CAMK2N2 Human

Calcium/Calmodulin-Dependent Protein Kinase II Inhibitor 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Overview

CAMK2N2 (HGNC: 24197) is located on chromosome 3q27.1 and encodes a 79-amino-acid protein that shares high similarity with rat CaM-KII inhibitory protein . Key features include:

PropertyDetails
Gene ID94032 (NCBI)
ProteinCK2N2_HUMAN (UniProt)
SynonymsCAMKIIN, CAM-KIIN
FunctionInhibits CAMK2 by trapping Ca²⁺/calmodulin, preventing kinase activation
ExpressionPredominantly in brain tissues, with lower levels in adrenal glands

Molecular Mechanism

CAMK2N2 binds to the autoregulatory domain of CAMK2, blocking its interaction with calcium/calmodulin and suppressing phosphorylation of downstream targets like AMPA receptors . Structural studies reveal:

  • Domain: Lacks catalytic activity but contains a conserved inhibitory motif .

  • Regulatory Role: Modulates synaptic plasticity by controlling CAMK2-driven phosphorylation cascades critical for long-term potentiation .

Neurodevelopmental Disorders

  • CAMK2N2 dysregulation is implicated in intellectual disability (ID) and autism spectrum disorders. It indirectly affects CAMK2A/B, whose mutations directly cause ID .

  • Overexpression in neuronal models disrupts dendritic spine formation, impairing cognitive function .

Cancer

  • Elevated CAMK2N2 expression correlates with colon adenocarcinoma progression by inhibiting CAMK2-mediated cell cycle regulation .

  • Genome-wide association studies identify CAMK2N2 as a potential biomarker in breast and lung cancers .

Protein Interaction Network

CAMK2N2 interacts with key neuronal and calcium-signaling components (STRING-db) :

Interacting ProteinFunctionInteraction Score
CAMK2A/CAMK2BSynaptic plasticity, memory formation0.84
CALM3Calcium signal transduction0.68
RIMBP2Synaptic vesicle regulation0.47

Expression Profiles

  • Tissue Specificity: Highest in the prefrontal cortex and hippocampus .

  • Cell Lines: Overexpressed in LoVo colon adenocarcinoma cells, where it suppresses apoptosis .

DatasetKey Insight
GTEx Tissue ExpressionUpregulated in brain tissues
CCLE Cell Line CNVCopy number variations in 12% of cancer lines

Research Gaps and Future Directions

  • Therapeutic Potential: CAMK2N2 inhibitors could mitigate CAMK2 hyperactivity in epilepsy or schizophrenia .

  • Mechanistic Clarity: Structural studies are needed to resolve how CAMK2N2 traps calmodulin selectively .

Product Specs

Introduction
Calcium/Calmodulin-Dependent Protein Kinase II Inhibitor 2, also known as CAMK2N2, is a protein encoded by the CAMK2N2 gene. This protein acts as a cellular inhibitor of CaM-kinase II (CAMK2) and has been shown to regulate cell growth when overexpressed in colon adenocarcinoma LoVo cells.
Description
Recombinant human CAMK2N2, expressed in E. coli, is available as a non-glycosylated polypeptide chain. This protein consists of 102 amino acids (including a 23 amino acid His-tag at the N-terminus) and has a molecular weight of 11kDa. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
The product is a clear, colorless solution that has been sterilized by filtration.
Formulation
The provided solution contains 1mg/ml CAMK2N2 in a buffer consisting of 20mM Tris-HCl (pH 8.0), 0.15M NaCl, 10% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) can further enhance stability during long-term storage. It is important to avoid repeated freezing and thawing of the product.
Purity
The purity of CAMK2N2 is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Calcium/calmodulin-dependent protein kinase II inhibitor 2, CAMK2N2, CaM-KII inhibitory protein, CaM-KIIN, CAMKIIN, Calcium/calmodulin-dependent protein kinase II.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMSEILPY SEDKMGRFGA DPEGSDLSFS CRLQDTNSFF AGNQAKRPPK LGQIGRAKRV VIEDDRIDDV LKGMGEKPPS GV.

