Recombinant Mouse Calcineurin subunit B type 1 (Ppp3r1)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder

Note: We will prioritize shipping the format currently in stock. If you have specific format requirements, please specify them during order placement.

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 advance notice 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 settle 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% and can serve as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer composition, 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. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.

The tag type will be determined during production. If you require a specific tag type, please inform us, and we will prioritize its development.

Synonyms
Ppp3r1; CnbCalcineurin subunit B type 1; Protein phosphatase 2B regulatory subunit 1; Protein phosphatase 3 regulatory subunit B alpha isoform 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-170
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Target Protein Sequence
GNEASYPLE MCSHFDADEI KRLGKRFKKL DLDNSGSLSV EEFMSLPELQ QNPLVQRVID IFDTDGNGEV DFKEFIEGVS QFSVKGDKEQ KLRFAFRIYD MDKDGYISNG ELFQVLKMMV GNNLKDTQLQ QIVDKTIINA DKDGDGRISF EEFCAVVGGL DIHKKMVVDV
Uniprot No.

Target Background

Function

Regulatory subunit of calcineurin, a calcium-dependent, calmodulin-stimulated protein phosphatase. Confer calcium sensitivity.

Gene References Into Functions
  1. Calcineurin B1 deficiency in glial cells impairs the small intestine, leading to malnutrition and death in mice, highlighting a crucial role for calcineurin in enteric glial cells. PMID: 29709916
  2. Expression of the RNA-binding protein, tristetraprolin (TTP), is down-regulated following calcineurin inhibition. PMID: 29723192
  3. Mice lacking Calcineurin B in CD11c(high)MHCII(+) cells (Cnb1(CD11c) mice) spontaneously develop intestinal inflammation and exhibit colitis susceptibility. PMID: 29549257
  4. Reduced calcineurin activity in the amygdala increases anxiety-like and, to some extent, depression-like behaviors. PMID: 24742621
  5. Calcineurin is essential for the development of the smooth muscle wall in coronary arteries. PMID: 23946498
  6. Macrophage Wnt-Calcineurin-Flt1 signaling regulates mouse wound angiogenesis and repair. PMID: 23303818
  7. Intact calcineurin/NFAT signaling is crucial for p53 and senescence-associated mechanisms protecting against skin squamous cancer development. PMID: 20485437
  8. Heart-specific CnB1 loss reduces expression of key Ca(2+)-handling genes, potentially leading to arrhythmia and decreased contractility. PMID: 20037164
  9. Calcineurin B1 is essential for positive selection during thymocyte development. PMID: 15030770
  10. Cardiac-specific ablation of the Cn regulatory subunit reduces gene expression of peroxisome proliferator-activated receptor gamma coactivator 1alpha and peroxisome proliferator-activated receptor alpha. PMID: 15262994
  11. Endothelin 1 (ET-1) activates calcineurin, causing NFATc1 nuclear translocation, implicating this pathway in ET-1-mediated osteoblast stimulation. PMID: 17237284
  12. Calcineurin B1 deletion significantly reduces skeletal muscle degeneration and histopathology compared to sarcoglycan-null genotypes. PMID: 17289669
  13. Inhibition of Cnb1 in osteoblasts increases bone mass by directly increasing osteoblast differentiation and indirectly decreasing osteoclastogenesis. PMID: 17884821
  14. Early calcineurin/NFAT signaling creates a developmental period of ERK hypersensitivity, enabling weak signals to induce positive selection. PMID: 18046413
  15. Calcineurin B1 gene deficiency reduces macrophage responsiveness to LPS. PMID: 19318421
  16. Cardiac-specific calcineurin b1-deficient mice exhibit impaired contractile function and calcium handling. PMID: 19700627
Database Links
Protein Families
Calcineurin regulatory subunit family
Subcellular Location
Cytoplasm, cytosol. Cell membrane. Cell membrane, sarcolemma. Cell membrane; Lipid-anchor.
Tissue Specificity
Brain specific.

Q&A

What is the structural composition of Calcineurin and how does Ppp3r1 function within this complex?

Calcineurin is a heterodimeric calcium-dependent serine-threonine phosphatase (also known as protein phosphatase 2B or PP2B) consisting of two primary subunits: a 60 kDa catalytic subunit (calcineurin A) and a 19 kDa calcium-binding regulatory subunit (calcineurin B, encoded by Ppp3r1) . The regulatory B subunit is structurally critical as it is abundantly expressed in the cytoplasm of neurons and functions to regulate calcineurin activity through calcium binding .

