects. First, researchers should consider using isogenic controls created through CRISPR-Cas9 correction of patient-derived iPSCs or introduction of mutations into wild-type lines to minimize variability from genetic background. The differentiation protocol is critical—protocols that closely recapitulate midbrain developmental patterning through careful temporal control of morphogens (SHH, FGF8, WNT agonists) yield more authentic midbrain dopaminergic neurons. Three-dimensional models like midbrain-like organoids (hMLOs) provide additional anatomical context that better represents the in vivo environment. Researchers should monitor key developmental markers throughout differentiation, particularly LMX1A, FOXA2, EN1, and NURR1, as these have been shown to be affected by DNAJC6 mutations. Finally, extended culture periods (60+ days) allow for the development of mature synaptic connections and more pronounced phenotypes in DNAJC6-mutant neurons .

What experimental timeline is optimal for detecting Dnajc6-related phenotypes in cellular models?

Based on current research findings, an optimal experimental timeline for detecting Dnajc6-related phenotypes in cellular models should span from early neural induction through mature neuron stages. Early developmental defects become apparent around days 15-20 of differentiation, as evidenced by altered expression of key developmental regulators like LMX1A and EN1. These early changes reflect disruptions in the WNT-LMX1A autoregulatory loop that is critical for proper midbrain dopaminergic neuron specification. The expression of later developmental markers like NURR1 should be assessed around day 30. For mature neuronal phenotypes, including synaptic dysfunction, neurite outgrowth defects, and dopaminergic markers, cultures should be maintained for at least 60-80 days. α-synuclein aggregation and neurodegeneration phenotypes typically require extended culture periods of 90-120 days. This staged analysis approach allows researchers to distinguish between developmental defects and later degenerative phenotypes, providing insight into the full spectrum of Dnajc6-related pathology .

Differentiation StageCulture DayKey Markers/Phenotypes to AssessRelevance to Dnajc6 Pathology
Neural Induction0-7Neural progenitor markersBaseline assessment
Early Regionalization15-20LMX1A, EN1, WNT signalingEarly developmental defects
Neuronal Specification30-45NURR1, PITX3, TH expressionDopaminergic specification
Neuronal Maturation60-80Neurite outgrowth, synaptic markersStructural/functional abnormalities
Late-stage Phenotypes90-120α-synuclein, cell viabilityNeurodegenerative processes

How does loss of Dnajc6 function affect WNT signaling in dopaminergic neuron development?

Loss of Dnajc6 function disrupts the WNT signaling pathway through impairment of clathrin-mediated endocytosis (CME) of WNT ligand-receptor complexes. This mechanism represents a crucial link between endocytic dysfunction and developmental abnormalities in midbrain dopaminergic neuron development. When DNAJC6/auxilin is absent, the endocytic capacity of neural stem cells is significantly reduced, as demonstrated by decreased uptake of FM1-43 dye in mutant cultures. This endocytic impairment prevents proper internalization and signaling of WNT ligand-receptor complexes, which is required for signal activation. Consequently, nuclear β-catenin levels are reduced, leading to decreased transcriptional activation of key target genes, including LMX1A and EN1. These genes are critical for midbrain dopaminergic neuron specification and identity. Furthermore, the WNT-LMX1A regulatory relationship is bidirectional—LMX1A also activates transcription of WNT cytokines and RSPO2, creating a positive feedback loop that is disrupted in the absence of DNAJC6. Treatment with CME inhibitors (Dyngo-4a and PitStop2) in wild-type neural stem cells replicates this phenotype, confirming that the endocytic deficit is responsible for WNT signaling impairment .

How do developmental defects from Dnajc6 mutations contribute to later neurodegeneration?

The developmental defects resulting from Dnajc6 mutations create a foundation for later neurodegeneration through several interconnected mechanisms. Most critically, reduced expression of the transcription factors LMX1A, EN1, and NURR1 during early development results in midbrain dopaminergic neurons with compromised cellular identity. These transcription factors normally continue to be expressed in adult neurons where they protect against toxic insults and support mitochondrial function. Neurons developing without proper expression of these factors are inherently more vulnerable to stressors throughout their lifespan. Additionally, early disruption of WNT signaling pathways, which are important for dopaminergic neuron specification and survival, creates neurons with altered gene expression profiles and compromised cellular resilience. The endocytic defects present from early development progressively impact multiple cellular functions, including receptor trafficking, neurotrophic signaling, and synaptic maintenance. Over time, these cumulative stresses combined with the inherent vulnerability of the improperly specified neurons lead to progressive neurodegeneration. This developmental vulnerability hypothesis explains why DNAJC6 mutations, despite being present from conception, typically manifest as juvenile-onset parkinsonism that progressively worsens over time .

