Recombinant Mouse Casein kinase I isoform gamma-1 (Csnk1g1)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Mouse Casein Kinase I Isoform Gamma-1 (Csnk1g1)

Recombinant Mouse Casein Kinase I Isoform Gamma-1 (Csnk1g1) is a serine/threonine protein kinase belonging to the Casein Kinase 1 (CK1) family. This enzyme plays a crucial role in various cellular processes, including cell proliferation, survival, and signaling pathways. The CK1 family is known for its involvement in numerous biological functions, and the gamma-1 isoform is particularly noted for its unique roles in cellular regulation.

Biological Functions

  • Cell Signaling: Csnk1g1 participates in signaling pathways that are crucial for cell growth and survival. For instance, it can interact with other proteins to modulate their activity through phosphorylation.

  • Necroptosis: Although primarily associated with CK1γ isoforms, the broader CK1 family's involvement in necroptosis suggests potential roles for Csnk1g1 in programmed cell death pathways, particularly in the context of cellular stress responses .

  • Cancer: The CK1 family, including gamma isoforms, has been implicated in cancer progression. CircRNAs associated with CK1 gamma have been shown to influence cancer cell proliferation and survival, suggesting indirect roles for Csnk1g1 in oncogenic processes .

Research Findings

Recent studies have highlighted the importance of CK1 family members in various diseases, including cancer and inflammatory disorders. While specific research on Csnk1g1 is limited, its involvement in cellular processes suggests potential applications in therapeutic strategies targeting CK1 pathways.

Table 1: Expression and Regulation of Csnk1g1

RegulatorEffect on Csnk1g1 ExpressionReference
EpoxiconazoleIncreases expression
Ethyl MethanesulfonateIncreases expression
FipronilIncreases expression
FlavonoidsDecreases expression
Folic AcidDecreases expression

Therapeutic Potential

The CK1 family, including Csnk1g1, presents opportunities for therapeutic intervention, particularly in diseases where CK1 activity is dysregulated. Targeting CK1 pathways has been explored in cancer therapy, with potential applications in other conditions where CK1 plays a critical role .

Product Specs

Form
Lyophilized powder
Lead Time
Delivery time varies depending on the purchase method and location. Please contact your local distributor for specific delivery timelines. Shipping is standard with blue ice packs. Dry ice shipping is available upon request with an additional fee; please contact us in advance to arrange this.
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 collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Csnk1g1; Casein kinase I isoform gamma-1; CKI-gamma 1; EC 2.7.11.1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-459
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Target Protein Sequence
MDHSNREKDD RQRTTKTMAQ RNTHCSRPSG TSTSSGVLMV GPNFRVGKKI GCGNFGELRL GKNLYTNEYV AIKLEPIKSR APQLHLEYRF YKQLGSAGEG LPQVYYFGPC GKYNAMVLEL LGPSLEDLFD LCDRTFTLKT VLMIAIQLLS RMEYVHSKNL IYRDVKPENF LIGRQGNKKE HVIHIIDFGL AKEYVDPETK KHIPYREHKS LTGTARYMSI NTHLGKEQSR RDDLEALGHM FMYFLRGSLP WQGLKADTLK ERYQKIGDTK RNTPIEALCE NFPEEMATYL RYVRRLDFFE KPDYEYLRTL FTDLFERKGY TFDYAYDWVG RPIPTPVGSV HVDSGASAIT RESHTHRDRP SQQQPLRNQT TSSERRGEWE IQPSRQTNTS YLTSHLAADR HGGSVQVVSS TNGELNVDDP TGAHSNAPIT AHAEVEVVEE AKCCCFFKRK RKKTAQRHK
Uniprot No.

