CALM2 Human 135 a.a

Calmodulin-2 135 a.a. Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to CALM2 Human 135 a.a

CALM2 Human 135 a.a refers to a recombinant protein corresponding to the full-length human calmodulin 2 (CALM2) gene product. This 135-amino acid protein (UniProt ID: P0DP24) is expressed in Escherichia coli and serves as a critical tool for studying calcium-mediated signaling pathways, ion channel regulation, and calmodulinopathy-related diseases . With a molecular weight of ~16 kDa, it retains the calcium-binding properties essential for its role as a multifunctional intracellular calcium sensor .

Functional Role in Calcium Signaling

CALM2 Human 135 a.a regulates calcium-dependent processes by interacting with:

  • Ion Channels: Modulates L-type calcium channels (CACNA1C) and ryanodine receptors (RyR2) .

  • Enzymes: Activates calmodulin-dependent kinases (CaMKII) and phosphatases .

  • Structural Proteins: Interacts with centrosomal proteins (e.g., CCP110) during cytokinesis .

Mutations in CALM2 disrupt Ca²⁺ binding, leading to severe arrhythmias such as long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT) .

Pathogenic Mutations

Over 30 CALM2 mutations (e.g., D130G, N98S) have been linked to calmodulinopathies. Key findings include:

MutationPhenotypeFunctional DefectCitation
D130GLQTS + neurodevelopmental issuesImpaired Ca²⁺-dependent inactivation of CACNA1C
N98SSevere LQTSReduced Ca²⁺ binding affinity (Kd = 150 ± 30 μM vs. WT = 27 ± 5 μM)
Q136PLQTS/CPVT overlapDisrupted RyR2 regulation, leading to diastolic Ca²⁺ leakage

Therapeutic Insights

  • CRISPR-mediated suppression of mutant CALM2 alleles restores normal action potential duration in iPSC-derived cardiomyocytes .

  • β-blockers (e.g., propranolol) reduce exertion-triggered arrhythmias in 74% of patients .

Industrial and Clinical Relevance

  • Drug Discovery: Used to screen small molecules targeting Ca²⁺/CALM2 interactions for arrhythmia therapies .

  • Structural Studies: Facilitates crystallography and NMR analyses of calcium-binding dynamics .

  • Diagnostics: Serves as an antigen for antibody development in calmodulinopathy testing .

Limitations and Future Directions

  • Challenges: Low stability in non-reducing buffers limits long-term storage .

  • Opportunities: Engineering thermostable CALM2 variants could enhance experimental reproducibility .

Product Specs

Introduction
Calmodulin (CaM) is a calcium-binding protein found inside cells. It plays a crucial role in regulating various cellular processes by binding to and activating other proteins, particularly enzymes. For instance, it activates myosin light chain kinase, which is responsible for myosin phosphorylation. CaM is involved in activating enzymes such as cyclic nucleotide-dependent phosphodiesterase, calcineurin, ATPase, Myosin Light Chain Kinases, and CAM kinase.
Description
This product consists of the full-length CALM2 protein, derived from humans and produced in E. coli bacteria. The protein encompasses 135 amino acids and has an approximate molecular weight of 16 kDa.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The CALM2 protein is supplied at a concentration of 1mg/ml in a solution containing 20mM HEPES-KOH at pH 7, 50mM NaCl, 1mM EDTA, and 1mM DTT.
Stability
For long-term storage, keep the vial at temperatures between -20°C and -80°C. Under these conditions, the protein remains stable for up to 12 months. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the protein is greater than 90%, as assessed by SDS-PAGE analysis.
Synonyms
CALM2, CAM2, CAMB, CAMII, PHKD2, calmodulin 2.
Source
Escherichia Coli.
Amino Acid Sequence
MADQLTEEQI AEFKEAFSLF DKDGDGTITT KELGTVMRSL GQNPTEAELQ DMINEVDADG NGTIDFPEFL TMMARKMKDT DSEEEIREAF RVFDKDGNGY ISAAELRHVM TNLGEKLTDE EVDEMIREAD IDGDGQ.

Q&A

What is the relationship between CALM1, CALM2, and CALM3 genes in humans?