Methodologically, researchers investigating the structural properties of Ppp3r1 should note that this subunit, rather than the catalytic subunit, is primarily responsible for the regulation of calcineurin activity. In experimental settings, it is essential to consider that Ppp3r1 achieves activation through calcium and calmodulin binding, which induces conformational changes that enable the phosphatase to dephosphorylate target proteins . This activation occurs specifically in response to calcium signaling triggered by external stimuli such as hormones, growth factors, or neurotransmitters.

What experimental methods are most effective for studying Ppp3r1 expression in neural tissues?

When investigating Ppp3r1 expression in neural tissues, researchers should employ a multi-method approach:

  • RNA-level analysis: Quantitative PCR (qPCR) for gene expression analysis provides a sensitive method for detecting changes in Ppp3r1 mRNA levels. RNA-seq can offer genome-wide context for expression patterns .

  • Protein-level analysis: Western blotting with specific anti-Ppp3r1 antibodies enables quantification of protein expression, while immunohistochemistry/immunofluorescence allows for spatial localization within brain regions .

  • Functional assays: Calcineurin activity assays using phosphatase substrates can measure the functional consequences of altered Ppp3r1 expression .

  • Single-cell approaches: For cell-type specific expression patterns, single-cell RNA-seq or multiplexed immunofluorescence can reveal heterogeneity across neural populations .

When designing these experiments, researchers should include appropriate controls and consider the impact of calcium concentration in their experimental buffers, as calcium directly influences Ppp3r1 function. Time-course experiments are particularly valuable for capturing dynamic changes in expression following stimulation.

How do recombinant Ppp3r1 preparations differ from endogenous protein in functional assays?

Recombinant Ppp3r1 preparations may exhibit important differences from endogenous protein that researchers must account for in experimental design:

  • Post-translational modifications: Endogenous Ppp3r1 undergoes various PTMs that may be absent or differently distributed in recombinant preparations depending on the expression system used .

  • Binding partners: Native Ppp3r1 exists in complex with calcineurin A and potentially other proteins, whereas recombinant Ppp3r1 may lack these physiological interactions unless specifically co-expressed .

  • Calcium sensitivity: Recombinant preparations may show altered calcium binding dynamics compared to endogenous protein, potentially affecting activation thresholds in functional assays .

For methodologically robust experiments, researchers should:

  • Validate recombinant protein functionality through calcineurin activity assays

  • Compare recombinant and endogenous protein behavior in parallel experiments when possible

  • Consider supplementing assays with calcineurin A subunit if using recombinant Ppp3r1 alone

  • Carefully control calcium concentrations to ensure appropriate activation conditions

What are the established signaling pathways involving Ppp3r1 in neuronal cells?

Ppp3r1, as part of the calcineurin complex, participates in several critical signaling pathways in neuronal cells:

PathwayPpp3r1 RoleKey SubstratesFunctional Outcome
Axon GuidanceRegulates phosphorylation of cytoskeletal proteinsMAP1B, CofilinDirects axonal growth and pathfinding
Glutamatergic SynapseModulates receptor traffickingAMPAR, NMDARRegulates synaptic strength and plasticity
Long-Term Potentiation (LTP)Counterbalances kinase activityCREB, PP1 inhibitorsInfluences memory formation
MAPK SignalingCross-talks with MAPK cascadeERK, MEKAffects neuronal survival and differentiation

Methodologically, when investigating these pathways, researchers should:

  • Employ phospho-specific antibodies to monitor substrate phosphorylation states

  • Use specific calcineurin inhibitors (FK506/tacrolimus or Cyclosporin A) as controls

  • Consider calcium imaging to correlate calcium dynamics with pathway activation

  • Design time-course experiments to capture the temporal dynamics of signaling cascades

What are the technical considerations for using Ppp3r1 knockout or knockdown models in neurodegenerative disease research?

When implementing Ppp3r1 knockout or knockdown models for neurodegenerative disease research, researchers should address several technical considerations:

  • Model selection:

    • Conventional knockouts may be embryonically lethal due to Ppp3r1's essential functions

    • Conditional (Cre-loxP) knockouts allow temporal and spatial control of gene deletion

    • AAV-mediated knockdown provides flexibility for region-specific manipulation in adult animals

    • CRISPR-Cas9 approaches enable precise genomic editing with reduced off-target effects

  • Compensatory mechanisms:

    • Monitor expression of other calcineurin regulatory subunits that might compensate for Ppp3r1 loss

    • Assess calcium-dependent phosphatase activity to determine functional consequences

