Fact-checked by Grok 2 weeks ago

CALM2

CALM2 is a protein-coding in humans that encodes 2, an essential calcium-sensing protein identical in sequence to those produced by the paralogous genes CALM1 and CALM3, which together mediate intracellular by binding Ca²⁺ ions and modulating the activity of numerous enzymes, ion channels, and other effectors involved in processes such as , , and neuronal signaling. The is situated on and produces a 149-amino-acid polypeptide that, upon calcium binding, undergoes a conformational shift enabling interactions with diverse partners, thereby transducing calcium signals into physiological responses. Calmodulin's structure consists of two globular N- and C-terminal domains, each containing two EF-hand motifs for high-affinity calcium coordination, connected by a flexible central that facilitates in both calcium-saturated and apo forms. Notable disease associations include rare germline mutations in CALM2 linked to life-threatening cardiac arrhythmias, such as type 15 and , where altered calcium handling disrupts regulation, particularly of voltage-gated calcium channels like CaV1.2.

Gene Characteristics

Genomic Location and Structure

The CALM2 gene resides on the short arm of at cytogenetic band 2p21. In the GRCh38.p14 , it occupies genomic coordinates 2:47,160,082-47,176,936 on the reverse strand. This positioning places CALM2 within a gene-dense region of , distinct from its paralogs CALM1 (on 14q32.2) and CALM3 (on 19q13.2). The gene spans approximately 16.9 kilobases (kb) of DNA, from the transcription start site to the polyadenylation signal. Structurally, CALM2 comprises 6 exons separated by 5 introns, encoding the 149-amino-acid calmodulin protein through alternative splicing variants that maintain the core coding exons. The exon-intron boundaries are conserved across the three human calmodulin genes, with introns interrupting the coding sequence at equivalent positions corresponding to structural domains of the protein; however, CALM2 introns are notably larger, contributing to its extended genomic footprint compared to CALM1 (∼9 kb) and CALM3 (∼8 kb). This architecture was elucidated through genomic library screening and PCR amplification of intron-spanning fragments. The 5' flanking region includes a TATA-like sequence and potential regulatory elements, such as GC-rich motifs, influencing basal transcription.

Expression and Regulation

The CALM2 gene, encoding one of three isoforms of the identical protein, demonstrates ubiquitous basal expression across human tissues, reflecting calmodulin's fundamental role in calcium-mediated signaling. However, the three paralogous genes (CALM1, CALM2, CALM3) exhibit differential expression patterns influenced by tissue type, developmental stage, and stimuli, allowing nuanced control of calmodulin levels despite producing the same protein. In the human heart, CALM1 and CALM2 mRNAs predominate, collectively contributing four-fold more to total calmodulin transcript levels than CALM3, which constitutes a minor fraction. This skew supports higher calmodulin demands in cardiac tissue for excitation-contraction . Transcriptional regulation of CALM2 features distinct promoter strengths and (UTR) sequences that dictate mRNA abundance and translational efficiency. In proliferating human teratoma cells, CALM2 displays lower transcriptional activity and mRNA levels compared to CALM1 and CALM3, with its extended inhibiting translation more potently, thereby fine-tuning protein output. These differences arise from sequence variations in regulatory elements, enabling tissue- and context-specific expression without altering the coding sequence. Post-transcriptional mechanisms further modulate CALM2 expression, including mRNA stability and (miRNA) targeting. CALM2 transcripts possess a predicted shorter relative to CALM1 and CALM3, facilitating rapid adjustments to fluctuating calcium signals or stressors. In states, such as , miR-651-5p suppresses CALM2 expression, curbing , , and , which highlights pathological dysregulation. Conversely, CALM2 upregulation in tissues correlates with advanced and reduced overall survival, implicating aberrant transcriptional or epigenetic controls in oncogenesis.