What are the optimal protocols for assessing clathrin-mediated endocytosis in Dnajc6 models?

To thoroughly assess clathrin-mediated endocytosis (CME) in Dnajc6 models, researchers should employ multiple complementary approaches. A fundamental method is the FM dye uptake assay, where lipophilic styryl dyes like FM1-43 are used to quantify endocytic capacity. In this protocol, neurons are incubated with FM dye during stimulation (typically using high K+ solution), followed by thorough washing and imaging to quantify internalized fluorescence. This provides a functional readout of endocytic efficiency. For more specific assessment of clathrin-dependent processes, transferrin uptake assays can be utilized, as transferrin receptor endocytosis relies exclusively on the clathrin pathway. Pulse-chase experiments with fluorescently labeled transferrin allow tracking of internalization kinetics and recycling rates. For molecular assessment, researchers should examine clathrin coat dynamics using live-cell imaging of fluorescently tagged clathrin light chain or adaptor proteins like AP2. Advanced visualization techniques such as total internal reflection fluorescence (TIRF) microscopy enable high-resolution imaging of clathrin pit formation and resolution at the plasma membrane. Finally, ultrastructural analysis using electron microscopy can directly visualize clathrin-coated vesicles and quantify their density, size, and morphology in synaptic terminals .

How can researchers effectively measure WNT signaling disruption in Dnajc6-deficient models?

Effectively measuring WNT signaling disruption in Dnajc6-deficient models requires a multi-level approach targeting different components of the pathway. At the transcriptional level, quantitative PCR should be used to assess expression of key WNT pathway genes, including WNT ligands (WNT1, WNT4), receptors (FZD), and downstream targets (AXIN2, LMX1A, EN1). RNA sequencing provides a more comprehensive view of pathway disruption and should be analyzed using gene set enrichment analysis focusing on WNT-related gene ontologies. At the protein level, Western blotting for total and nuclear β-catenin provides a direct measure of canonical WNT pathway activation, with particular attention to nuclear translocation. Immunocytochemistry for β-catenin with nuclear counterstaining allows visualization of nuclear translocation at the single-cell level. For functional assessment, TOPFlash reporter assays (using luciferase reporters driven by TCF/LEF binding sites) provide quantitative measurement of WNT-dependent transcriptional activity. Chromatin immunoprecipitation (ChIP) assays targeting β-catenin binding to promoter regions of key genes like LMX1A and EN1 directly assess transcriptional regulation. Finally, rescue experiments using WNT agonists (e.g., CHIR99021) or by expressing constitutively active β-catenin can confirm the causative relationship between endocytic defects and WNT signaling impairment .

What techniques are most reliable for quantifying dopaminergic neuron vulnerability in Dnajc6 models?

Quantifying dopaminergic neuron vulnerability in Dnajc6 models requires a comprehensive approach combining multiple assessment techniques. For survival analysis, researchers should perform longitudinal quantification of tyrosine hydroxylase (TH)-positive neurons through immunostaining, tracking changes over extended culture periods (60-120 days). This should be complemented with general cell death markers like cleaved caspase-3 or TUNEL assays to distinguish between selective dopaminergic degeneration and general cytotoxicity. Functional assessment should include measurement of dopamine release using high-performance liquid chromatography (HPLC) or fast-scan cyclic voltammetry, as well as electrophysiological recordings to detect alterations in firing patterns and synaptic transmission. Molecular vulnerability can be assessed by measuring expression levels of neuroprotective transcription factors (LMX1A, EN1, NURR1) using both qPCR and immunostaining approaches. Researchers should also quantify mitochondrial function through measurements of membrane potential (using dyes like TMRM), respiratory capacity (using Seahorse analyzers), and ROS production. Assessment of protein homeostasis should include quantification of α-synuclein aggregation, ubiquitinated protein accumulation, and lysosomal function. Finally, stressed conditions using oxidative stressors (H₂O₂, rotenone) or proteotoxic stress (proteasome inhibitors) can reveal latent vulnerability that might not be apparent under basal conditions .

How might differences in Dnajc6 splice variants contribute to tissue-specific phenotypes?