The human genome contains three independent calmodulin genes (CALM1, CALM2, and CALM3) that are all functional and encode the exact same 135 amino acid calmodulin protein. This unusual genetic redundancy underscores the critical importance of maintaining precise calmodulin levels in cells. The identical protein product from these three genes exhibits extraordinary evolutionary conservation, with the amino acid sequence completely preserved across all vertebrates, highlighting the essential nature of calmodulin's precise structure for its diverse functions . When designing experiments targeting CALM2 specifically, researchers must employ gene-specific primers or probes that target unique untranslated regions, as the coding sequences are identical across all three genes.

How is calmodulin involved in calcium signaling pathways?

Calmodulin functions as a primary intracellular calcium sensor that detects local changes in Ca²⁺ concentration and relays this information to hundreds of interaction partners. Upon binding calcium ions, calmodulin undergoes conformational changes that enable it to interact with and regulate numerous target proteins including ion channels, enzymes, and transcription factors. This conformational flexibility allows calmodulin to function as a versatile signal transducer in calcium-dependent pathways . Experimentally, this can be studied using calcium imaging techniques combined with FRET-based sensors that detect calmodulin-target interactions in real-time within living cells.

What experimental approaches are most effective for studying CALM2 expression?

For differential analysis of CALM2 expression:

  • Gene-specific RT-qPCR targeting unique UTR regions

  • RNA-seq with specific mapping parameters to distinguish between the three genes

  • In situ hybridization with gene-specific probes

  • CRISPR-based tagging of endogenous CALM2 for visualization

When analyzing expression data, researchers should normalize to appropriate housekeeping genes stable under the experimental conditions and include controls to verify gene-specific amplification, as cross-reactivity is a common technical challenge when studying highly similar genes.

What mutations in CALM2 have been identified in human disease, and what are their functional consequences?

Multiple pathogenic mutations in CALM2 have been identified in patients with severe cardiac arrhythmias:

MutationClinical PresentationFunctional EffectAge of Onset
N98SLQTSAltered Ca²⁺ bindingEarly childhood
N98ILQTSAltered Ca²⁺ bindingEarly childhood
D134HLQTSAltered Ca²⁺ bindingEarly childhood
D132ELQTS and CPVTImpaired CaV1.2 and RyR2 regulationEarly childhood
Q136PLQTS and CPVTImpaired CaV1.2 and RyR2 regulationEarly childhood
D130GLQTSDecreased Ca²⁺ binding affinityEarly childhood

These mutations predominantly affect the calcium-binding domains of calmodulin, resulting in altered calcium binding affinity and disrupted regulation of cardiac ion channels. Unlike mutations in CALM1 or CALM3, mutations in CALM2 tend to more commonly present with mixed LQTS and CPVT phenotypes . When studying these mutations, it is critical to consider that they exist in a heterozygous state in patients, where wildtype calmodulin is still produced from five other alleles (the unaffected CALM2 allele plus both alleles of CALM1 and CALM3).

How can researchers effectively model CALM2 mutations in experimental systems?

For modeling CALM2 mutations, multiple complementary approaches provide robust data:

  • Biochemical characterization: Purified recombinant wildtype and mutant calmodulin proteins should be analyzed for calcium binding using isothermal titration calorimetry and fluorescence spectroscopy.

  • Cellular systems: CRISPR/Cas9-mediated knock-in of mutations in relevant cell types (cardiomyocytes, neurons) while preserving endogenous expression levels.

  • iPSC-derived models: Patient-derived iPSCs or gene-edited iPSCs differentiated into cardiomyocytes provide a clinically relevant model system .

  • Target interaction assays: Co-immunoprecipitation and FRET-based interaction studies with key targets (CaV1.2, RyR2) under varying calcium concentrations.

When designing these experiments, researchers should consider the stoichiometry of wildtype to mutant calmodulin, as this ratio significantly impacts experimental outcomes and better represents the in vivo situation where both normal and mutant forms coexist.

Why are CALM2 mutations predominantly associated with cardiac phenotypes despite calmodulin's ubiquitous expression?