Protein Structure and Function

Molecular Structure

The protein product of the CALM2 gene, calmodulin-2, consists of 149 and shares an identical sequence with calmodulin isoforms from CALM1 and CALM3, exhibiting a molecular weight of approximately 16.7 kDa. Its tertiary structure forms a dumbbell-like shape with two compact globular lobes—an N-terminal (residues 1-77) and a C-terminal (residues 82-148)—linked by a flexible seven-turn central α-helix (residues 78-81). Each lobe contains a pair of EF-hand motifs, which are conserved helix-loop-helix calcium-binding sites comprising approximately 12 residues in the loop that coordinate Ca²⁺ ions through side-chain carboxylates from aspartic and glutamic acids, as well as main-chain carbonyl oxygens. In the apo (calcium-free) state, maintains an extended, open conformation with the lobes oriented away from each other, as observed in NMR and crystal structures such as PDB entry 1CFC. Binding of four Ca²⁺ ions—one per EF-hand—triggers a conformational shift: the central bends, allowing the two lobes to collapse toward each other and exposing hydrophobic surfaces for interaction with target proteins. The four EF-hands are non-identical, with the C-terminal sites exhibiting higher Ca²⁺ affinity (dissociation constants around 10⁻⁶ M) compared to the N-terminal sites (around 10⁻⁵ M), enabling sequential binding that fine-tunes activation. Secondary structure analysis reveals predominantly α-helical content (about 70%), with eight α-helices (labeled I-VIII) flanking the four EF-hand loops, and minimal β-sheet elements. structures, such as the 1.7 Å resolution refinement of calcium-bound (PDB 1CLL), confirm this helical dominance and highlight the plasticity of the central linker, which lacks secondary in the apo form but adopts helical character upon target engagement. This structural versatility underpins 's role as a versatile calcium , with no reported isoforms unique to CALM2 altering the core architecture.

Calcium-Dependent Mechanisms


, encoded by the CALM2 gene, binds four calcium ions via two pairs of EF-hand motifs in its N- and C-terminal lobes, enabling it to transduce calcium signals into cellular responses. Each EF-hand features a 12-residue loop that coordinates Ca²⁺ through seven oxygen atoms, primarily from carboxylates of aspartic and residues and main-chain carbonyls. The N-lobe exhibits higher Ca²⁺ affinity (K_d ≈ 10⁻⁶ M) and faster association/dissociation kinetics compared to the C-lobe (K_d ≈ 10⁻⁵ M), allowing sequential lobe during transient Ca²⁺ elevations.
In the apo form, calmodulin maintains a closed conformation with intra-lobe helix packing that buries hydrophobic surfaces. Calcium binding induces lobe-specific opening: the C-lobe unfolds first to expose a methionine-rich patch (Met¹⁴⁵, Met¹⁴⁹, Met¹⁵¹), followed by N-lobe exposure (Met³⁶, Met⁵¹, Met⁷¹), resulting in a dumbbell-shaped, extended structure with a flexible central linker. This transition, driven by Ca²⁺ coordination that neutralizes negative charges and stabilizes helical rearrangements, increases target-binding affinity by over 10⁶-fold. The Ca²⁺-calmodulin complex allosterically regulates targets by wrapping around amphipathic helices in enzymes like Ca²⁺/calmodulin-dependent protein kinase II (CaMKII), displacing autoinhibitory segments to activate catalytic domains. For instance, in CaMKII, Ca²⁺/calmodulin binding to the regulatory domain triggers T-site to R-site , exposing the and promoting autophosphorylation at Thr²⁸⁶ for sustained activity. Similar mechanisms govern activation of for contraction and adenylyl cyclase modulation, underscoring 's role in diverse Ca²⁺-dependent pathways without intrinsic enzymatic activity.

Physiological Roles

Calcium Signaling Pathways

Calmodulin, the protein product of the CALM2 gene, functions as a ubiquitous calcium sensor that decodes fluctuations in intracellular Ca²⁺ concentrations to regulate diverse signaling cascades essential for cellular , contraction, secretion, and . Encoded by three paralogous genes including CALM2, calmodulin comprises 148 with four EF-hand motifs that bind Ca²⁺ ions with differing affinities, the C-terminal sites exhibiting higher affinity while N-terminal sites enable rapid response. Upon Ca²⁺ binding, calmodulin transitions from a compact, inactive state to an extended conformation, exposing amphipathic helices that interact with target proteins, thereby amplifying and specifying Ca²⁺ signals. A primary pathway involves the activation of Ca²⁺/calmodulin-dependent protein kinases (CaMKs), where Ca²⁺-calmodulin binds and relieves autoinhibition, enabling phosphorylation of downstream substrates; for instance, CaMKII autophosphorylates at Thr286 to sustain activity post-Ca²⁺ transient, influencing , , and vascular tone. , upstream in this kinase cascade, phosphorylates CaMKI and CaMKIV, linking Ca²⁺ signals to AMPK activation for energy homeostasis and CREB-mediated transcription. In parallel, Ca²⁺-calmodulin activates (PP2B), a serine/ phosphatase that dephosphorylates nuclear factor of activated T-cells (NFAT), promoting its nuclear import and in immune cells. Calmodulin further modulates cyclic nucleotide signaling by stimulating phosphodiesterase 1 (PDE1), which hydrolyzes and cGMP, thereby integrating Ca²⁺ with second messenger pathways to control processes like relaxation and neuronal excitability. It regulates ion channels and transporters, including inhibition of (RyR2) to prevent aberrant Ca²⁺ release and activation of plasma membrane Ca²⁺-ATPase (PMCA) for cytosolic Ca²⁺ extrusion, maintaining signal fidelity. In enzymatic pathways, Ca²⁺- enhances (nNOS) activity, generating NO for and . These interactions, modulated by post-translational modifications like at Thr79 or oxidation at Met144, ensure context-specific responses, with CALM2 mutations disrupting affinity and leading to dysregulated signaling in conditions like arrhythmias.

