RCAN1 Human

Regulator of Calcineurin 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure and Isoforms

RCAN1 spans ~45 kb and contains seven exons. Alternative splicing generates four major isoforms:

IsoformExonsMolecular WeightKey Functions
RCAN1-11 + 5-748 kDaCalcineurin inhibition, stress response
RCAN1-44 + 5-724 kDaNFAT signaling regulation
RCAN1-2/-3Rare variantsN/ALimited data

Sources:

RCAN1-1 and RCAN1-4 are the most studied isoforms, with distinct tissue expression patterns. RCAN1-1 is upregulated under oxidative or calcium stress, while RCAN1-4 is linked to calcineurin-dependent pathways .

Calcineurin Inhibition

RCAN1 binds calcineurin A (a calcium-dependent phosphatase) and inhibits its dephosphorylation of NFAT (Nuclear Factor of Activated T-cells), thereby blocking NFAT nuclear translocation and transcriptional activity . This regulation impacts:

  • Immune responses: T-cell activation and survival .

  • Cardiac hypertrophy: RCAN1 overexpression suppresses pathological hypertrophy in mice .

  • Neurodegeneration: Chronic overexpression in Down syndrome brains contributes to Alzheimer-like pathology .

Calcineurin-Independent Roles

ProcessMechanismImpact
Oxidative stressIncreases reactive oxygen species (ROS)Disrupts mitochondrial function
Mitochondrial Ca²⁺ uptakeElevates mitochondrial Ca²⁺ levelsReduces ATP production
AngiogenesisInhibits VEGF expressionSuppresses tumor vascularization

In vitro studies using LS8 cells overexpressing RCAN1 showed:

  • ROS increase: GSH/GSSG ratio ↓ (oxidized state) .

  • Mitochondrial dysfunction: Basal respiration ↓, ATP production ↓ .

  • Enamel defects: Downregulated enamel genes (e.g., AMELX, ENAM) .

Down Syndrome

Trisomy 21 leads to RCAN1 overexpression, contributing to:

  • Neurodegeneration: Enhanced amyloid-β aggregation, neurofibrillary tangles .

  • Cognitive deficits: Impaired neurogenesis and synaptic plasticity .

  • Dental anomalies: Hypoplastic enamel due to disrupted Ca²⁺ signaling .

Alzheimer’s Disease

RCAN1 is upregulated in Alzheimer’s brains, exacerbating:

  • Calcineurin dysregulation: Promotes tau phosphorylation and neurofibrillary tangles .

  • Oxidative stress: Elevated ROS damages neuronal mitochondria .

Cardiac Hypertrophy

RCAN1 overexpression in transgenic mice:

  • Blocks pathological hypertrophy: Attenuates NFAT-driven gene expression .

  • Reduces infarct remodeling: Preserves left ventricular function post-myocardial infarction .

Cancer

RCAN1’s dual role in:

ProcessEffectOutcome
Angiogenesis↓ VEGF, ↓ vascular densityReduced tumor growth
Apoptosis↑ Glucocorticoid-induced cell deathLeukemia susceptibility

DS patients show reduced incidence of solid cancers but higher leukemia risk, highlighting RCAN1’s context-dependent effects .

Calcineurin-NFAT Axis

RCAN1 binds calcineurin’s catalytic domain, blocking NFAT dephosphorylation. Phosphorylated RCAN1 (e.g., at Ser residues) enhances calcineurin inhibition, while unphosphorylated forms may paradoxically activate it .

Neurotrophin Trafficking

In Dp(16)1Yey/+ mice (trisomic for chromosome 21), excess RCAN1:

  • Impairs TrkA endocytosis: Disrupts NGF signaling, causing sympathetic nervous system defects .

  • Reduces calcineurin activity: ↓ NFAT activation, ↓ neurotrophic support .

Therapeutic Potential

RCAN1 modulation is a promising strategy:

  • Inhibitors: Restoring calcineurin activity in Down syndrome brains.

  • Overexpression: Suppressing cardiac hypertrophy or tumor angiogenesis.

  • Phosphorylation-targeted therapies: Enhancing RCAN1’s calcineurin-inhibiting capacity .

