NCS1 acts as a Ca²⁺ sensor, modulating diverse cellular processes through protein interactions:
NCS1 binds to target proteins via its hydrophobic crevice, with Ca²⁺ occupancy altering binding affinity . In C. elegans and mammals, it enhances neuroprotection and axonal regeneration .
NCS1 is implicated in neurological and non-neurological disorders:
In cancer, high NCS1 expression correlates with poor prognosis in skin (SKCM), liver (LIHC), and kidney (KIRC) cancers .
Mice models: NCS1-KO males show impaired displaced object recognition and altered thermal/motor recovery post-injury, while females remain unaffected .
Mitochondrial regulation: NCS1 deficiency reduces ATP synthesis (↓ ND1, ENO2) and increases metabolic stress (↓ UCP4/5, DJ-1) in dopaminergic neurons .
Synaptic modulation: NCS1/Ric-8A complex stabilizes synapses; disruption improves fragile X syndrome phenotypes .
Prognostic value: High NCS1 expression predicts reduced survival in SKCM, LIHC, BRCA, COAD, and KIRC .
Pathway enrichment: Co-regulated with immune-related and COVID-19 response genes, suggesting immunomodulatory roles .
NCS1 is a highly conserved calcium binding protein abundantly expressed in neurons. It functions primarily as a modulator of intracellular calcium homeostasis, calcium-dependent signaling pathways, as well as neuronal transmission and plasticity . Unlike other neuronal calcium sensors, NCS1 is found outside the nervous system and does not contain a Ca²⁺/Myr switch, causing it to be constantly bound to the membrane .
The protein structure of NCS1 consists of two pairs of EF-hand motifs, with three functional calcium-binding sites: EF-2, EF-3, and EF-4. EF-2 and EF-3 can also recognize Mg²⁺ and serve as structural sites that allow the protein to adopt its tertiary fold. Meanwhile, EF-4 functions as a regulatory Ca²⁺ binding site capable of sensing changes in cytosolic calcium levels in neurons .
Methodologically, researchers can study NCS1 function through calcium imaging techniques, electrophysiology, and protein-protein interaction assays to understand how it modulates neuronal activity and signaling pathways.
Several experimental models exist for studying NCS1 function in humans, with CRISPR-Cas9 genome editing being particularly valuable. Researchers have successfully generated NCS1 knockout human induced pluripotent stem cell (hiPSC) lines that show regular expression of pluripotent markers, normal iPSC morphology and karyotype, with no detectable off-target effects on potentially affected genes .
These knockout cell lines serve as valuable tools for studying the role of NCS1 in the pathophysiology of various neuropsychiatric disorders and non-neurological diseases . The methodology for creating such models typically involves:
Design of guide RNAs targeting specific regions of the NCS1 gene
Transfection of hiPSCs with CRISPR-Cas9 components
Clonal selection and expansion
Validation through sequencing and expression analysis
Functional characterization through calcium imaging and electrophysiology
Additionally, researchers can differentiate these hiPSCs into neurons to study the effects of NCS1 knockout on neuronal development, synapse formation, and electrophysiological properties.
NCS1 participates in a wide range of protein interactions due to its ability to recognize and regulate different target proteins. The multifunctionality of NCS1 relies on its ability to interact with G-protein-coupled receptors (GPCRs) and some of their regulators, Ca²⁺ channels, guanine nucleotide exchange factors (GEF), and kinases, both in a Ca²⁺-dependent and -independent manner .
One well-characterized interaction is between NCS1 and Ric-8A, where they coregulate synapse number and probability of neurotransmitter release . NCS1 uses a surface-exposed hydrophobic crevice to recognize its targets, which generally present short helical motifs that bind to the N- or C-terminal part of this large cavity. The shape and size of this hydrophobic crevice, along with hydrophilic residues at its border, determine target specificity .
Methodological approaches to study these interactions include:
Co-immunoprecipitation assays
Crystal structure analysis
Fluorescence resonance energy transfer (FRET)
Surface plasmon resonance (SPR)
Yeast two-hybrid screening
NCS1 participates in a wide range of important neuronal functions that collectively impact neural circuit development and function. It serves as a regulator of Ca²⁺ channels, which are critical for controlling neuronal excitability and neurotransmitter release . Additionally, NCS1 regulates exocytosis, the process by which neurotransmitters are released into the synaptic cleft.
Beyond its role in neurotransmission, NCS1 is involved in synaptogenesis (the formation of synapses between neurons) and axonal growth . These developmental functions ultimately affect higher cognitive processes such as learning and memory. Research has also revealed roles for NCS1 in neuroprotection and axonal regeneration, suggesting it contributes to neuronal resilience and recovery after injury .