Q&A

What is CAMK2N2 and what is its basic function in the brain?

CAMK2N2, also known as CaMKIINβ, is an endogenous inhibitor of calcium/calmodulin-dependent kinase II (CaMKII), an enzyme critical for memory and long-term potentiation (LTP). It functions primarily as a regulatory protein that inhibits CaMKII activity, thereby potentially modulating synaptic plasticity and memory processes . The inhibitor acts by binding to CaMKII and reducing its enzymatic activity, particularly inhibiting the autophosphorylation of T286, which is essential for maintaining CaMKII's autonomous activity and synaptic targeting .

How does CAMK2N2 differ from CAMK2N1 in terms of expression patterns?

While both are endogenous CaMKII inhibitors, CAMK2N2 and CAMK2N1 show distinct patterns of expression following memory-related activities. Research has shown that Camk2n1 (but not Camk2n2) is upregulated 60 minutes after LTP induction in hippocampal CA3-CA1 connections . In contrast, after contextual fear conditioning, Camk2n1 is transiently upregulated in the hippocampus and amygdala at 30 minutes post-training, while CaMK2N2 protein levels increase for several hours afterward . This temporal difference suggests distinct regulatory roles in memory processes.

What experimental models are commonly used to study CAMK2N2 function?

Research on CAMK2N2 frequently employs rodent models, particularly in contextual fear conditioning paradigms. These experiments typically use:

  • Hippocampal slice preparations for LTP induction studies

  • Adeno-associated virus (rAAV) vectors for gene manipulation

  • Synapsin 1 promoter-based expression systems for targeted neuronal expression

  • Quantitative PCR for measuring mRNA expression changes

  • Western blot analyses for protein level quantification

  • Behavioral assessments including contextual fear conditioning

The dorsal hippocampus is often targeted in these studies due to its critical role in contextual memory formation.

What viral vector systems are most effective for manipulating CAMK2N2 expression in neural tissue?

For manipulating CAMK2N2 expression in neural tissue, adeno-associated virus (rAAV) bicistronic vectors have demonstrated effectiveness. These vectors typically employ:

  • The synapsin 1 promoter for neuron-specific expression

  • Independent expression cassettes for the human CAMK2N2 gene and fluorescent markers

  • Stereotaxic delivery methods targeting specific brain regions (often dorsal hippocampus)

When designing viral constructs, researchers should consider the promoter strength, viral serotype for optimal neural tropism, and inclusion of reporter genes for verification of transduction efficiency. Stereotaxic coordinates must be carefully selected to ensure targeting of the intended brain region without affecting adjacent structures.

How should time points be selected when studying CAMK2N2 expression changes after behavioral training?

Based on the temporal dynamics observed in previous studies, researchers should consider the following time points when studying CAMK2N2 expression changes:

  • Baseline (pre-training) measurements

  • 30 minutes post-training/induction (capturing immediate early gene responses)

  • 60 minutes post-training (when differential regulation between CAMK2N1 and CAMK2N2 is apparent)

  • 2-6 hours post-training (when protein level changes become more pronounced)

  • 24 hours post-training (for long-term changes)

The selection should be guided by whether the focus is on mRNA expression (earlier time points) or protein level changes (later time points). For memory retrieval experiments, similar time windows after the retrieval session should be considered.

What control conditions are essential when studying the effects of CAMK2N2 manipulation on memory?

When designing experiments to study CAMK2N2's role in memory, the following controls should be included:

  • Non-manipulated animals to establish baseline performance

  • Vector controls expressing only reporter molecules (e.g., GFP)

  • Scrambled shRNA controls for knockdown experiments

  • Time-matched controls without behavioral training

  • Brain region controls (e.g., ventral hippocampus vs. dorsal hippocampus)

  • Post-surgery recovery period controls to account for surgical effects

Additionally, researchers should verify successful viral transduction through fluorescence imaging and confirm target protein levels via Western blot or qPCR before interpreting behavioral results.