Target Background

Function
Recombinant Mouse Casein kinase I isoform gamma-1 (Csnk1g1) is a serine/threonine-protein kinase. Casein kinases are characterized by their preference for acidic proteins, such as caseins, as substrates. Csnk1g1 phosphorylates numerous proteins and is involved in Wnt signaling. It phosphorylates CLSPN and regulates fast synaptic transmission mediated by glutamate.
Database Links
Protein Families
Protein kinase superfamily, CK1 Ser/Thr protein kinase family, Casein kinase I subfamily
Subcellular Location
Cytoplasm.
Tissue Specificity
Expressed in both the striatum and the neocortex.

Q&A

What is the basic structure and function of Mouse Csnk1g1?

Mouse Casein kinase I isoform gamma-1 (Csnk1g1) is a serine/threonine protein kinase that belongs to the casein kinase I family. Its structure features a highly conserved kinase domain with distinct N- and C-terminal regions that differentiate it from other casein kinase isoforms. The protein preferentially utilizes acidic proteins such as caseins as substrates for phosphorylation . Functionally, Csnk1g1 is involved in the growth and morphogenesis of cells, with ubiquitous expression across multiple tissue types . In neuronal tissue, Csnk1g1 plays a particularly important role in regulating the phosphorylation of N-methyl-D-aspartate receptors, thereby influencing synaptic transmission .

How does the expression pattern of Csnk1g1 vary across different tissues and developmental stages?

Csnk1g1 expression exhibits variability across different tissues, with notable presence in the brain, though it is considered ubiquitously expressed . The expression levels are known to vary across tissues, which may correlate with tissue-specific functions of the protein. While all three casein kinase I gamma isoforms (CSNK1G1, CSNK1G2, and CSNK1G3) are widely expressed, their relative abundance differs between tissues, suggesting tissue-specific roles . Developmental expression patterns of Csnk1g1 show dynamic regulation, with particular importance during neurological development, as evidenced by the association between CSNK1G1 mutations and neurodevelopmental disorders .

What are the key differences between mouse Csnk1g1 and other casein kinase I gamma isoforms?

The casein kinase I gamma family includes three main isoforms: Csnk1g1, Csnk1g2, and Csnk1g3. These isoforms share a highly conserved kinase domain but exhibit considerable heterogeneity in their N- and C-terminal regions, which are believed to determine the functional specificities of each subtype . Interestingly, while the most distal C-terminal region is highly conserved among all three CK1G isoforms, this region has specific functionality in cellular compartmentalization . Mouse Csnk1g1 also differs from CK1 delta, another family member, in terms of antibody cross-reactivity profiles, indicating structural differences that can be exploited for experimental detection .

How does compartmentalization regulate Csnk1g1 activity and substrate access?

The compartmentalization of Csnk1g1 is primarily controlled by its C-terminal region, which determines its cytoplasmic punctate localization pattern. Recent research has revealed that the C-terminal region serves as a determinant for the subcellular localization of Csnk1g1 and other gamma isoforms . The loss of the C-terminus redistributes the kinase from specific cytoplasmic compartments to the nucleocytoplasm, significantly altering its ability to access and phosphorylate specific substrate proteins .

This compartmentalization-based regulatory mechanism allows Csnk1g1 to access substrates present in specific intracellular organelles. For example, research has demonstrated that C-terminal truncation-induced redistribution of CK1G3 (a related isoform) causes functional changes in cellular processes . Similar mechanisms are likely operational for Csnk1g1, suggesting that proper subcellular localization is essential for the normal function of this kinase in regulating its downstream targets.

What methodologies are most effective for studying Csnk1g1 interactions with potential substrates?