Methodologically, these functions can be studied through:
Electrophysiological recordings to measure synaptic transmission
Live imaging to track synapse formation and axonal growth
Calcium imaging to visualize calcium dynamics in neurons
Behavioral assays in animal models with altered NCS1 expression
NCS1 has been implicated in several pathological processes related to neuropsychiatric disorders. Research has linked NCS1 to X-linked mental retardation and autism, schizophrenia, and bipolar disorder . These associations suggest that dysregulation of calcium signaling through NCS1 may contribute to the pathophysiology of these conditions.
The generation of NCS1 knockout human induced pluripotent stem cell lines provides a valuable tool for studying these relationships . These cell lines can be differentiated into neurons to investigate how the absence of NCS1 affects neuronal development, connectivity, and function in the context of these disorders.
Methodological approaches to studying NCS1's role in neuropsychiatric disorders include:
Gene association studies in patient populations
Expression analysis in post-mortem brain tissue
Functional studies using knockout or knockdown models
Electrophysiological and calcium imaging analyses in patient-derived neurons
Behavioral assessments in animal models with altered NCS1 expression
The structural determinants of NCS1 target specificity are based on the shape and size of its hydrophobic crevice. NCS1 contains a dynamic C-terminal helix (helix H10) that can insert into the crevice, thus contributing to its shape . Since Ca²⁺ binding promotes structural rearrangements, the occupancy of the three Ca²⁺ binding sites also determines affinity for protein partners .
The structures of several NCS proteins bound to their corresponding targets have shown that these Ca²⁺ sensors use a surface-exposed hydrophobic crevice to recognize their targets, which generally present short helical motifs that bind to the N- or C-terminal part of this large cavity . Additionally, the presence of hydrophilic residues at the border of the crevice contributes to target specificity and constitutes hot spots for interactions with different targets .
Methodologically, researchers can study these mechanisms through:
X-ray crystallography of NCS1-target complexes
Site-directed mutagenesis of key residues
Isothermal titration calorimetry to measure binding affinities
Molecular dynamics simulations to observe conformational changes
Nuclear magnetic resonance spectroscopy to study dynamics in solution
The interaction between NCS1 and Ric-8A has been extensively studied, revealing important structural and functional insights. Crystal structure analysis identified that the region of Ric-8A necessary and sufficient for NCS1 recognition corresponds to the two-helix bundle that constitutes the HEAT repeat 9 of the ARM-HEAT repeat domain .
When binding to NCS1, the HEAT repeat 9 of Ric-8A undergoes a substantial conformational change: helix a9 unfolds while helix b9 refolds, resulting in regions named R1 and R2 . This interaction is functionally significant as NCS1 and G-proteins compete for Ric-8A binding, suggesting they could share certain interaction surfaces .
Through a combination of crystallographic work using Ric-8A peptides of different lengths and cell-based protein-protein interaction assays with various Ric-8A and NCS1 mutants, researchers have identified specific regions necessary for this interaction . Interestingly, a C-terminally truncated construct of human Ric-8A (hRic-8A-424) showed significantly higher affinity for NCS1 compared to full-length hRic-8A, suggesting that the HEAT repeat 9, but not the rH10 or rH11 helices, is implicated in the protein-protein interaction .
Methodological approaches to study this interaction include:
Co-immunoprecipitation assays with different protein constructs
Crystal structure analysis of protein complexes
Mutational analysis of binding interfaces
Functional assays measuring G-protein activation
Live-cell imaging of protein interactions
Recent research has highlighted NCS1's involvement in immune regulation and its potential use as a prognostic biomarker for multiple cancer types, including SKCM (skin cutaneous melanoma), LIHC (liver hepatocellular carcinoma), BRCA (breast cancer), COAD (colon adenocarcinoma), and KIRC (kidney renal clear cell carcinoma) .
Researchers have employed multiple methodological approaches to study NCS1's role in cancer:
Analysis of RNA sequencing expression data from TCGA (The Cancer Genome Atlas) and GTEx (Genotype-Tissue Expression) programs using tools like GEPIA 2.0
Assessment of NCS1's correlation with clinicopathological features using Kaplan-Meier Plotter and The Human Protein Atlas
Evaluation of NCS1 DNA promoter region methylation in pan-cancer data using the UALCAN database
Immune score assessment using the "immuneeconv" R package, which integrates six algorithms: TIMER, xCell, MCP-counter, CIBERSORT, EPIC, and stromal score
Through these approaches, researchers have analyzed the association between NCS1 and immunosuppressive, immunostimulatory, and MHC molecules, as well as tumor-infiltrating lymphocytes (TILs) and chemokines regulating T-cell trafficking in pan-cancer . This research direction provides valuable insights for developing new therapeutic approaches combining NCS1-targeted treatments with immunotherapy.