The differential expression and function of Dnajc6 splice variants likely contribute significantly to tissue-specific phenotypes in both normal physiology and disease states. Human DNAJC6 undergoes alternative splicing to produce multiple isoforms, with neuronal specificity potentially arising from tissue-specific splicing regulation. To investigate this complex question, researchers should first comprehensively characterize the expression patterns of different splice variants across brain regions and developmental stages using RNA-seq with splice junction analysis or isoform-specific qPCR. Particular attention should be paid to variants expressed in vulnerable midbrain dopaminergic neurons versus those in resistant neuronal populations. Functional differences between splice variants can be assessed through rescue experiments, transfecting specific isoforms into DNAJC6-deficient cellular models and measuring their capacity to restore normal endocytic function, WNT signaling, and dopaminergic marker expression. Domain-specific functions may be elucidated by creating chimeric proteins with swapped domains between variants. Additionally, mass spectrometry analysis of protein-protein interactions for different splice variants might reveal isoform-specific binding partners that could explain regional vulnerability. Finally, researchers should investigate whether disease-causing mutations differentially affect specific splice variants, potentially explaining why some mutations cause predominantly neurological phenotypes while others affect broader developmental processes .

What is the interplay between Dnajc6 and GAK (auxilin-2) in compensating for each other's function?

The interplay between Dnajc6 (auxilin-1) and GAK (auxilin-2) represents a critical area for understanding differential vulnerability in DNAJC6-related disorders. While DNAJC6 is neuron-specific, GAK is ubiquitously expressed and shares significant structural and functional similarity, suggesting potential compensatory mechanisms. To investigate this relationship, researchers should first quantify relative expression levels of both proteins across different neural cell types and brain regions, particularly noting whether GAK expression increases in response to DNAJC6 deficiency as a compensatory mechanism. Conditional knockout models allow temporal control of gene deletion, enabling researchers to determine whether developmental versus adult loss of DNAJC6 differentially engages GAK compensation. Double knockdown/knockout experiments can reveal synergistic effects that suggest functional redundancy. Biochemical approaches using co-immunoprecipitation can identify shared versus distinct binding partners and substrates. Single-cell transcriptomics comparing vulnerable dopaminergic neurons with resistant neuronal populations may reveal cell type-specific differences in the DNAJC6:GAK ratio that explain selective vulnerability. Finally, overexpression of GAK in DNAJC6-deficient models can test whether GAK upregulation represents a viable therapeutic strategy. Understanding this compensatory relationship may explain why Dnajc6-knockout mice show less severe phenotypes than human patients and could provide insights for therapeutic approaches .

How do Dnajc6 mutations affect non-canonical functions beyond clathrin-mediated endocytosis?

Investigating the non-canonical functions of Dnajc6 beyond clathrin-mediated endocytosis is essential for fully understanding its role in disease pathogenesis. Several methodological approaches can address this complex question. Unbiased protein interactome analysis using proximity labeling techniques (BioID or APEX) coupled with mass spectrometry can identify novel interaction partners beyond the known clathrin and HSC70 associations. Subcellular fractionation followed by Western blotting can determine whether auxilin localizes to unexpected cellular compartments like mitochondria or the nucleus, suggesting functions beyond the synaptic terminal. Phosphoproteomics can evaluate changes in cellular signaling networks in Dnajc6-deficient models, as the protein has putative phosphatase domains that might directly regulate signaling pathways. Lipidomics analysis may reveal alterations in membrane composition or lipid signaling that could affect numerous cellular processes. Transcriptomic analysis comparing early versus late changes in gene expression patterns can distinguish between primary effects of auxilin loss and secondary adaptations. Additionally, researchers should investigate potential interactions with α-synuclein and other Parkinson's disease-associated proteins to determine whether auxilin has direct effects on proteostasis beyond its role in endocytosis. Understanding these non-canonical functions may explain the complex neurodevelopmental phenotypes observed in patients with DNAJC6 mutations .

What gene therapy approaches show the most promise for treating Dnajc6-related disorders?