Product Specs

Introduction
RCAN1, encoded by a gene located on human chromosome 21, interacts with calcineurin A and inhibits calcineurin-dependent signaling pathways, potentially influencing the development of the central nervous system. This gene resides within the minimal candidate region associated with the Down syndrome phenotype and exhibits overexpression in the brains of Down syndrome fetuses. Chronic overexpression of RCAN1 may contribute to the formation of neurofibrillary tangles, a characteristic observed in Alzheimer's disease. Three transcript variants of this gene have been identified, encoding three distinct isoforms.
Description
RCAN1 Isoform-b Human Recombinant, produced in E. coli, is a single, non-glycosylated polypeptide chain composed of 117 amino acids. It has a molecular weight of 13 kDa.
Physical Appearance
Sterile filtered, colorless solution.
Formulation
This protein solution (1mg/ml) is supplied in 20mM Tris-HCl (pH 7.5), 100mM NaCl, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), store at 4°C. For extended storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 90.0% as determined by SDS-PAGE analysis.
Synonyms
Calcipressin-1, Regulator of calcineurin 1, Down syndrome critical region protein 1, Myocyte-enriched calcineurin-interacting protein 1, MCIP1, Adapt78, RCAN1, ADAPT78, CSP1, DSC1, DSCR1, RCN1.
Source
Escherichia Coli.
Amino Acid Sequence
MKLYFAQTLH IGSSHLAPPN PDKQFLISPP ASPPVGWKQV EDATPVINYD LLYAISKLGPGEKYELHAAT DTTPSVVVHV CESDQEKEEE EEMERMRRPK PKIIQTRRPE YTPIHLS.

Q&A

What is the nomenclature history of RCAN1 and its significance?

RCAN1 (Regulator of Calcineurin 1) has undergone several name changes in scientific literature that reflect its discovery context and functional understanding. Originally identified as DSCR1 (Down Syndrome Critical Region 1) by researchers mapping chromosome 21, it was simultaneously discovered as Adapt78 by investigators studying cellular adaptation to oxidative stress . The gene was renamed RCAN1 to better reflect its primary biochemical function as a regulator of calcineurin activity.

The protein has also been referred to as calcipressin 1 and MCIP1 (myocyte-enriched calcineurin interacting protein) in various research contexts. This nomenclature evolution is important to consider when conducting literature searches, as significant research findings may be published under different gene names. When designing experiments or interpreting historical data, researchers should be aware of this nomenclature transition to ensure comprehensive analysis of available literature .

Which RCAN1 isoforms are expressed in human brain, and how are they structurally distinct?

Human brain expresses at least three distinct RCAN1 isoforms resulting from differential splicing and alternative promoters:

  • RCAN1-1 Short (RCAN1-1S): 31 kDa protein

  • RCAN1-1 Long (RCAN1-1L): 38 kDa protein

  • RCAN1-4: Distinct isoform with different expression regulation

These isoforms differ in their N-terminal regions due to alternative promoter usage and differential splicing of exons. The 5'-UTR region of the RCAN1 gene contains multiple regulatory elements that contribute to isoform-specific expression patterns. Researchers have characterized these promoter regions using primer extension assays and gel shift assays to identify transcription initiation sites and regulatory elements .

When investigating RCAN1 in human brain, it's essential to distinguish between these isoforms as they may serve different functional roles. RCAN1-1S specifically has been correlated with GSK-3β levels, while RCAN1-1L and RCAN1-4 do not show this correlation . This suggests isoform-specific functions that should be considered in experimental design.

How does RCAN1 interact with the calcineurin pathway?

RCAN1 functions as a critical regulator of calcineurin (protein phosphatase 2B or PP2B), a calcium/calmodulin-dependent serine/threonine phosphatase. The primary mechanism involves direct physical interaction between RCAN1 and calcineurin subunit A, resulting in inhibition of calcineurin phosphatase activity both in vitro and in vivo .

The regulatory relationship between RCAN1 and calcineurin is complex and bidirectional:

  • RCAN1 physically binds calcineurin and inhibits its phosphatase activity

  • Phosphorylation state of RCAN1 determines whether it acts as an inhibitor or facilitator of calcineurin

  • Phosphorylated RCAN1 can itself become a substrate for calcineurin

This creates a sophisticated regulatory circuit where RCAN1's effects on calcineurin are modulated by its own phosphorylation status. RCAN1 contains an FLISPP motif whose phosphorylation can enhance calcineurin inhibition while also accelerating RCAN1 degradation . For experimental interrogation of this pathway, researchers should consider:

  • Using phosphorylation-specific antibodies to distinguish between different RCAN1 states

  • Employing phosphorylation site mutants to determine functional consequences

  • Implementing pharmacological manipulation of kinases that modify RCAN1

What is the relationship between RCAN1 and GSK-3β, and how can this interaction be studied?

RCAN1 stimulates expression of glycogen synthase kinase-3β (GSK-3β), establishing a critical regulatory link between these important signaling molecules. Research has demonstrated that regulated overexpression of RCAN1 transgene stimulates GSK-3β expression at a post-transcriptional level . This relationship creates an important regulatory circuit, as GSK-3β can also phosphorylate RCAN1, affecting its function.