Tissue-Specific Contributions

CALM2 demonstrates ubiquitous expression across human tissues, with particularly elevated mRNA levels in the (median RPKM of 152.2) and testis (median RPKM of 98.3), as determined from GTEx . This pattern reflects the broader role of isoforms in , though CALM2's specific contributions vary by tissue due to differential gene regulation among the three calmodulin-encoding genes (CALM1, CALM2, CALM3). In cerebral tissue, CALM2 supports neuronal calcium-dependent processes, including long-term synaptic potentiation, which underlies learning and formation through modulation of synaptic strength. Its high expression in brain regions such as the middle temporal gyrus facilitates interactions with targets like ion channels and kinases in presynaptic and postsynaptic compartments. Within , CALM2 provides a substantial portion of total , with CALM1 and CALM2 collectively contributing four-fold more to calmodulin levels than CALM3, based on GTEx sequencing across heart samples. This supports excitation-contraction coupling by regulating calcium release from the and activation of contractile proteins, essential for rhythmic heart function. Murine models confirm Calm2 as the predominant isoform in ventricular and atrial cardiomyocytes, underscoring its primacy in myocardial calcium homeostasis.

Pathophysiological Associations

Cardiac Disorders

Mutations in the CALM2 gene, which encodes calmodulin 2, have been linked to rare but severe inherited cardiac arrhythmias, collectively termed calmodulinopathies. These mutations disrupt calmodulin's role in regulating cardiac ion channels, particularly by impairing calcium-dependent inhibition of the (CaV1.2) and the rapid delayed rectifier potassium channel (/KCNH2), leading to prolonged duration and increased risk of ventricular arrhythmias. Affected individuals often present with life-threatening events such as sudden in infancy or early childhood, distinguishing these from more common forms of channelopathies. The primary cardiac phenotypes associated with CALM2 variants include (LQTS), classified as LQT15, and (CPVT). In LQTS cases, CALM2 mutations such as p.Asn98Ser (N98S) and variants like p.Gly13Asp have been identified in patients with severe QT prolongation (QTc often exceeding 500 ms) and recurrent syncope or , frequently triggered at rest or during sleep rather than exertion. CPVT linked to CALM2 manifests as bidirectional or polymorphic during adrenergic , with cases reporting exercise-induced arrhythmias in young patients. Approximately 30% of mutation carriers, including those with CALM2 variants, exhibit concomitant structural abnormalities such as left ventricular dysfunction, , or congenital heart defects, potentially exacerbating arrhythmic risk. Pathogenic CALM2 variants are typically missense mutations altering key residues in calmodulin's EF-hand motifs, reducing calcium affinity or altering conformational changes necessary for target binding. Functional studies demonstrate that these mutations fail to suppress calcium influx through CaV1.2, promoting early afterdepolarizations and . mutations predominate, with inheritance patterns showing incomplete but high lethality; for instance, over 70% of reported cases involve cardiac events before age 10. for CALM1, CALM2, and CALM3 is recommended in infants with unexplained prolongation or , given the overlap in encoded protein function. Therapeutic management relies on beta-blockers, implantable cardioverter-defibrillators, and left cardiac sympathetic , though outcomes remain guarded due to refractoriness.