Current research indicates that lentiviral vector-based gene therapy approaches show significant promise for treating DNAJC6-related disorders. Based on available data, the most effective gene therapy strategies should address several key considerations. First, the therapeutic vector should contain the full-length wild-type DNAJC6 gene under the control of a neuron-specific promoter to ensure appropriate cell-type expression. Preliminary studies have demonstrated that transfection of mutant midbrain dopaminergic neurons with lentiviral vectors containing wild-type DNAJC6 can restore auxilin protein levels and improve clathrin-mediated endocytosis, providing proof-of-concept for this approach. AAV9 vectors may be particularly suitable for clinical translation due to their ability to cross the blood-brain barrier and efficiently transduce neurons. For optimal efficacy, treatment should ideally begin early, before extensive neurodegeneration has occurred, although even later intervention might slow disease progression. The therapeutic construct should be designed to resist silencing through the use of appropriate regulatory elements. A critical consideration is the potential immunogenicity of the expressed protein in patients with null mutations who might recognize the wild-type protein as foreign. Researchers should also evaluate whether partial restoration of auxilin function is sufficient for therapeutic benefit, as this would influence the required efficiency of gene transfer .

How can the WNT-LMX1A pathway be targeted to rescue developmental defects in Dnajc6 models?

Targeting the WNT-LMX1A pathway represents a promising strategy for rescuing developmental defects in Dnajc6 models, particularly during early stages of midbrain dopaminergic neuron development. Several methodological approaches can be employed. Direct activation of the WNT pathway using small molecule GSK3β inhibitors like CHIR99021 can bypass the endocytic requirement for WNT signal activation, directly stabilizing β-catenin. The timing and duration of treatment are critical—treatment should coincide with the normal developmental window of WNT signaling activation during dopaminergic specification. Alternatively, researchers can use genetic approaches to overexpress key transcription factors downstream of WNT signaling, particularly LMX1A and EN1, which are reduced in DNAJC6-mutant models. This approach directly addresses the deficiency in these critical factors without relying on upstream WNT activation. Targeted epigenetic modifiers that enhance expression of silenced developmental genes offer another potential approach. For in vivo application, researchers should investigate whether localized delivery of WNT agonists to the developing midbrain can prevent the developmental defects in animal models. It's important to note that this approach would likely be most effective as a preventive measure during development rather than as a treatment for established disease, highlighting the importance of early intervention or genetic screening for DNAJC6 mutations in high-risk populations .

What combined therapeutic approaches might address both developmental and degenerative aspects of Dnajc6-related pathology?

Addressing both the developmental and degenerative aspects of DNAJC6-related pathology requires sophisticated combinatorial therapeutic strategies. An effective approach would target multiple aspects of disease pathophysiology through staged interventions. For developmental defects, early intervention with gene therapy to restore DNAJC6 expression, combined with WNT pathway modulators during critical developmental windows, could correct the fundamental specification defects in dopaminergic neurons. This early intervention should be followed by maintenance strategies targeting the degenerative aspects of the disease. These could include neuroprotective approaches such as neurotrophic factor delivery (GDNF, BDNF) to support vulnerable neurons, antioxidants to manage increased oxidative stress, and compounds that enhance mitochondrial function. Additionally, agents that promote α-synuclein clearance, such as autophagy enhancers or specific anti-aggregation compounds, could address the proteostatic aspects of the pathology. Small molecules that enhance residual clathrin-mediated endocytosis or promote alternative endocytic pathways might partially compensate for the fundamental endocytic deficit. For advanced disease stages, cell replacement therapies using properly specified dopaminergic progenitors could replace lost neurons. The optimal therapeutic regimen would likely change over the disease course, with initial focus on correcting developmental specification followed by shifting emphasis to neuroprotection and eventually to symptom management and cell replacement as the disease progresses .

Therapeutic ApproachTarget MechanismDisease StagePotential AdvantagesChallenges
Gene therapy (AAV/lentiviral)Restore DNAJC6 expressionEarly/preventiveAddresses root causeDelivery efficiency, immune response
WNT agonistsRescue LMX1A/EN1 expressionDevelopmentalTargets key pathwayTiming dependency, off-target effects
LMX1A/EN1 overexpressionRestore TF expressionEarly/developmentalBypasses upstream defectsProper regulation of expression
Neurotrophic factorsSupport vulnerable neuronsProgressiveWell-established neuroprotectionDelivery challenges, broad effects
Autophagy enhancersPromote protein clearanceDegenerativeAddresses α-synuclein aggregationSpecificity, off-target effects
Endocytic enhancersCompensate for CME defectsMultiple stagesTargets core cellular deficitFew specific compounds available
Cell replacementReplace lost neuronsAdvancedAddresses end-stage degenerationIntegration challenges, cell source

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.