MethodologyApplicationAdvantagesLimitations
Co-immunoprecipitationProtein-protein interactionsIdentifies physiological interactionsMay miss transient interactions
Phosphorylation assaysSubstrate identificationDirect measurement of kinase activityRequires purified proteins
Proximity ligation assayIn situ interaction detectionVisualizes interactions in native contextLimited quantification
Mass spectrometryPhosphoproteomic analysisLarge-scale, unbiased approachComplex data analysis
CRISPR-Cas9 gene editingFunctional validationPrecise genetic manipulationOff-target effects

For studying Csnk1g1 interactions with potential substrates, researchers should employ both biochemical and cellular approaches. In vitro kinase assays using recombinant Csnk1g1 and potential substrates can establish direct phosphorylation relationships . For cellular studies, co-immunoprecipitation followed by Western blotting has proven effective for detecting Csnk1g1-substrate complexes . Additionally, more advanced techniques like proximity ligation assays or FRET-based approaches can detect interactions in their native cellular context. Mass spectrometry-based phosphoproteomics following Csnk1g1 manipulation (overexpression, knockdown, or inhibition) can identify global changes in phosphorylation patterns attributable to Csnk1g1 activity .

What is the role of Csnk1g1 in neurodevelopmental disorders?

Heterozygous de novo variants in the CSNK1G1 gene have been associated with neurodevelopmental disorders characterized by intellectual disability and, in some cases, autism spectrum disorder . Clinical studies of individuals with CSNK1G1 mutations have revealed a consistent pattern of developmental delay across all patients, with at least 60% of affected individuals also presenting with diagnoses of autism spectrum disorder . Additionally, dysmorphic facial features have been noted in patients, although the specific features vary considerably between individuals .

The mechanistic basis for these associations likely stems from Csnk1g1's role in regulating synaptic transmission through phosphorylation of N-methyl-D-aspartate receptors . Disruption of this regulatory function may alter neural circuit development and function, leading to the observed neurodevelopmental phenotypes. The absence of additional major malformations in affected individuals suggests that CSNK1G1's role in development may be relatively specific to neurological systems, despite its ubiquitous expression .

What detection methods are most reliable for recombinant mouse Csnk1g1 in different experimental contexts?

For the detection of recombinant mouse Csnk1g1 in experimental systems, several validated methodologies exist with varying applications based on the research context:

Western blotting provides reliable detection of Csnk1g1 in cell and tissue lysates. Antibodies specific to Csnk1g1 have been validated for this purpose, with specific bands typically detected at approximately 49 kDa . When selecting antibodies, researchers should verify cross-reactivity with other casein kinase isoforms, as some antibodies may detect multiple family members .

For immunocytochemistry and immunohistochemistry applications, several antibodies have been validated for the detection of Csnk1g1 in fixed cells and tissues . This approach allows visualization of the subcellular localization of Csnk1g1, which is particularly relevant given the importance of compartmentalization for its function . Typical protocols involve fixation with 4% paraformaldehyde, permeabilization with 0.1% Triton X-100, and immunostaining with validated primary antibodies followed by fluorophore-conjugated secondary antibodies .

For quantitative analysis of gene expression, quantitative real-time PCR (qPCR) using primers specific to mouse Csnk1g1 mRNA provides sensitive and specific detection . This method is particularly useful for analyzing expression patterns across tissues or experimental conditions.

How can researchers effectively modulate Csnk1g1 function in experimental models?

ApproachMechanismAdvantagesConsiderations
siRNA/shRNATransient/stable knockdownRapid reduction in expressionVariable efficiency
CRISPR-Cas9Gene knockout/mutationComplete loss of functionPotential compensation
C-terminal truncationDomain-specific modificationAltered localizationPreserves kinase activity
Kinase-dead mutationsCatalytic inactivationDominant-negative effectMay act as scaffold
Chemical inhibitorsATP-competitive bindingRapid and reversibleLimited specificity

Researchers can modulate Csnk1g1 function through several genetic and pharmacological approaches. For genetic manipulation, RNA interference (siRNA or shRNA) targeting Csnk1g1 has been successfully employed to reduce expression levels . For complete knockout, CRISPR-Cas9 genome editing targeting exonic regions can generate cell lines or animal models lacking functional Csnk1g1 .