Oncogenic and Other Diseases

CALM2 overexpression is implicated in the progression of multiple malignancies. In , elevated CALM2 expression correlates with reduced overall survival and disease-free survival, with immunohistochemical analyses showing higher levels in tumor tissues compared to adjacent normal tissue. Similarly, in (HCC), CALM2 is upregulated in tumor samples, and its knockdown via siRNA reduces , migration, and invasion while suppressing tumor growth in xenograft models; pharmacological inhibition of activity likewise impairs HCC progression, suggesting therapeutic potential. In gastric cancer, CALM2 facilitates and by activating the JAK2//HIF-1α/VEGFA pathway and promoting M2 polarization, as demonstrated in orthotopic models where CALM2 knockdown diminished metastases and vascular density. Common genetic variants in CALM2 have been linked to increased risk, particularly in women of ancestry, based on genome-wide association studies identifying cooperative transcriptional effects with other loci. Downregulation of CALM2 also sensitizes HER2-positive gastric cancer cells to the afatinib, indicating a role in mechanisms involving signaling. These findings position CALM2 as a potential and target, though requires further validation beyond correlative expression data. Beyond , mutations in CALM2 predominantly cause life-threatening cardiac arrhythmias. Missense variants, such as p.Asn98Ser (N98S), disrupt calmodulin's calcium-binding affinity, leading to prolonged QT intervals and (CPVT), with affected individuals experiencing syncope or sudden as early as infancy; functional studies in cardiomyocytes show increased calcium spark frequency and altered L-type . Other mutations, including changes like p.Phe89Leu, impair interactions with ion channels such as Cav1.2 and RyR2, exacerbating ventricular arrhythmias under stress; clinical registries report near-uniform lethality without interventions like implantable defibrillators. Calmodulinopathies from CALM2 variants exhibit incomplete penetrance and variable expressivity, with some carriers developing neurodevelopmental comorbidities including autism spectrum disorder (8 cases), (8 cases), and ADHD (5 cases) alongside cardiac symptoms, potentially due to disrupted neuronal . No direct associations with neurodegenerative diseases like Alzheimer's have been established for CALM2 mutations, despite calmodulin's broader role in calcium-dependent kinases. Therapeutic strategies focus on beta-blockers and left cardiac sympathetic denervation, as mutation-specific correctors remain experimental.

Molecular Interactions

Known Protein Partners

Calmodulin-2 (CALM2), encoding the identical calmodulin protein as CALM1 and CALM3, functions as a calcium sensor that interacts with over 300 target proteins to transduce calcium signals, primarily in a calcium-dependent manner through electrostatic and hydrophobic interactions. These partners span enzymes, ion channels, receptors, and cytoskeletal elements, with binding often mediated by conserved motifs such as the 1-10, 1-5-10, or 1-14 patterns in amphipathic alpha-helices for calcium-dependent interactions, and IQ motifs for calcium-independent ones. Enzymatic partners include (MLCK), which CALM2 activates to phosphorylate myosin for contraction; (PP2B), a serine/ regulated for T-cell activation and neuronal signaling; and calcium/calmodulin-dependent protein kinases (CaMKs), notably CaMKII, essential for in synapses via autophosphorylation and substrate targeting. Additional enzyme targets encompass adenylate cyclase and phosphodiesterases (PDEs), modulating levels, as well as (NOS) isoforms for . Ion channel and receptor partners feature ryanodine receptors (RyR1/RyR2) for calcium release in excitation-contraction coupling; receptors (IP3R) for calcium mobilization; voltage-gated sodium channels (Nav1.2/Nav1.5) for modulation; and small-conductance calcium-activated channels (SK) for after-hyperpolarization. Connexins (e.g., Cx43) represent proteins gated by CALM2 to control intercellular communication, while metabotropic glutamate receptors (mGluR5/mGluR7) link to G-protein signaling in . Cytoskeletal and other structural partners include myosins and actinin, facilitating , alongside adaptor proteins like those in invadopodia for cell invasion dynamics. Experimental evidence derives from structural studies (e.g., of complexes), affinity assays, and , confirming high-confidence interactions via databases like the Calmodulin Target Database.