How does CAMK2N2 interact with CaMKII-dependent signaling pathways during memory formation versus retrieval?

CAMK2N2 appears to have distinct roles in memory formation versus retrieval processes. For memory formation, overexpression of CAMK2N2 prior to training blocks contextual memory formation, suggesting that inhibition of CaMKII activity during acquisition prevents proper memory encoding . This indicates that CaMKII activity is necessary during the learning phase.

In contrast, overexpression of CAMK2N2 after training has no effect on contextual long-term memory maintenance, suggesting that persistent CaMKII activity is not required for memory storage . This functional dissociation points to a complex temporal regulation of CaMKII signaling, where CAMK2N2 may serve as a negative regulator during encoding but play a different role during maintenance phases.

Methodologically, researchers investigating these differential roles should:

  • Use temporally controlled expression systems (e.g., inducible promoters)

  • Examine downstream effectors like GluA1 and c-fos

  • Analyze phosphorylation states of CaMKII at T286 after manipulation

  • Consider the subcellular localization of both CaMKII and its inhibitors

What is the significance of the correlation between LTP magnitude and CAMK2N1 expression, and how does this compare to CAMK2N2?

Research has demonstrated a significant positive correlation between LTP magnitude and Camk2n1 (but not Camk2n2) expression, with Spearman's coefficient analysis showing r = .943, p = .018 . This indicates that changes in Camk2n1 expression induced by synaptic activity are triggered in a graded manner, following a monotonic direct relationship with the percent fEPSP increase induced by high-frequency stimulation.

This correlation suggests that CAMK2N1 may function as part of a feedback mechanism that scales with synaptic strengthening, potentially serving to modulate subsequent plasticity (metaplasticity). In contrast, CAMK2N2 does not show this correlation, indicating fundamentally different regulatory mechanisms.

To investigate these differences, researchers should:

  • Perform dose-response experiments with varying stimulation intensities

  • Examine subregion-specific expression patterns using microdissection techniques

  • Assess the temporal relationship between CaMKII activation and inhibitor expression

  • Explore the molecular mechanisms linking synaptic strengthening to inhibitor expression

What methodological approaches can resolve contradictions in the literature regarding CAMK2N2's role in memory maintenance?

Several methodological approaches can help resolve apparent contradictions regarding CAMK2N2's role in memory maintenance:

  • Temporal specificity: Use temporally controlled expression systems (e.g., doxycycline-regulated promoters) to manipulate CAMK2N2 at precise time points during memory formation, consolidation, and retrieval.

  • Spatial specificity: Employ region-specific promoters or Cre-dependent expression in specific cell populations to determine whether CAMK2N2's effects vary across brain regions or cell types.

  • Dosage considerations: Implement a range of expression levels through titrated viral delivery to determine whether CAMK2N2's effects are concentration-dependent.

  • Molecular interaction analysis: Use co-immunoprecipitation and proximity ligation assays to examine direct interactions between CAMK2N2 and CaMKII under different conditions.

  • Functional readouts: Combine electrophysiological recordings with molecular manipulations to directly link CAMK2N2 levels to synaptic function.

These approaches should be complemented by comprehensive analysis of downstream molecular events, including examination of AMPA receptor trafficking, spine morphology changes, and protein synthesis pathways.

What are the challenges in distinguishing between the functions of CAMK2N1 and CAMK2N2 in human neuronal systems?

Distinguishing between CAMK2N1 and CAMK2N2 functions presents several methodological challenges:

  • Overlapping binding sites: Both inhibitors interact with CaMKII at similar sites, making selective pharmacological targeting difficult.

  • Compensatory mechanisms: Knockdown of one inhibitor may lead to compensatory upregulation of the other, confounding interpretations.

  • Temporal dynamics: The inhibitors show different expression kinetics following stimulation, requiring careful time-course analyses.