This question represents an ongoing research puzzle. Several hypotheses exist:

  • Screening bias hypothesis: Most mutations have been identified through cardiac phenotype screening programs, potentially missing non-cardiac manifestations .

  • Tissue sensitivity hypothesis: Cardiac tissue may be especially sensitive to disruptions in calcium handling, given the central role of calcium in excitation-contraction coupling.

  • Functional redundancy hypothesis: Non-cardiac tissues may have compensatory mechanisms that mitigate the effects of calmodulin mutations.

  • Target-specific dysregulation: The particular calmodulin targets disrupted by these mutations (CaV1.2 and RyR2) have especially critical roles in cardiac function.

To investigate this question methodically, researchers should consider comprehensive phenotyping of mutation carriers, including neurological, immunological, and metabolic assessments, as calmodulin regulates critical processes in these systems as well. Additionally, tissue-specific knock-in mouse models would help determine if certain tissues are inherently more sensitive to calmodulin mutations .

What are the most effective methods for studying calmodulin-target interactions in the context of CALM2 mutations?

For robust characterization of calmodulin-target interactions:

  • In vitro binding assays: Surface plasmon resonance (SPR) and microscale thermophoresis (MST) to quantify binding affinities under varying calcium concentrations.

  • Structural studies: X-ray crystallography or cryo-EM of calmodulin-target complexes to visualize binding interfaces.

  • Live-cell interaction studies: FRET/BRET sensors with genetically encoded calcium indicators to simultaneously monitor calcium levels and calmodulin-target binding.

  • Proximity labeling approaches: BioID or APEX2 fused to calmodulin variants to identify the interactome in living cells under different conditions.

  • Electrophysiological recordings: Patch-clamp studies of ion channels (CaV1.2, RyR2) in the presence of wildtype versus mutant calmodulin.

These methodologies should be employed with proper controls, including calcium-binding deficient calmodulin mutants (e.g., with EF-hand mutations) as reference standards .

How can researchers overcome the challenge of studying CALM2 specifically given the identical protein product from all three CALM genes?

This represents a significant technical challenge requiring multifaceted approaches:

  • Gene-specific knockdown/knockout: Use siRNAs, shRNAs, or CRISPR targeting unique UTR regions of CALM2.

  • Allele-specific expression analysis: Employ SNP-based approaches or digital PCR to distinguish transcripts.

  • Rescue experiments: Knockdown all three genes followed by selective re-expression of CALM2.

  • Gene-edited cellular models: CRISPR-based introduction of tags or mutations specifically in the CALM2 locus.

  • Patient-derived cells: Utilize cells from individuals with identified CALM2 mutations for functional studies.

A comprehensive approach combining genetic manipulation with functional readouts provides the most reliable data. Validation of gene-specific targeting should be demonstrated using multiple methods, as cross-reactivity remains a significant concern .

What advanced techniques are available for studying calcium-dependent structural changes in calmodulin with CALM2 mutations?

Several sophisticated biophysical techniques can elucidate structural impacts of mutations:

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): Provides data on conformational dynamics and solvent accessibility changes upon calcium binding.

  • Single-molecule FRET: Reveals population distributions of different conformational states and transition kinetics between them.

  • NMR spectroscopy: Yields residue-specific information on structural perturbations and dynamics in solution.

  • Molecular dynamics simulations: Complements experimental approaches by predicting conformational changes and energetics at atomic resolution.

  • Cryo-electron microscopy: For visualizing calmodulin-target complexes, especially with larger interaction partners.

These techniques should be applied across a range of calcium concentrations (from nM to μM) to capture the full spectrum of calmodulin's conformational landscape and how mutations alter this energy landscape .

How do CALM2 mutations differentially affect regulation of CaV1.2 versus RyR2 channels?

CALM2 mutations show target-specific effects on ion channel regulation:

  • CaV1.2 regulation: Calmodulin acts as both a calcium sensor for calcium-dependent inactivation (CDI) and facilitates calcium-dependent facilitation (CDF). LQTS-associated mutations primarily disrupt CDI, leading to prolonged calcium influx and QT interval prolongation. Experimental approaches include patch-clamp recordings of calcium currents in heterologous expression systems or cardiomyocytes, with specific protocols to isolate CDI and CDF components .