Functional Networks

Calmodulin-2, the protein product of CALM2, functions as a central hub in calcium-dependent signaling networks, binding Ca²⁺ ions to regulate downstream effectors including enzymes, ion channels, and pumps. Upon Ca²⁺ binding, it undergoes conformational changes that enable interactions with targets such as calcium/calmodulin-dependent protein kinases (CaMKs), , and , thereby transducing signals for processes like , neurotransmitter release, and . In protein-protein interaction (PPI) networks, CALM2 exhibits high connectivity, with verified partners including CAMK1, CAMK2B, RYR2 (ryanodine receptor 2), CACNA1C ( subunit alpha-1C), KCNN2 (potassium calcium-activated channel subfamily N member 2), KCNQ1 (potassium voltage-gated channel subfamily Q member 1), and DAPK2 (death-associated protein kinase 2), as mapped in databases like and BioGRID. These interactions cluster around ion homeostasis, , and , with network analyses showing CALM2's role in modules regulating Ca²⁺ influx/efflux and kinase activation. CALM2 integrates into broader pathway networks, notably the KEGG calcium signaling pathway, where it modulates targets like CaMKII for long-term potentiation and circadian entrainment, and Reactome pathways such as activation of CaMK IV and kainate receptor signaling upon glutamate binding. In cardiac contexts, it links to oxytocin signaling and arrhythmia susceptibility via RyR2 stabilization, while in neuronal networks, it supports glutamatergic synapse regulation through CaMKII-ERK cascades. Tissue-specific expression amplifies its network roles, with higher CALM2 levels in brain and heart correlating to enriched interactions in neurodevelopmental and arrhythmogenic modules. Structural studies of CALM2 complexes, such as with CaMK fragments, underscore its network versatility, revealing flexible domain rearrangements that accommodate diverse partners for signal specificity. Experimental data from affinity capture and co-immunoprecipitation confirm these hubs, though isoform redundancy with CALM1/CALM3 necessitates context-specific validation.

Recent Research Advances

In 2023, structural and functional studies revealed that mutations at glycine 114, including those in CALM2, disrupt binding to the IQ domain of the voltage-gated NaV1.5, leading to altered channel inactivation and increased late sodium current, which contributes to arrhythmogenic phenotypes in calmodulinopathy. This finding highlights a specific molecular interaction defect amenable to targeted interventions. Research in 2024 demonstrated proof-of-principle for single-construct suppression-replacement in calmodulinopathy models harboring CALM2 variants, where mutant CALM2 suppression combined with wild-type CALM1 delivery restored normal interactions with ion channels, shortening prolonged duration by modulating calcium-dependent signaling. Similarly, antisense therapy targeting CALM2 and related genes reduced mutant protein expression, normalizing interactions in patient-derived cardiomyocytes and mitigating risk without off-target effects on wild-type calmodulin. A elucidated isoform-specific spatiotemporal mRNA localization of CALM2 in cardiac myocytes, showing distinct intracellular distribution that enables non-redundant calmodulin-dependent signaling interactions, such as localized calcium-calmodulin activation, thereby influencing excitation-contraction coupling efficiency. These advances underscore gene-specific regulatory mechanisms modulating calmodulin's broad protein partnership network.

Therapeutic Prospects

Therapeutic strategies targeting CALM2 primarily focus on addressing gain-of-function mutations associated with severe cardiac arrhythmias, such as long-QT syndrome (LQTS) and (CPVT), where CALM2 variants disrupt calcium-dependent regulation of ion channels. A proof-of-principle demonstrated single-construct suppression-replacement using interference (CRISPRi) to silence both mutant and wild-type CALM2 alleles while introducing a codon-optimized wild-type CALM2 transgene, effectively rescuing arrhythmogenic phenotypes in patient-derived cardiomyocytes without off-target effects on CALM1 or CALM3 isoforms. This approach highlights isoform-specific gene editing as a precision medicine tool for calmodulinopathies, with modular guide RNAs adaptable to individual CALM2 mutations. In , elevated CALM2 expression correlates with aggressive tumor behavior and poor prognosis, positioning it as a candidate for inhibitory interventions. In hepatocellular carcinoma (HCC), RNA interference-mediated knockdown of CALM2 suppressed , migration, and invasion and reduced tumor growth in xenograft models, suggesting potential as a strategy to prevent and recurrence alongside standard therapies. Similarly, CALM2 silencing in HER2-positive gastric cancer cells enhanced sensitivity to the afatinib by modulating signaling and pathways, indicating combinatorial potential to overcome . High CALM2 levels in predict reduced overall and disease-free survival, further supporting downregulation as a therapeutic avenue, though clinical translation requires validation beyond preclinical models. Challenges in CALM2 targeting include its structural similarity to other calmodulin isoforms, risking non-specific effects on essential , and the absence of approved small-molecule inhibitors selective for CALM2. While general antagonists like trifluoperazine bind CALM2 in structural studies, their broad activity limits therapeutic specificity. Ongoing research emphasizes nucleic acid-based approaches, such as isoform-selective siRNAs or antisense , to exploit CALM2's role in downstream effectors like CaMKII without disrupting basal cellular functions. No CALM2-specific drugs have entered clinical trials as of 2025, underscoring the need for improved delivery systems and biomarkers to monitor efficacy in mutation-driven or overexpression contexts.