Specifically, GSK-3β phosphorylates RCAN1 at Ser108, following a priming phosphorylation at Ser112 by BMK1 (big MAP kinase 1) . This phosphorylation can alter RCAN1's effects on calcineurin activity - phosphorylated RCAN1 can release its inhibitory effect on calcineurin.

For studying this relationship, researchers should consider:

  • Correlation analysis: RCAN1-1S levels specifically correlate with GSK-3β levels in human brain tissues, while RCAN1-1L and RCAN1-4 do not show this correlation .

  • Experimental approaches:

    • Co-immunoprecipitation to detect physical interactions

    • Kinase assays to measure GSK-3β-mediated phosphorylation of RCAN1

    • Use of phospho-specific antibodies to detect RCAN1 phosphorylation state

    • GSK-3β inhibitors to determine functional consequences

  • Systems to consider:

    • Cell models: Use of SH-SY5Y neuroblastoma cells for in vitro studies

    • Primary neurons: Isolated from mouse or rat brain

    • Human brain tissue: From normal, DS, or AD patients

How does RCAN1 contribute to neurodegeneration in Down syndrome and Alzheimer's disease?

RCAN1 is implicated in neurodegeneration through several mechanisms supported by both clinical observations and experimental data:

  • Overexpression in pathological states: RCAN1 expression is elevated in the cortex of both Down syndrome (DS) and Alzheimer's disease (AD) patients . This overexpression may result from gene dosage effects in DS (extra copy of chromosome 21) and from stress-responsive activation in AD.

  • Neuronal apoptosis pathway: Overexpression of RCAN1-1 in primary neurons activates caspase-9 and subsequently caspase-3, inducing neuronal apoptosis . This finding provides a direct mechanistic link between RCAN1 overexpression and neuronal death.

  • Stress-response activation: RCAN1 expression can be activated by the stress hormone dexamethasone through a functional glucocorticoid response element (GRE) identified in the RCAN1-1 promoter region . This suggests RCAN1 acts as a mediator of stress-induced neuronal death.

  • Amyloid-beta (Aβ) sensitivity: RCAN1 overexpression renders neurons more vulnerable to apoptosis induced by Aβ, a key pathological protein in AD .

Experimental validation has shown that the neurotoxicity of RCAN1-1 is inhibited in caspase-3 knockout (caspase-3−/−) neurons, confirming the dependence on caspase-3 activation . This suggests potential therapeutic approaches targeting this pathway.

Researchers investigating RCAN1 in neurodegeneration should consider:

  • Using both DS and AD brain tissues for comparative analyses

  • Employing primary neuronal cultures for mechanistic studies

  • Utilizing caspase inhibitors or genetic knockouts to validate apoptotic pathways

  • Examining interactions between RCAN1 and other neurodegenerative factors like Aβ

What experimental systems are most appropriate for studying RCAN1's role in neuronal apoptosis?

Several experimental systems have proven valuable for investigating RCAN1's role in neuronal apoptosis:

  • Primary neuronal cultures:

    • Rat hippocampal and cortical neurons cultivated on poly-d-lysine coated plates

    • Maintained at 37°C in 5% CO₂ for 7-14 days before experimental use

    • Particularly useful for apoptosis studies and overexpression experiments

  • Genetically modified systems:

    • Caspase-3 knockout (caspase-3−/−) mouse neurons have been instrumental in establishing the dependence of RCAN1-induced apoptosis on caspase-3

    • Comparison between wild-type and knockout neurons provides clear mechanistic insights

  • Cell lines:

    • SH-SY5Y human neuroblastoma cells serve as a model for studying RCAN1 promoter regulation and expression

    • Human embryonic kidney 293 cells (HEK293) provide an additional system for molecular studies

  • Viral expression systems:

    • Semliki Forest virus vector (pSFV) with RCAN1 constructs allows efficient gene delivery

    • pSFV-RCAN1 and pSFV-GFP constructs enable visualization and functional studies

    • Viral infection protocols typically involve 1-hour infection followed by 12-18 hour incubation

  • Human tissue samples:

    • Brain tissues from AD patients (available from brain banks)

    • Brain tissues from DS abortuses (17-21 gestational weeks)

    • Provide clinically relevant context for RCAN1 expression studies

For protein detection and quantification, specific methodologies include:

  • Western blotting using custom antibodies (e.g., rabbit anti-RCAN1 polyclonal antibody against C-terminus)

  • 12% SDS-PAGE for RCAN1 detection

  • 16% Tris-Tricine PAGE for caspase-3 and caspase-9 detection

  • Immunostaining with 9E10 antibody for Myc-tagged RCAN1

How is RCAN1 gene expression regulated at the transcriptional level?