  • Subcellular localization: Determining the precise subcellular distribution of each inhibitor requires high-resolution imaging techniques.

  • Translation to human systems: Most studies have been conducted in rodent models, and translation to human neuronal systems requires careful validation.

Researchers can address these challenges by:

  • Using simultaneous knockdown approaches with rescue experiments

  • Employing super-resolution microscopy to track subcellular localization

  • Developing inhibitor-specific antibodies with minimal cross-reactivity

  • Utilizing human iPSC-derived neurons to validate findings from animal models

What are the most reliable methods for quantifying CAMK2N2 protein levels in specific neuronal compartments?

For reliable quantification of CAMK2N2 protein in specific neuronal compartments, researchers should consider:

  • Subcellular fractionation: Prepare synaptosomal, PSD, and cytosolic fractions through differential centrifugation followed by Western blotting with compartment-specific markers as controls.

  • Immunocytochemistry: Use confocal or super-resolution microscopy with validated antibodies against CAMK2N2 and compartment markers (PSD-95 for postsynaptic, synaptophysin for presynaptic regions).

  • Proximity ligation assay: Detect in situ protein interactions between CAMK2N2 and compartment-specific proteins.

  • Live imaging: Express fluorescently tagged CAMK2N2 to track its dynamics in different neuronal compartments over time.

  • Electron microscopy immunogold labeling: Provide nanometer-scale resolution of CAMK2N2 localization.

When using these techniques, appropriate controls should include:

  • Validation of antibody specificity using knockout or knockdown tissues

  • Quantification against housekeeping proteins specific to each compartment

  • Cross-validation using multiple independent methods

How can researchers differentiate between the direct inhibitory effects of CAMK2N2 on CaMKII enzymatic activity versus its influence on CaMKII-GluN2B binding?

Differentiating between CAMK2N2's effects on CaMKII enzymatic activity versus CaMKII-GluN2B binding requires specialized methodological approaches:

  • In vitro kinase assays: Measure CaMKII enzymatic activity with purified components in the presence of varying CAMK2N2 concentrations, using synthetic substrates or autophosphorylation as readouts.

  • Pull-down assays: Assess CaMKII-GluN2B binding using purified components with and without CAMK2N2, analyzing binding through co-immunoprecipitation or surface plasmon resonance.

  • Structure-function analyses: Use mutated forms of CAMK2N2 that selectively disrupt either enzymatic inhibition or GluN2B binding interference.

  • Dose-response experiments: Compare the concentration-dependency of enzymatic inhibition versus binding interference, as research indicates larger concentrations of inhibitor are required to disrupt CaMKII-GluN2B binding than to inhibit enzymatic activity .

  • Live cell FRET sensors: Deploy fluorescence resonance energy transfer sensors to monitor CaMKII-GluN2B interactions in real-time while manipulating CAMK2N2 levels.

These approaches should be employed in combination to build a comprehensive understanding of CAMK2N2's dual mechanisms of action.

What are the promising approaches for targeting CAMK2N2 in neurological disorder research?

Several promising approaches for targeting CAMK2N2 in neurological disorder research include:

  • Cell-type specific manipulation: Using Cre-dependent viral vectors to manipulate CAMK2N2 expression in specific neuronal populations implicated in neurological disorders.

  • Temporal regulation: Employing chemogenetic or optogenetic approaches to control CAMK2N2 function with precise temporal resolution during critical disease periods.

  • Peptide-based therapeutics: Developing membrane-permeable peptides based on CAMK2N2 structure that can modulate CaMKII activity in a controlled manner.

  • Small molecule screening: Identifying compounds that can specifically modulate the interaction between CAMK2N2 and CaMKII without affecting other CaMKII functions.

  • Gene therapy approaches: Exploring viral vector-based delivery of modified CAMK2N2 to restore normal CaMKII regulation in disorders characterized by dysregulated calcium signaling.

These approaches should be evaluated in relevant disease models, with careful attention to potential off-target effects and compensatory mechanisms that might emerge with chronic manipulation of the CaMKII regulatory system.