Cancer Type | NCS1 as Prognostic Biomarker | Associated Immune Features |
---|---|---|
SKCM | Yes | Correlation with immune checkpoint expression |
LIHC | Yes | Association with tumor microenvironment scores |
BRCA | Yes | Linked to specific immune subtypes |
COAD | Yes | Related to tumor mutation burden |
KIRC | Yes | Correlation with specific immune cell infiltration |
While not explicitly discussed in the search results, single-cell analysis techniques represent a cutting-edge methodological approach for studying NCS1 function in heterogeneous cell populations. These techniques can overcome limitations of bulk tissue analysis, which may mask cell type-specific expression patterns and functions of NCS1.
Methodological approaches include:
Single-cell RNA sequencing (scRNA-seq) to profile NCS1 expression across different cell types and correlate it with expression of other genes involved in calcium signaling, synaptic function, and disease pathways
Single-cell proteomics to analyze NCS1 protein levels and post-translational modifications
Spatial transcriptomics to map NCS1 expression within tissue architecture
Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) to simultaneously analyze NCS1 expression and cell surface protein markers
Patch-seq, which combines electrophysiological recording with single-cell transcriptomics to link NCS1 expression to functional properties
These approaches can reveal cell type-specific roles of NCS1 in both normal physiology and disease states, potentially identifying new therapeutic targets for conditions associated with NCS1 dysfunction.
Phosphorylation of proteins like NCS1 can significantly alter their binding properties and functions. While the search results don't specifically address NCS1 phosphorylation, they do mention phosphorylated Ric-8A (pRic-8A) and its interaction with Gα . This suggests that phosphorylation states are important in the signaling pathways involving NCS1 and its binding partners.
Effective methodological approaches for studying NCS1 phosphorylation include:
Mass spectrometry-based phosphoproteomics to identify phosphorylation sites
Phospho-specific antibodies for Western blotting and immunoprecipitation
Phosphomimetic and phospho-deficient mutants to study functional consequences
In vitro kinase assays to identify kinases responsible for NCS1 phosphorylation
Live-cell imaging using phospho-sensors to monitor dynamics of phosphorylation
Structural studies comparing phosphorylated and non-phosphorylated forms
Understanding the phosphorylation status of NCS1 in different cellular contexts could provide insights into its regulation and function in health and disease.
NCS-1 is a high-affinity, low-capacity calcium-binding protein that is ubiquitously expressed, with the highest abundance in neuronal tissues . It is N-terminally myristoylated, which allows it to bind intracellular membranes . The protein primarily transduces calcium signals through interactions with its target proteins, including dopamine receptor D2 (D2R), voltage-gated calcium channels (Cavs), and inositol 1,4,5-trisphosphate receptors (InsP3Rs) . These interactions modulate various downstream effects such as neurotransmission, synaptic plasticity, neurite outgrowth, and neuronal survival .
NCS-1 has been implicated in several neurological and psychiatric conditions, including autism spectrum disorder (ASD), fragile X syndrome, Parkinson’s disease, Alzheimer’s disease, bipolar disorder, and schizophrenia . Knockout studies in model organisms like Caenorhabditis elegans and mice have shown that the absence of NCS-1 results in impaired memory and learning . Conversely, overexpression of NCS-1 in the dentate gyrus of mice enhances synaptic plasticity and memory .
NCS-1 regulates synaptic transmission and helps control the dynamics of nerve terminal growth . It is critical for some forms of learning and memory in both C. elegans and mammals . The protein also regulates corticohippocampal plasticity, and enhancing levels of NCS-1 in the mouse dentate gyrus increases spontaneous exploration of safe environments . This potentially links NCS-1 to curiosity .
In addition to calcium ions, NCS-1 can bind mobile, or signaling, intracellular zinc, which is a characteristic feature of cells under oxidative stress . Zinc coordination under these conditions stimulates NCS-1 oxidation to form a disulfide dimer (dNCS-1) with altered functional properties . This combined effect of mobile zinc and increased redox potential can induce aberrant NCS-1 activity, promoting either the survival of neuronal cells or their apoptosis, potentially leading to neurodegenerative processes .