    • Evaluate pathway adaptation through phosphoproteomic analysis

  • Phenotypic analysis timeline:

    • Early changes in dendritic spine morphology (2-4 weeks)

    • Intermediate alterations in synaptic protein composition (1-3 months)

    • Late-stage behavioral and cognitive deficits (3-12 months)

  • Experimental readouts:

    • Tau phosphorylation status in Alzheimer's models

    • Synaptic density measurements using Golgi staining or EM

    • Electrophysiological properties (LTP/LTD)

    • Behavioral assessment (memory, learning, motor function)

In Alzheimer's disease research specifically, evidence suggests that low PPP3R1 expression accurately predicts disease onset, with area under curve (AUC) analysis demonstrating predictive value. Therefore, researchers should incorporate quantitative expression analysis in their experimental designs to correlate with phenotypic progression .

How do differential interactions between Ppp3r1 and TLR pathways influence neuroinflammatory responses?

The interaction between Ppp3r1 and Toll-like receptor (TLR) pathways represents a complex intersection between calcium signaling and innate immunity that influences neuroinflammation. Researchers investigating this relationship should consider:

  • Mechanistic interaction: Recombinant calcineurin subunit B has been shown to interact directly with the ectodomain of TLR4 in vitro, potentially serving as an endogenous regulator of TLR signaling . Experimental approaches should include:

    • Co-immunoprecipitation assays to confirm physical interactions

    • Surface plasmon resonance to determine binding kinetics

    • FRET-based approaches to visualize interactions in living cells

  • Downstream signaling consequences:

    • Ppp3r1 stimulation leads to phosphorylation of interferon regulatory factor 3 (IRF3)

    • Degradation of IκB-α occurs following Ppp3r1 activation

    • Both processes culminate in enhanced transcription and production of β-interferon

  • Pathway-specific effects:

    • Unlike LPS stimulation, Ppp3r1-mediated signaling does not promote degradation of TRAF3

    • Ppp3r1 may preferentially promote K63 ubiquitination of TRAF3 for type I interferon production

    • Both TAK-1 and TBK-1 mRNA levels are upregulated following Ppp3r1 stimulation

For neuroinflammation studies, researchers should implement time-course experiments measuring:

  • Pro-inflammatory cytokine production (TNF-α, IL-12p70, IL-1β, IL-6)

  • Chemokine secretion (IL-8, IP-10, MIP-1α, MIP-1β)

  • Interferon pathway activation markers

  • Microglial activation status

The methodological approach should include both in vitro models (primary microglia, mixed glial cultures) and in vivo paradigms (stereotaxic injection of recombinant Ppp3r1, neuroinflammation models).

What contradictions exist in the literature regarding Ppp3r1's role in neuroprotection versus neurodegeneration?

The literature presents several apparent contradictions regarding Ppp3r1's role in neuroprotection versus neurodegeneration that researchers should carefully consider:

  • Alzheimer's disease context:

    • Protective evidence: PPP3R1 expression is inversely associated with AD neuropathy and clinical dementia rating, suggesting a neuroprotective role

    • Detrimental evidence: Calcineurin activation by calcium dysregulation can promote tau hyperphosphorylation and amyloid pathology

  • Cell survival regulation:

    • Pro-survival: Calcineurin regulates adaptive responses to environmental stress

    • Pro-death: Excessive calcineurin activation can trigger apoptotic pathways

  • Neuroinflammatory effects:

    • Anti-inflammatory: β-interferon production stimulated by Ppp3r1 has shown protective effects

    • Pro-inflammatory: Ppp3r1 stimulation enhances pro-inflammatory cytokine secretion

  • Therapeutic implications:

    • Inhibition paradigm: Calcineurin inhibitors like FK506 show neuroprotective effects in some models

    • Activation paradigm: CNB injection has shown anti-tumor effects, suggesting beneficial immune stimulation

To resolve these contradictions, researchers should:

  • Conduct dose-response studies to determine threshold effects

  • Implement cell-type specific manipulations to distinguish neuronal versus glial contributions

  • Use temporal control systems to separate acute versus chronic effects

  • Employ systems biology approaches to model network-level outcomes

  • Design experiments that directly compare Ppp3r1 function across different disease models and stages

How can Ppp3r1-based therapeutic strategies be optimized for specific neurological disorders?