References

  1. [1]
    Entry - *114182 - CALMODULIN 2; CALM2 - OMIM - (OMIM.ORG)
    Calmodulin is an essential calcium-sensing, signal-transducing protein. Three calmodulin genes, CALM1 (114180), CALM2, and CALM3 (114183), ...
  2. [2]
    805 - Gene ResultCALM2 calmodulin 2 [ (human)] - NCBI
    Calmodulin is a calcium binding protein that plays a role in signaling pathways, cell cycle progression and proliferation.
  3. [3]
    CALM2 - Calmodulin-2 - Homo sapiens (Human) | UniProtKB | UniProt
    May 10, 2017 · Calmodulin acts as part of a calcium signal transduction pathway by mediating the control of a large number of enzymes, ion channels, aquaporins and other ...
  4. [4]
    Novel Calmodulin (CALM2) Mutations Associated with Congenital ...
    In the recent reports of human calmodulin gene mutations, there was only one CALM2 allele identified compared to five CALM1 mutations. The previously identified ...
  5. [5]
    Spectrum and Prevalence of CALM1-, CALM2-, and CALM3 ...
    Calmodulin (CaM) is an essential Ca2+ sensing, signal-transducing protein. Calcium-induced activation of CaM regulates many calcium-dependent processes and ...
  6. [6]
    Calmodulinopathy: Functional Effects of CALM Mutations and Their ...
    Dec 11, 2018 · This review discusses how mutations affect CaM signaling function and how this may relate to the distinct arrhythmia phenotypes/mechanisms observed in patients.
  7. [7]
    Characterization of the human CALM2 calmodulin gene ... - PubMed
    1 Department of Biochemistry and Molecular Biology, Mayo Graduate School, Mayo Foundation, Rochester, MN 55905, USA.
  8. [8]
    CALM1, CALM2, and CALM3 expression and translation efficiency ...
    May 15, 2025 · Our findings reveal a four-fold greater contribution of CALM1 and CALM2 to the total calmodulin levels in the human heart compared to CALM3.
  9. [9]
    Characterization of the human CALM2 calmodulin gene and ...
    Human calmodulin is encoded by three genes CALM1, CALM2 and CALMS located on different chromosomes. To complete the characterization of this family, ...<|control11|><|separator|>
  10. [10]
    Differential expression of the three independent CaM genes coding ...
    The CALM2 gene is the longest of the three CaM genes [51], and all human ... genes) have 6 exons and 5 introns (see Table 1). Of interest and ...
  11. [11]
    Calmodulin 2 expression is associated with poor prognosis in breast ...
    CALM2 is overexpressed in BRCA and its upregulation is significantly correlated with poor patient prognosis. Elevated CALM2 expression holds promise as a ...
  12. [12]
    Structural Consequences of Calmodulin EF Hand Mutations
    It consists of two globular domains, each containing two EF hand pairs capable of binding Ca2+, joined by a flexible central linker region.Experimental Procedures · Results · Discussion · Abbreviations
  13. [13]
    Conserved properties of individual Ca 2+ -binding sites in calmodulin
    CaM binds Ca2+ at four, nonidentical sites that contain the structural motif called an EF-hand (10, 11), each of which contains an acidic Ca2+-coordinating loop ...
  14. [14]
    Identification of the critical structural determinants of the EF-hand ...
    EF-hand calcium binding proteins (CaBPs) share strong sequence homology, but exhibit great diversity in structure and function. Thus although calmodulin (CaM) ...
  15. [15]
    Full article: Calmodulin Mutations in Human Disease
    Here, we review the research that has been done to identify calmodulinopathic CaM mutations and evaluate the mechanisms underlying their role in disease.Cam Regulation In Diverse... · Cam Regulation Of Ion... · Lqts-Associated Cam...
  16. [16]
    Structural basis for calmodulin as a dynamic calcium sensor - PMC
    Upon binding Ca2+, calmodulin changes its conformation to form complexes with a diverse array of target proteins. Despite a wealth of knowledge on calmodulin, ...
  17. [17]
    Molecular Dynamics Study of the Changes in Conformation ... - Nature
    Jul 23, 2019 · The conformational changes induced by calcium and/or target binding are crucial to the functions of calmodulin. Calmodulin binds four Ca2+ ions ...
  18. [18]
    Mechanisms of Ca2+/calmodulin-dependent kinase II activation in ...