Domain-specific modifications represent a sophisticated approach to studying Csnk1g1 function. Particularly, C-terminal truncations that remove the final 20-38 amino acids have been shown to significantly alter the subcellular localization and consequently the function of casein kinase I gamma isoforms . These truncations can be introduced through genome editing or expression of mutant constructs.

For transient modulation, overexpression of wild-type or mutant Csnk1g1 (such as kinase-dead mutants) using plasmid or viral vectors provides a means to study gain-of-function or dominant-negative effects, respectively . When designing such experiments, careful consideration should be given to the expression level and cellular compartmentalization of the recombinant protein.

What considerations are important when designing functional studies of Csnk1g1 in neurological contexts?

When designing functional studies of Csnk1g1 in neurological contexts, several key considerations should be addressed:

Cellular models should be selected based on relevance to the neurological process under investigation. Primary neuronal cultures, neuronal cell lines, or induced pluripotent stem cell (iPSC)-derived neurons may be appropriate depending on the research question. For studies related to neurodevelopmental disorders, developmental models that recapitulate the temporal aspects of neural differentiation may be particularly valuable .

The subcellular compartmentalization of Csnk1g1 is critical for its function, with the C-terminal region playing an essential role in localization . Experimental designs should account for this by including imaging studies to verify proper localization of wild-type or mutant Csnk1g1 proteins. When using tagged versions of Csnk1g1, researchers should verify that the tag does not interfere with normal localization or function.

Given Csnk1g1's role in phosphorylating N-methyl-D-aspartate receptors and influencing synaptic transmission , functional readouts should include measures of synaptic function, such as electrophysiological recordings, calcium imaging, or biochemical assessment of receptor phosphorylation states. Additionally, behavioral assays in animal models may be relevant for connecting molecular functions to neurodevelopmental phenotypes associated with CSNK1G1 mutations .

How can mouse models be used to investigate Csnk1g1's role in neurodevelopmental disorders?

Mouse models represent valuable tools for investigating Csnk1g1's role in neurodevelopmental disorders. Several approaches can be employed to create informative models:

Genetic models harboring mutations that mirror those identified in human patients with neurodevelopmental disorders can provide insights into pathophysiological mechanisms . These models can be generated using CRISPR-Cas9 genome editing to introduce specific mutations corresponding to clinically relevant variants. Particularly, heterozygous de novo variants similar to those found in patients with developmental delay and autism spectrum disorder would be most relevant .

Conditional knockout models using tissue-specific or temporally controlled Cre-lox systems allow for selective deletion of Csnk1g1 in specific brain regions or developmental stages. This approach can help dissect the regional and temporal requirements for Csnk1g1 function in neurodevelopment, potentially identifying critical periods during which Csnk1g1 function is essential .

For phenotypic analysis, comprehensive behavioral testing batteries should assess cognitive function, social behavior, and neurological function to parallel human clinical presentations . Additionally, electrophysiological studies examining synaptic transmission, particularly involving NMDA receptors, can provide mechanistic insights into how Csnk1g1 mutations affect neural circuit function .

What are the potential consequences of different types of Csnk1g1 mutations on protein function and disease phenotypes?

Different types of Csnk1g1 mutations can have distinct consequences for protein function and resultant disease phenotypes:

Mutations affecting the kinase domain may directly impair catalytic activity, potentially resulting in reduced phosphorylation of physiological substrates such as NMDA receptors . These mutations would be expected to produce loss-of-function effects, potentially explaining neurodevelopmental phenotypes through reduced synaptic plasticity.

Mutations in the C-terminal region may alter subcellular localization without necessarily affecting kinase activity, as demonstrated by studies showing that truncation of the C-terminus redistributes the protein from cytoplasmic puncta to the nucleocytoplasm . Such alterations in compartmentalization could disrupt the ability of Csnk1g1 to access its proper substrates, effectively causing functional deficiency despite normal kinase activity.