RCAN1 expression is regulated through multiple transcriptional mechanisms:

  • Promoter structure: The RCAN1 gene contains multiple promoter regions that drive expression of different isoforms. Researchers have characterized these regions using cloning techniques and luciferase reporter assays .

  • Glucocorticoid regulation: A functional glucocorticoid response element (GRE) has been identified in the RCAN1-1 promoter region, specifically:

    • Located between positions -272 and -237bp

    • Core sequence: 5'-cagctgtcagaaaagcggaactggggacgaggactt-3'

    • Mediates up-regulation by stress hormone dexamethasone

    • Verified through gel shift assays

  • NFAT pathway regulation: RCAN1 is a downstream gene in the NFAT signaling pathway and can be activated by:

    • VEGF

    • Angiotensin II

    • G protein-coupled receptor 54

    • TNF-α

    • Thrombin

    • Calcium ionophores

  • Calcineurin-mediated activation: RCAN1 can be activated by dephosphorylation in neural cells via calcium current increase through L-type calcium channels .

For experimental investigation of RCAN1 transcriptional regulation, researchers have employed:

  • Primer extension assays to determine transcription initiation sites

  • Gel shift assays to verify protein-DNA interactions at regulatory elements

  • Luciferase reporter constructs to quantify promoter activity

  • PCR amplification and cloning of promoter regions (e.g., -684 to +46bp of RCAN1 exon 1)

This complex regulation creates a feedback loop where RCAN1, as an inhibitor of calcineurin, can indirectly regulate its own expression through the calcineurin-NFAT pathway.

What post-translational modifications regulate RCAN1 function, and how can they be detected?

RCAN1 undergoes several post-translational modifications that significantly impact its function:

  • Phosphorylation by multiple kinases:

    • BMK1 (big MAP kinase 1) phosphorylates Ser112, priming for subsequent modifications

    • GSK-3β phosphorylates Ser108 following BMK1 priming

    • NF-κB-inducing kinase (NIK) phosphorylates RCAN1, increasing its stability

    • TAK1 (TGF-β-activated kinase 1) phosphorylates RCAN1 via interaction with TAB2

  • Functional consequences of modifications:

    • Phosphorylation of the FLISPP motif enhances calcineurin inhibition while accelerating RCAN1 degradation

    • Phosphorylated RCAN1 can release inhibition of calcineurin

    • Phosphorylated RCAN1 can itself become a substrate for calcineurin

    • Some phosphorylated forms may act as calcineurin facilitators rather than inhibitors

For experimental detection and analysis of these modifications, researchers should consider:

  • Phospho-specific antibodies:

    • Development of antibodies targeting specific phosphorylation sites

    • Western blotting with phospho-specific antibodies

  • Phosphorylation site mutants:

    • Generation of constructs with alanine substitutions at key phosphorylation sites

    • Expression of mutants in cellular systems to determine functional consequences

  • Kinase inhibitors:

    • Use of specific inhibitors for GSK-3β, BMK1, NIK, and TAK1

    • Assessment of RCAN1 phosphorylation state following inhibitor treatment

  • Mass spectrometry:

    • Identification and quantification of specific phosphorylation sites

    • Temporal analysis of modification patterns

  • In vitro kinase assays:

    • Reconstitution of phosphorylation reactions with purified components

    • Assessment of sequential phosphorylation events

These experimental approaches can provide insights into how the complex pattern of RCAN1 post-translational modifications regulates its function in different cellular contexts and disease states.

How do the three RCAN1 isoforms differ in their regulation of GSK-3β and neuronal survival?

The three RCAN1 isoforms exhibit distinct relationships with GSK-3β and differential effects on neuronal survival:

  • RCAN1-1S (31 kDa):

    • Shows strong correlation with GSK-3β levels in human brain

    • Likely induces GSK-3β production in vivo

    • Primary mediator of RCAN1's neurotoxic effects

  • RCAN1-1L (38 kDa):

    • No correlation with GSK-3β levels observed

    • Distinct regulatory properties from RCAN1-1S despite sharing the same C-terminal region

  • RCAN1-4:

    • No correlation with GSK-3β levels

    • Regulated by different promoter elements than RCAN1-1 variants

    • May have distinct functional properties in neuronal contexts

These findings suggest that RCAN1-1S specifically might be the critical isoform mediating neurodegeneration in Down syndrome and Alzheimer's disease. The isoform-specific effects highlight the importance of distinguishing between RCAN1 variants in experimental design and analysis.