How might single-cell transcriptomics advance our understanding of CAMK2N2 expression patterns in different neuronal populations?

Single-cell transcriptomics offers several advantages for understanding CAMK2N2 expression patterns:

  • Cell-type specific expression profiling: Identifying which neuronal subtypes express CAMK2N2 at baseline and how this changes with activity.

  • Co-expression networks: Determining which genes are co-regulated with CAMK2N2, potentially revealing functional relationships and regulatory pathways.

  • Activity-dependent regulation: Capturing the heterogeneity in CAMK2N2 expression changes following neuronal activation across different cell populations.

  • Developmental trajectories: Mapping how CAMK2N2 expression changes throughout neuronal maturation and circuit formation.

  • Disease-associated alterations: Identifying cell-specific dysregulation of CAMK2N2 in neurological or psychiatric conditions.

Methodologically, researchers should:

  • Use validated neuronal markers for cell type identification

  • Implement computational approaches to account for technical noise

  • Integrate spatial information through spatial transcriptomics

  • Validate findings with in situ hybridization or immunohistochemistry

  • Combine with functional assays to link expression patterns to neuronal activity

What experimental design would best determine if CAMK2N2 functions as a metaplasticity regulator across different memory systems?

To investigate CAMK2N2's potential role as a metaplasticity regulator across memory systems, an optimal experimental design would include:

  • Multi-region analysis: Simultaneous manipulation and recording in multiple memory-related brain regions (hippocampus, amygdala, prefrontal cortex) to compare CAMK2N2's effects across systems.

  • Sequential learning paradigms: Implement behavioral protocols where animals learn one task followed by another, while manipulating CAMK2N2 expression between learning events.

  • Electrophysiological assessment: Perform in vivo recordings during and after learning to measure how CAMK2N2 manipulation affects subsequent plasticity induction at the same synapses.

  • Molecular priming: Examine how prior manipulation of CAMK2N2 alters the molecular response to subsequent learning, focusing on plasticity-related proteins.

  • Reversible manipulations: Use temporally controlled, reversible manipulation of CAMK2N2 function through optogenetics or chemogenetics.

The experimental design should incorporate:

  • Within-subject controls where possible

  • Careful timing of manipulations relative to learning events

  • Dose-dependency analyses to detect threshold effects

  • Multiple behavioral readouts to assess different memory components

  • Correlation analyses between molecular, electrophysiological, and behavioral measures

What statistical approaches are most appropriate for analyzing the relationship between CAMK2N2 expression levels and behavioral outcomes?

For analyzing relationships between CAMK2N2 expression and behavioral outcomes, researchers should consider:

  • Correlation analyses: Both Pearson and Spearman correlations can assess relationships between CAMK2N2 levels and behavioral metrics. Spearman's coefficient analysis has previously revealed significant correlations between gene expression changes and LTP magnitude (r = .943, p = .018) .

  • Regression models: Linear and non-linear regression can determine the predictive value of CAMK2N2 levels for behavioral outcomes while controlling for confounding variables.

  • Mixed-effects models: Account for both fixed effects (manipulation conditions) and random effects (individual differences between animals).

  • Path analysis: Test causal hypotheses about how CAMK2N2 mediates relationships between neuronal activity and behavioral outcomes.

  • Bayesian approaches: Incorporate prior knowledge about CAMK2N2 function while updating models based on new experimental data.

Researchers should control for:

  • Multiple comparisons when analyzing multiple brain regions or time points

  • Inter-individual variability in baseline CAMK2N2 expression

  • Behavioral variability unrelated to the manipulation

  • Potential non-linear relationships between expression and function

How should researchers interpret apparent contradictions in CAMK2N2 function between different brain regions or memory tasks?

When faced with contradictions in CAMK2N2 function across brain regions or memory tasks, researchers should:

  • Consider circuit-specific functions: CAMK2N2 may have different roles depending on the neural circuit in which it operates. The protein might promote plasticity in some circuits while constraining it in others.