    Jun 25, 2019 · CaMKII is composed of 12 subunits, each of which is a kinase that is activated by the binding of Ca2+/calmodulin (CaM)1.
  19. [19]
    Calmodulin in Action: Diversity in Target Recognition and Activation ...
    Highly regulated changes in the concentration of cytosolic calcium ions control biological processes as diverse as muscle contraction, fertilization, ...
  20. [20]
    Calmodulin: The switch button of calcium signaling - PMC
    Calmodulin (CaM), a well-defined calcium sensor, is central to the activation of calcium-dependent signaling pathways and certain types of mutations in CaM's ...
  21. [21]
    Calcium Signalling in Heart and Vessels: Role of Calmodulin and ...
    Dec 17, 2022 · In this review, we present the current literature regarding the role of calcium/calmodulin pathways in the heart and vessels with the aim to ...
  22. [22]
    CaMKK2: bridging the gap between Ca2+ signaling and energy ...
    CaMKK2 converts Ca2+-signals generated by these receptors into cellular responses by activating four downstream signaling pathways, which are regulated by the ...
  23. [23]
    Calcineurin | Cell Communication and Signaling | Full Text
    Aug 28, 2020 · Calcineurin is activated by increases in intracellular calcium concentrations, a process that requires the calcium sensing protein calmodulin ...
  24. [24]
    CALM2 ENSG00000143933 expression in Homo sapiens (human)
    General information. Expressed in middle temporal gyrus and 215 other cell types or tissues. Gene identifier. ENSG00000143933. Name.
  25. [25]
    [PDF] Enrichment of mutant calmodulin protein in a murine model of a ...
    Overall, our numerical estimates suggest that Calm2 is the most highly expressed of all 3 Calm genes within wild-type and mutant hearts, while chamber-specific.
  26. [26]
    Spectrum and Prevalence of CALM1-, CALM2-, and ... - PubMed
    Background: Calmodulin (CaM) is encoded by 3 genes, CALM1, CALM2, and CALM3, all of which harbor pathogenic variants linked to long QT syndrome (LQTS) with ...
  27. [27]
    Calmodulin 2 Mutation N98S Is Associated with Unexplained ...
    Calmodulin 1, 2 and 3 (CALM) mutations have been found to cause cardiac arrest in children at a very early age. The underlying aetiology described is long QT ...
  28. [28]
    Novel Association of a De Novo CALM2 Mutation With Long QT ...
    Oct 15, 2018 · With a calcium transporter gene, such as CALM2, disruptions in calcium signaling may contribute to cardiomyopathy either independently or in ...
  29. [29]
    UNCOMMON CALM2 MUTATION IN CATECHOLAMINERGIC ...
    Apr 6, 2024 · We present a case of a young patient with CPVT found to have an unusual, potentially causative, CALM2 mutation.
  30. [30]
    [PDF] Clinical presentation of calmodulin mutations
    CALM-LQTS and CALM-CPVT were the most prevalent phenotypes. • Cardiomyopathies and congenital heart defects were present in 30% of patients. • Of 111 evaluable ...<|separator|>
  31. [31]
    Calmodulin Mutations Associated with Long QT Syndrome Prevent ...
    Recent work has identified missense mutations in calmodulin (CaM) that are associated with severe early-onset long-QT syndrome (LQTS), leading to the ...
  32. [32]
    Clinical presentation of calmodulin mutations: the International ...
    Aug 2, 2023 · We identified 12 patients, mostly LQTS (n = 9), with coexistence of both primary neurological features and cardiac structural abnormalities.Abstract · Introduction · Results · Discussion
  33. [33]
    Early-onset cardiac arrest, prolonged QT interval, and left ventricular ...
    CALM1 and CALM2 variants were initially described in infants with recurrent cardiac arrest, all of whom exhibited a severe cardiac phenotype of recurrent ...
  34. [34]
    Targeting CALM2 Inhibits Hepatocellular Carcinoma Growth and ...
    CALM is overexpressed in different types of tumor tissues, and its altered expression is significantly associated with malignant tumor pathogenesis and ...
  35. [35]
    Calmodulin 2 Facilitates Angiogenesis and Metastasis of Gastric ...
    CALM2 modulates the JAK2/STAT3/HIF-1/VEGFA axis and bolsters macrophage polarization, thus facilitating GC metastasis and angiogenesis.
  36. [36]
    CALM2 Gene - Ma'ayan Laboratory, Computational Systems Biology
    Beyond its roles in the heart and breast, dysregulation of CALM2 expression appears to be involved in fibrogenic processes and neuronal stress responses. Lower ...