  • RyR2 regulation: Calmodulin inhibits RyR2 activity in both calcium-free and calcium-bound states, though through different mechanisms. CPVT-associated mutations often reduce this inhibitory effect, leading to inappropriate calcium release from the sarcoplasmic reticulum. Single-channel recordings, calcium spark analysis, and [³H]ryanodine binding assays can quantify these effects .

Researchers should design experiments that can directly compare effects on both channels within the same cellular context, as differential regulation might explain mixed clinical phenotypes observed with some mutations.

What therapeutic strategies show promise for treating calmodulinopathies caused by CALM2 mutations?

Several approaches show potential based on understanding of molecular mechanisms:

  • Allele-specific silencing: CRISPR/Cas9 or antisense oligonucleotides targeting the mutant CALM2 allele specifically. Experimental evidence shows that silencing the mutant allele in patient-derived cardiomyocytes can restore normal calcium channel function .

  • Target-specific therapies: Beta-blockers and sodium channel blockers have shown effectiveness in controlling symptoms in patients with CALM2 mutations .

  • Calcium channel modulators: Compounds that can compensate for defective CDI of CaV1.2 channels may be beneficial for LQTS patients.

  • RyR2 stabilizers: For patients with CPVT phenotypes, RyR2 stabilizers may reduce inappropriate calcium release.

  • Gene therapy approaches: Delivery of wildtype CALM genes to increase the ratio of functional to mutant calmodulin.

When designing preclinical studies, researchers should consider the heterozygous nature of these mutations and the presence of wildtype calmodulin from other alleles .

How do CALM2 mutations outside calcium-binding domains affect protein function and disease pathogenesis?

While most pathogenic CALM2 mutations affect calcium-binding EF-hand domains, mutations outside these regions present unique research opportunities:

Research approaches should include stability assays (differential scanning fluorimetry, proteolytic susceptibility), interactome analysis comparing wildtype and mutant forms, and functional assays for specific target regulation. Additionally, computational prediction of mutation effects through molecular dynamics simulations can guide experimental design .

What non-cardiac phenotypes might be associated with CALM2 mutations that are currently overlooked?

Given calmodulin's ubiquitous role, several non-cardiac manifestations warrant investigation:

  • Neurological phenotypes: CaV1.2 and CaV2 channels regulated by calmodulin play crucial roles in neuronal function. Subtle neurological phenotypes might be masked by the severe cardiac manifestations or misattributed to secondary effects of cardiac episodes .

  • Immune dysfunction: Calmodulin regulates calcium signaling in immune cells, suggesting potential immunological phenotypes.

  • Metabolic abnormalities: Calmodulin regulates multiple metabolic enzymes, warranting investigation of glucose homeostasis and lipid metabolism.

  • Developmental effects: Given calmodulin's role in cell division and differentiation, subtle developmental phenotypes might exist.

Research approaches should include comprehensive phenotyping of mutation carriers, development of tissue-specific conditional knock-in models, and collaboration with specialties beyond cardiology to identify subtle non-cardiac manifestations .

How do rare variants in CALM2 identified in population databases contribute to disease risk?

The GnomAD database contains several rare CALM2 variants not currently associated with arrhythmias, raising important questions:

  • Subclinical phenotypes: These variants might confer subtle phenotypes or disease risk not severe enough to cause overt clinical presentation.

  • Conditional pathogenicity: May cause disease only under specific environmental conditions or genetic backgrounds.

  • Modified calcium affinity: Even small changes in calcium binding could alter signaling under specific physiological stress conditions.

To investigate these variants, researchers should perform comprehensive functional characterization comparing them to known pathogenic mutations and wildtype calmodulin. Additionally, association studies in large cohorts with detailed phenotyping could reveal subtle genotype-phenotype correlations .

What are the methodological considerations for developing gene therapy approaches for CALM2 mutations?

Developing gene therapies for CALM2 mutations requires addressing several challenges:

  • Allele-specific targeting: Methods for selectively silencing or correcting the mutant allele while preserving wildtype expression, such as CRISPR/Cas9 with appropriate guide RNA design or antisense oligonucleotides.