  • Examine task demands: Different memory tasks engage distinct cognitive processes, and CAMK2N2's role may vary depending on the specific computational requirements.

  • Account for temporal dynamics: Apparent contradictions might reflect different time points in the expression and function of CAMK2N2 after learning or memory retrieval.

  • Assess concentration-dependent effects: CAMK2N2 might have opposing effects at different expression levels, possibly through differential effects on CaMKII enzymatic activity versus structural functions.

  • Evaluate interaction with other molecular players: Regional differences in other signaling molecules might alter how CAMK2N2 affects memory processes.

Methodologically, researchers should:

  • Use within-subject designs where possible to control for individual variability

  • Implement parallel manipulations across brain regions with identical methods

  • Combine regional manipulations with circuit-specific activity measurements

  • Consider compensatory mechanisms that might emerge in different regions

How can findings from rodent studies on CAMK2N2 be most effectively translated to human brain function and pathology?

To effectively translate CAMK2N2 findings from rodent studies to human applications:

  • Human tissue validation: Verify expression patterns and regulatory mechanisms in post-mortem human brain tissue and compare with rodent findings.

  • iPSC-derived neuronal models: Generate human neurons from induced pluripotent stem cells to test whether CAMK2N2 mechanisms observed in rodents are conserved.

  • Genetic association studies: Examine whether polymorphisms in the CAMK2N2 gene or its regulatory regions are associated with cognitive function or neuropsychiatric disorders in humans.

  • Neuroimaging correlates: Identify functional or structural neuroimaging correlates that might reflect CAMK2N2-dependent processes established in animal models.

  • Cross-species behavioral paradigms: Develop parallel behavioral tasks that can be administered to both rodents and humans to bridge translational gaps.

Researchers should be particularly careful to:

  • Account for species differences in CaMKII expression and regulation

  • Consider the increased complexity of human neural circuits

  • Acknowledge the longer timescales of human development and aging

  • Evaluate the influence of genetic and environmental factors unique to humans

What methodological approaches would best determine if CAMK2N2 dysfunction contributes to specific neuropsychiatric conditions?

To investigate potential contributions of CAMK2N2 dysfunction to neuropsychiatric conditions:

  • Case-control studies: Compare CAMK2N2 expression, localization, and function in post-mortem brain tissue from patients versus matched controls.

  • Genetic analyses: Perform targeted sequencing of CAMK2N2 in patient populations to identify potential disease-associated variants.

  • Patient-derived models: Generate neurons from patient iPSCs to examine CAMK2N2 expression, localization, and function in a disease context.

  • Animal models of disease: Determine whether CAMK2N2 manipulation can ameliorate or exacerbate phenotypes in animal models of neuropsychiatric conditions.

  • Pharmacological studies: Test whether drugs that indirectly affect CAMK2N2 function have differential effects in patient populations.

Researchers should incorporate:

  • Careful phenotypic characterization of patient populations

  • Analysis of potential sex differences in CAMK2N2 function and dysfunction

  • Consideration of developmental trajectories in CAMK2N2 expression and function

  • Examination of gene-environment interactions that might modulate CAMK2N2 effects

What are the critical quality control measures for validating CAMK2N2 antibodies in experimental protocols?

Critical quality control measures for CAMK2N2 antibody validation include:

  • Knockout/knockdown controls: Test antibodies on tissues or cells with confirmed CAMK2N2 knockout or knockdown to verify specificity.

  • Western blot validation: Confirm a single band of appropriate molecular weight (approximately 8-10 kDa for CAMK2N2).

  • Peptide competition assays: Preincubate antibodies with the immunizing peptide to demonstrate specific blocking of signal.

  • Cross-reactivity testing: Evaluate potential cross-reactivity with the closely related CAMK2N1 protein through parallel testing.

  • Multiple antibody comparison: Use at least two antibodies targeting different epitopes of CAMK2N2 to confirm staining patterns.