  37. [37]
    Knockdown of CALM2 increases the sensitivity to afatinib in HER2 ...
    Our study reveals for the first time that downregulation of CALM2 can overcome the resistance of GC cells to afatinib.
  38. [38]
    Effect of Ca2+ on the promiscuous target-protein binding of calmodulin
    Calmodulin is a protein involved in the regulation of a variety of cell signaling pathways. It acts by making usually calcium-insensitive proteins sensitive to ...<|separator|>
  39. [39]
    Structural Aspects and Prediction of Calmodulin-Binding Proteins
    Calmodulin (CaM) is an important intracellular protein that binds Ca2+ and functions as a critical second messenger involved in numerous biological ...
  40. [40]
    Calmodulin: a highly conserved and ubiquitous Ca2+ sensor - PMC
    Jul 31, 2024 · Among these, calmodulin-dependent enzymes are major targets. Adenylate cycle and PDE, act in opposite directions on calmodulin ...
  41. [41]
    Gene - CALM2 calmodulin 2 [Homo sapiens (human)] - NCBI - NIH
    Sep 13, 2025 · microRNA-651-5p affects the proliferation, migration, and invasion of lung cancer cells by regulating Calmodulin 2 expression.
  42. [42]
    Calmodulin 2 expression is associated with poor prognosis in breast ...
    CALM2 expression was significantly elevated in BRCA, with increased levels predicting poor overall survival (OS) and disease-free survival (DFS).
  43. [43]
    Reactome Pathway Database: Home
    Reactome is pathway database which provides intuitive bioinformatics tools for the visualisation, interpretation and analysis of pathway knowledge.Manually · Pathway Browser · Download · Analysis ToolsMissing: CALM2 | Show results with:CALM2
  44. [44]
    Role of Ca 2+ /Calmodulin-Dependent Protein Kinase Type II in ...
    The article will review the multiple roles played by CaMKII in glutamatergic synapses and how they are affected in disease conditions.
  45. [45]
    ANK2 - CALM2 Interaction Summary - BioGRID
    An interaction is inferred when a bait protein is affinity captured from cell extracts by either polyclonal antibody or epitope tag and the associated ...
  46. [46]
    Calmodulin mutations affecting Gly114 impair binding to the NaV1.5 ...
    Missense variants in CALM genes encoding the Ca 2+ -binding protein calmodulin (CaM) cause severe cardiac arrhythmias.
  47. [47]
    Single Construct Suppression and Replacement Gene Therapy for ...
    Jul 29, 2024 · The CALM-SupRep gene therapy shortened the pathologically prolonged APD90 in CALM1-, CALM2-, and CALM3-variant CaM-mediated long-QT syndrome ...
  48. [48]
    Antisense Oligonucleotide Therapy for Calmodulinopathy - PubMed
    Oct 8, 2024 · Background: Calmodulinopathies are rare inherited arrhythmia syndromes caused by dominant heterozygous variants in CALM1, CALM2, or CALM3, which ...<|control11|><|separator|>
  49. [49]
    Distinct intracellular spatiotemporal expression of Calmodulin genes ...
    Jul 8, 2025 · Our findings reveal that Calm1, Calm2, and Calm3 fulfil distinct, non-redundant roles in cardiac myocytes through their spatially regulated mRNA localization.
  50. [50]
    Single Construct Suppression and Replacement Gene Therapy for ...
    Jul 29, 2024 · We provide the first proof-of-principle suppression-replacement gene therapy for CaM-mediated long-QT syndrome.
  51. [51]
    A Precision Medicine Approach to the Rescue of Function on ... - NIH
    Having established a viable model system, we next use CRISPRi to selectively silence the expression of the CALM2 gene (both mutant and wild-type CALM2 alleles) ...
  52. [52]
    Targeting CALM2 Inhibits Hepatocellular Carcinoma Growth and ...
    Targeting CALM2 may be a molecular strategy for both primary HCC treatment and prevention of metastasis or recurrence.
  53. [53]
    Targeting of a Novel Ca+2/Calmodulin-Dependent Protein Kinase II ...
    As a positive control, cells were treated with calyculin-A, which is a type-1 and type-2A protein phosphatase inhibitor that has been shown to depolymerize at ...
  54. [54]
    Challenges and Opportunities for Therapeutic Targeting of ...
    Calmodulin kinase II inhibition protects against structural heart disease. Nat. Med. 11, 409–417. doi: 10.1038/nm1215. PubMed Abstract | CrossRef Full Text ...