Research approaches to further investigate isoform-specific functions should include:

  • Isoform-specific overexpression:

    • Use of viral vectors expressing individual RCAN1 isoforms

    • Assessment of differential effects on neuronal viability and GSK-3β expression

  • Selective knockdown:

    • Design of siRNAs targeting isoform-specific sequences

    • Assessment of effects on downstream pathways

  • Correlation studies:

    • Analysis of isoform ratios in different brain regions

    • Comparison between normal, DS, and AD brain tissues

  • Protein interaction analysis:

    • Identification of isoform-specific binding partners

    • Characterization of differential effects on calcineurin activity

This research direction could lead to more targeted therapeutic approaches focusing on specific RCAN1 isoforms rather than global RCAN1 inhibition or activation.

What is the proposed equilibrium model between RCAN1, calcineurin, and GSK-3β, and how can it be experimentally tested?

A sophisticated equilibrium model has been proposed to explain the complex relationships between RCAN1, calcineurin, and GSK-3β in cellular homeostasis:

  • Key components of the model:

    • RCAN1 inhibits calcineurin activity through direct binding

    • RCAN1 stimulates GSK-3β expression at a post-transcriptional level

    • GSK-3β can phosphorylate RCAN1, affecting its interaction with calcineurin

    • Calcineurin can dephosphorylate RCAN1, creating a feedback loop

  • Equilibrium dynamics:

    • Cells maintain a balance between these three components

    • Disruption of this equilibrium may contribute to pathological states

    • The model incorporates both inhibitory and stimulatory interactions

    • Different RCAN1 isoforms may differentially affect this equilibrium

Experimental approaches to test this model include:

  • Perturbation analysis:

    • Selective overexpression or knockdown of individual components

    • Observation of compensatory changes in the other components

    • Time-course studies to capture dynamic equilibrium shifts

  • Pharmacological interventions:

    • Calcineurin inhibitors (e.g., cyclosporin A, FK506)

    • GSK-3β inhibitors (e.g., lithium, SB216763)

    • Assessment of effects on the third component

  • Mathematical modeling:

    • Development of computational models incorporating known parameters

    • Simulation of perturbations and prediction of system responses

    • Validation of model predictions with experimental data

  • Correlation studies in disease states:

    • Quantification of all three components in DS and AD brain tissues

    • Assessment of equilibrium disruption in pathological states

    • Correlation with markers of cellular stress and neurodegeneration

This equilibrium model provides a conceptual framework for understanding how RCAN1 dysregulation might contribute to neurodegeneration through disruption of a delicate balance between key signaling components. Further experimental validation of this model could provide insights into potential points of therapeutic intervention.

Product Science Overview

Introduction

The Regulator of Calcineurin 1 (RCAN1), also known as Down Syndrome Critical Region 1 (DSCR1), is a protein encoded by the RCAN1 gene located on chromosome 21q22. This protein plays a crucial role in various physiological processes, including neurodegeneration, mitochondrial dysfunction, inflammation, and protein glycosylation .

Structure and Isoforms

RCAN1 has two main isoforms, RCAN1-1 and RCAN1-4, which result from differential promoter use and first exon choice . These isoforms have distinct functions and regulatory mechanisms. RCAN1-1 consists of 252 amino acids, while RCAN1-4 comprises 197 amino acids .

Function

RCAN1 is an endogenous inhibitor of the serine/threonine phosphatase calcineurin. Calcineurin is a heterodimer composed of a catalytic subunit (calcineurin A) and a regulatory subunit (calcineurin B) . By binding to the catalytic domain of calcineurin A, RCAN1 inhibits calcineurin-dependent transcriptional responses . This inhibition affects various signaling pathways, including the nuclear factor of activated T cells (NFAT) and NF-κB pathways .

Role in Diseases

RCAN1 is implicated in several diseases, most notably Down syndrome and Alzheimer’s disease. Overexpression of RCAN1 in the brain of Down syndrome fetuses is associated with neurofibrillary tangles, similar to those observed in Alzheimer’s disease . Additionally, RCAN1 plays a role in cardiovascular diseases (CVDs) by participating in the development and functional maintenance of the cardiovascular system .

Research and Therapeutic Potential

Recent studies have highlighted the potential of RCAN1 as a therapeutic target. For instance, RCAN1 has been identified as a novel RNA-binding protein that regulates neuronal apoptosis . The discovery of RNA aptamers that block RCAN1-induced inhibition of NFAT and NF-κB signaling pathways offers promising avenues for neuroprotective therapies .

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.