  • Recombinant protein standards: Include purified recombinant CAMK2N2 protein as a positive control for quantification.

  • Species validation: Confirm that antibodies recognize the target protein across all species used in the research program.

These validation steps should be systematically documented and reported in publications to ensure reproducibility across research groups.

What considerations are important when designing primers for qPCR analysis of CAMK2N2 expression?

When designing primers for qPCR analysis of CAMK2N2 expression, researchers should consider:

  • Isoform specificity: Design primers that either specifically target individual isoforms or span common regions to detect all isoforms.

  • Exon junction spanning: Create primers that span exon-exon junctions to prevent amplification of genomic DNA.

  • Primer efficiency testing: Verify primer efficiency using standard curves with 90-110% efficiency for accurate quantification.

  • Reference gene selection: Carefully select stable reference genes that are not affected by the experimental conditions.

  • Cross-reactivity prevention: Ensure primers do not amplify the related CAMK2N1 gene by performing specific checks against sequence alignments.

  • Melting curve analysis: Perform melting curve analysis after each qPCR run to confirm a single specific amplification product.

  • Amplicon size optimization: Design primers to generate amplicons of 70-200 bp for optimal PCR efficiency.

  • Validation with sequencing: Confirm the identity of qPCR products by sequencing to ensure target specificity.

These considerations will help ensure reliable and reproducible quantification of CAMK2N2 expression levels across experimental conditions.

Product Science Overview

Introduction

Calcium/Calmodulin-Dependent Protein Kinase II (CaMKII) is a multifunctional serine/threonine kinase that plays a pivotal role in various cellular processes, including synaptic plasticity, memory formation, and cardiac function. The enzyme is activated by the binding of calcium/calmodulin and is known for its ability to phosphorylate a wide range of substrates. Inhibitors of CaMKII, such as the human recombinant CaMKII Inhibitor 2, are crucial tools for studying the enzyme’s function and potential therapeutic applications.

Structure and Function of CaMKII

CaMKII is a holoenzyme composed of multiple subunits, each containing a catalytic domain, a regulatory domain, and an association domain. The enzyme is activated when calcium-bound calmodulin binds to the regulatory domain, causing a conformational change that exposes the catalytic site. This activation allows CaMKII to phosphorylate various target proteins, influencing numerous cellular pathways.

Role of CaMKII in Cellular Processes

CaMKII is highly expressed in the brain, where it is involved in synaptic plasticity and memory formation. It is also present in cardiac tissue, where it regulates calcium handling and cardiac contractility. Dysregulation of CaMKII activity has been implicated in various diseases, including Alzheimer’s disease, heart failure, and diabetes .

CaMKII Inhibitors

CaMKII inhibitors, such as the human recombinant CaMKII Inhibitor 2, are essential for dissecting the enzyme’s role in cellular processes. These inhibitors can be used to study the effects of CaMKII inhibition on cellular function and to explore potential therapeutic applications. For example, CaMKII inhibitors have been shown to restore cardiac contractility and relaxation in diabetic hearts, suggesting a potential role in treating diabetic cardiomyopathy .

Human Recombinant CaMKII Inhibitor 2

The human recombinant CaMKII Inhibitor 2 is a synthetic peptide designed to specifically inhibit CaMKII activity. This inhibitor is produced using recombinant DNA technology, which allows for the generation of highly pure and consistent peptide products. The inhibitor works by binding to the catalytic domain of CaMKII, preventing its activation and subsequent phosphorylation of target proteins.

Applications in Research and Medicine

Human recombinant CaMKII Inhibitor 2 is widely used in research to study the role of CaMKII in various cellular processes. It has been employed in studies investigating the enzyme’s involvement in synaptic plasticity, memory formation, and cardiac function. Additionally, CaMKII inhibitors are being explored as potential therapeutic agents for diseases associated with CaMKII dysregulation, such as Alzheimer’s disease, heart failure, and diabetes .

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.