  • Dosage considerations: Given that calmodulin levels are tightly regulated, determining optimal expression levels for therapeutic intervention is critical.

  • Delivery systems: Cardiac-specific delivery vectors with appropriate tropism and efficiency, particularly for reaching cardiomyocytes.

  • Safety assessment: Evaluating off-target effects and potential consequences in non-cardiac tissues.

Proof-of-concept studies have demonstrated that silencing mutant CALM2 alleles can restore normal calcium channel function in patient-derived cardiomyocytes, suggesting the viability of this approach . Future research should focus on optimizing delivery methods and assessing long-term efficacy and safety.

What are the most significant unanswered questions in CALM2 research?

Despite substantial progress, several fundamental questions remain:

  • Why do identical mutations in different CALM genes (CALM1, CALM2, CALM3) sometimes produce different clinical phenotypes?

  • What determines the tissue-specific manifestations of ubiquitously expressed calmodulin mutations?

  • How does the complex interplay between wildtype and mutant calmodulin influence disease severity and progression?

  • What are the long-term effects of calmodulin mutations on cellular proteostasis and calcium homeostasis?

  • Are there protective factors that explain incomplete penetrance observed with some CALM2 mutations?

Addressing these questions requires interdisciplinary approaches combining clinical genetics, biochemistry, structural biology, electrophysiology, and systems biology. Longitudinal studies of mutation carriers and development of more sophisticated animal models will be particularly valuable .

How can researchers better integrate multi-omics data in studying CALM2 function and dysfunction?

To comprehensively understand CALM2 biology:

  • Integrative genomics: Combining genome, transcriptome, and epigenome data to understand CALM2 regulation across tissues and conditions.

  • Proteomics approaches: Global and targeted proteomics to map the calmodulinome and how it changes with mutations.

  • Metabolomics integration: Linking calmodulin dysfunction to metabolic perturbations that may contribute to disease phenotypes.

  • Single-cell multi-omics: Characterizing heterogeneity in calmodulin function and mutation effects at the single-cell level.

  • Computational modeling: Developing mathematical models that integrate multi-omics data to predict system-level effects of CALM2 mutations.

These approaches should be applied in relevant model systems, including patient-derived cells and tissues, to capture physiologically relevant contexts .

Product Science Overview

Structure and Function

Calmodulin-2 is composed of 135 amino acids and has a molecular weight of approximately 16 kDa . The protein contains four functional Ca²⁺-binding sites, known as EF-hands, which are crucial for its ability to bind calcium ions . These binding sites are located at the following amino acid positions:

  • aa 20-31
  • aa 56-67
  • aa 93-104
  • aa 129-140

Upon binding to Ca²⁺, calmodulin undergoes a conformational change that allows it to interact with and regulate a variety of target proteins and enzymes. Some of the key enzymes activated by calmodulin include:

  • Myosin Light Chain Kinase (MLCK)
  • Cyclic Nucleotide-Dependent Phosphodiesterase
  • Calcineurin
  • ATPase
  • CAM Kinase
Biological Significance

Calmodulin-2 is involved in numerous cellular processes, including muscle contraction, cell division, and signal transduction. It acts as an intracellular receptor for Ca²⁺ ions, translating calcium signals into cellular responses. This makes it a crucial component in the regulation of various physiological functions.

Recombinant Production

Recombinant human calmodulin-2 is typically expressed in Escherichia coli (E. coli) and purified for research and therapeutic applications . The recombinant protein is often tagged with a His•Tag sequence at the N-terminus to facilitate purification and detection . It is available in various forms, including full-length proteins and specific fragments, depending on the research requirements.

Applications

Recombinant calmodulin-2 is widely used in biochemical and biophysical studies to understand its interaction with target proteins and its role in cellular processes. It is also employed in drug discovery and development, particularly in the context of diseases where calcium signaling is disrupted.

Storage and Stability

Recombinant calmodulin-2 should be stored at -20°C to -80°C to maintain its stability. It is important to avoid repeated freeze-thaw cycles to prevent degradation . When stored properly, the protein remains stable for up to 12 months.

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.