Missense mutations may have more subtle effects, potentially altering protein-protein interactions, substrate specificity, or regulatory mechanisms without completely abolishing function. The clinical presentations associated with different CSNK1G1 mutations suggest variable expressivity, with consistent developmental delay but variable presence of autism spectrum disorder and differences in dysmorphic features .

How does Csnk1g1 function in oncogenic pathways, particularly in thyroid cancer?

Recent research has uncovered a role for circular RNA derived from the CSNK1G1 gene (circ-CSNK1G1) in thyroid cancer pathogenesis . Circ-CSNK1G1 functions as a microRNA sponge for miR-149-5p, effectively reducing the availability of this microRNA to regulate its downstream targets . This mechanism represents a distinct function from the protein kinase activity of Csnk1g1 itself.

Experimental evidence for circ-CSNK1G1's oncogenic role comes from various in vitro and in vivo assays. In vitro, manipulation of circ-CSNK1G1 levels affects cancer cell viability, proliferation, apoptosis, cell cycle progression, and invasion capacity as measured by CCK-8, colony formation, EdU incorporation, flow cytometry, and transwell assays, respectively . Animal models have further validated these findings, demonstrating that circ-CSNK1G1 modulation affects tumor growth in vivo .

Mechanistically, circ-CSNK1G1 appears to function through the miR-149-5p/MAPK1 axis, with MAPK1 serving as a downstream effector that promotes thyroid cancer progression when de-repressed by circ-CSNK1G1's sponging of miR-149-5p . This pathway represents a potential therapeutic target, as disruption of the circ-CSNK1G1/miR-149-5p interaction or inhibition of downstream MAPK1 activity could potentially mitigate the oncogenic effects.

What emerging technologies might advance our understanding of Csnk1g1 function and regulation?

Several emerging technologies hold promise for advancing our understanding of Csnk1g1 function and regulation:

Cryo-electron microscopy could provide high-resolution structural information about Csnk1g1, particularly in complex with substrates or regulatory proteins. Such structural insights would enhance our understanding of the molecular mechanisms underlying Csnk1g1's substrate specificity and regulation, potentially informing structure-based drug design efforts.

Optogenetic and chemogenetic approaches allow for precise temporal control of protein function, which could be applied to study Csnk1g1 activity in specific cellular compartments or developmental timepoints. These techniques might reveal time-sensitive aspects of Csnk1g1 function that are particularly relevant to neurodevelopmental processes.

Single-cell technologies, including single-cell RNA sequencing and single-cell proteomics, could reveal cell type-specific functions of Csnk1g1 and identify rare cell populations that are particularly sensitive to Csnk1g1 dysfunction. This approach might be especially valuable for understanding the cellular basis of neurodevelopmental phenotypes associated with CSNK1G1 mutations .

Advanced microscopy techniques, such as super-resolution microscopy and live-cell imaging, could provide detailed insights into the dynamic compartmentalization of Csnk1g1 and its interactions with substrates in real-time. Given the importance of compartmentalization for Csnk1g1 function , these approaches could reveal novel regulatory mechanisms.

What are the most promising therapeutic strategies targeting Csnk1g1 for neurodevelopmental disorders?

The association between CSNK1G1 mutations and neurodevelopmental disorders suggests several potential therapeutic strategies:

Small molecule modulators of Csnk1g1 activity or localization represent another potential therapeutic avenue. Compounds that enhance the remaining Csnk1g1 activity in individuals with partial loss-of-function mutations, or that correct mislocalization of compartmentalization-defective Csnk1g1 variants, might ameliorate associated phenotypes.

Targeting downstream pathways affected by Csnk1g1 dysfunction may offer more immediate therapeutic opportunities. Given Csnk1g1's role in NMDA receptor regulation , modulators of glutamatergic signaling might compensate for altered synaptic function resulting from CSNK1G1 mutations. Such approaches would benefit from detailed characterization of the signaling pathways disrupted in specific CSNK1G1-associated conditions.

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.