Optimizing Ppp3r1-based therapeutic strategies requires systematic consideration of several factors:

  • Target specificity engineering:

    • Design peptide or small molecule modulators that target specific Ppp3r1 interaction surfaces

    • Develop isoform-selective approaches that distinguish between calcineurin subunit variants

    • Create cell-type specific delivery systems using targeted nanoparticles or viral vectors

    • Engineer conditional activation systems responsive to disease-specific microenvironments

  • Context-dependent intervention:

    • For Alzheimer's disease: Strategies should focus on maintaining PPP3R1 levels, as downregulation correlates with disease progression and tau hyperphosphorylation

    • For neuroinflammatory conditions: Controlled Ppp3r1 administration might stimulate beneficial β-interferon production

    • For autoimmune disorders: Selective calcineurin inhibition shows promise, as evidenced by clinical studies in lupus nephritis

  • Delivery optimization parameters:

ParameterConsiderationsMethodological Approach
Blood-brain barrier penetrationMolecular size, lipophilicity, transport mechanismsIn vitro BBB models, in vivo biodistribution studies
Temporal controlDisease stage-specific intervention, circadian effectsInducible expression systems, chronotherapy
Spatial targetingRegion-specific delivery, cellular compartmentalizationStereotaxic delivery, organelle-targeted constructs
Dose optimizationTherapeutic window, side effect profileDose-escalation studies, PK/PD modeling
  • Combinatorial approaches:

    • Pair Ppp3r1 modulation with complementary pathway interventions

    • Consider sequential treatment protocols to address different disease phases

    • Explore synergistic effects with existing approved therapies

For methodological validation, researchers should implement:

  • Target engagement biomarkers

  • Functional readouts of calcineurin activity

  • Disease-relevant endpoints

  • Long-term safety monitoring

In Alzheimer's disease specifically, PPP3R1-targeted therapeutic strategies should focus on the identified cross-talking pathways: axon guidance, glutamatergic synapse, LTP, and MAPK signaling pathways, as these represent the mechanisms through which PPP3R1 influences disease pathogenesis .

What are the translational considerations when using mouse Ppp3r1 findings to inform human clinical research?

When translating findings from mouse Ppp3r1 research to human clinical applications, researchers must address several critical considerations:

ParameterMouseHumanResearch Implication
Brain region expressionHigher in hippocampusMore uniform distributionRegion-specific intervention may require adjustment
Age-related changesRapid changes (months)Gradual changes (years)Timeline for intervention must be scaled
Immune responseTh1-dominantMore balanced Th1/Th2Immunomodulatory effects may differ
Drug metabolismFaster clearanceSlower metabolismDosing regimens require adjustment
  • Clinical trial design considerations:

    • Include PPP3R1 expression as a stratification biomarker

    • Consider genetic variants (e.g., rs1868402) that affect PPP3R1 expression in population selection

    • Implement longitudinal monitoring of PPP3R1 levels as a pharmacodynamic marker

How can Ppp3r1 expression be effectively quantified in clinical biospecimens?

Accurate quantification of PPP3R1 expression in clinical biospecimens requires robust methodological approaches:

  • Cerebrospinal fluid (CSF) analysis:

    • ELISA-based quantification with validated antibodies

    • Mass spectrometry-based proteomics for absolute quantification

    • Digital ELISA (Single Molecule Array) for ultra-sensitive detection

    Researchers should establish standardized collection protocols, as PPP3R1 measurements may be affected by diurnal variation, sample processing time, and storage conditions.

  • Blood-based measurements:

    • Peripheral blood mononuclear cell (PBMC) expression via qPCR

    • Plasma/serum protein levels via immunoassays

    • Exosomal PPP3R1 as a potential CNS-derived biomarker

  • Tissue analysis from biopsies or post-mortem samples:

    • RNA-Seq or NanoString technology for gene expression profiling

    • Immunohistochemistry with digital pathology quantification

    • Laser capture microdissection for cell-type specific analysis

    • Spatial transcriptomics for regional expression patterns

  • Quality control considerations:

ParameterChallengeRecommended Solution
Pre-analytical variablesSample degradationStandardized processing within 30 minutes
Reference standardsBatch variationInclude recombinant protein calibrators
Antibody specificityCross-reactivityValidate with knockout controls
NormalizationInter-individual variationUse multiple housekeeping references
  • Clinical correlation:

    • Establish reference ranges across age, sex, and disease status

    • Correlate PPP3R1 levels with clinical dementia rating and other cognitive assessments

    • Perform longitudinal measurements to establish prognostic value

Research indicates that PPP3R1 expression is inversely associated with AD neuropathy and clinical dementia rating, making quantification particularly valuable for neurodegenerative disease monitoring .

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.