KEGG: pon:100173979
STRING: 9601.ENSPPYP00000010220
MAPRE2 (Microtubule Associated Protein RP/EB Family Member 2) belongs to the evolutionary conserved microtubule plus-end tracking proteins, specifically the end-binding (EB) family. Unlike its paralogs MAPRE1 and MAPRE3, MAPRE2 is not required for the persistent growth and stabilization of microtubules . Instead, it plays crucial roles in:
Reorganization of microtubules during early apical-basal differentiation in epithelia
Regulation of cell adhesion and focal adhesion turnover
Mitotic progression and genome stability
Neural crest cell migration, particularly cranial neural crest cells
Cardiac electrophysiology and ventricular conduction
The protein contains a highly conserved calponin-homology (CH) domain that is responsible for tracking and interacting with the growing plus-end tips of microtubules . MAPRE2 acts as an adaptor to recruit signaling molecules to microtubule ends and interacts with proteins such as MAP4K4 and HAX1 in the focal adhesion turnover complex .
MAPRE2 mutations significantly alter neural crest cell migration, which has been demonstrated through multiple methodological approaches:
In vitro methodologies:
Scratch assay combined with live imaging allows tracking of single neural crest cells as they migrate into an open area
Nuclei labeling with H2B-RFP for easy tracking of individual cells
Quantitative assessment of migration speed and directionality
Key findings:
Loss-of-function mutations (Q152* heterozygous or homozygous) result in decreased migration speed of cranial neural crest cells
The N68S/N68S mutation, in contrast, increases migration speed compared to isogenic controls
Haploinsufficiency is sufficient to cause migration phenotypes, as seen in patients with these mutations
In vivo approaches:
Xenotransplantation of human-derived MAPRE2 neural crest progenitors into developing chicken embryos
Zebrafish models showing that knockdown of MAPRE2 results in craniofacial defects and aberrant branchial arch formation
MAPRE2 plays a critical role in focal adhesion (FA) dynamics through the following mechanisms:
Molecular interactions:
MAPRE2 interacts with MAP4K4 and HAX1 in the focal adhesion turnover complex
It influences wound healing and cell motility through these interactions
Experimental observations:
Microtubules specifically grow toward focal adhesions and promote FA turnover rate at the cell periphery
MAPRE2 appears to serve as an adaptor to recruit signaling molecules to microtubule ends
Quantitative findings:
MAPRE2 Q152* knockdown lines (both heterozygous and homozygous) show significantly larger vinculin-positive focal adhesion spots compared to wild-type controls
This can be visualized using immunohistochemistry and confocal microscopy of iPSC-derived cranial neural crest cells plated on fibronectin-coated plates
The altered focal adhesion dynamics likely contribute to the abnormal cell motility observed in MAPRE2 mutant cranial neural crest cells, providing a mechanism for the craniofacial phenotypes seen in patients.
The end-binding (EB) protein family has three main members (EB1, EB2, and EB3), with MAPRE2 encoding EB2. Key differences include:
These differences indicate that despite sharing structural similarities, MAPRE2 has evolved distinct functions from its paralogous proteins, particularly in specialized cellular contexts.
Creating reliable MAPRE2 loss-of-function models requires careful consideration of several factors:
CRISPR/Cas9 system for germline knockout:
Target the calponin homology domain for functional disruption
Generate isogenic control lines simultaneously for proper comparison
Validate knockout efficiency through both Western blotting and RT-qPCR
Assess both mRNA levels (using primers in different exons) and protein expression
siRNA for transient knockdown:
Design and validate multiple siRNAs targeting different regions of MAPRE2
Confirm knockdown efficiency by Western blot in established cell lines (e.g., HeLa)
Verify knockdown in target cells (e.g., iPSC-CMs) by immunofluorescence
Validation considerations:
For Q152* mutation models, expect approximately 95% reduction in mRNA and 96% reduction in protein for homozygous knockouts
For heterozygous models, expect approximately 45% reduction in mRNA and 55% reduction in protein
For N68S mutations, protein levels may be reduced despite normal mRNA levels due to protein instability (observed 70% reduction)
Rescue experiments:
Include rescue controls to confirm phenotype specificity
The "Goldilocks effect" should be considered - both overexpression and insufficient protein can lead to similar phenotypes
MAPRE2 has recently been implicated in Brugada syndrome through genome-wide association studies. To investigate its role in cardiac electrophysiology:
Zebrafish model approaches:
Generate mapre2 knockout zebrafish using CRISPR/Cas9
Perform voltage mapping of larval hearts at 5 days post-fertilization
Measure ventricular maximum upstroke velocity of action potentials and conduction velocity
Conduct ECG on adult fish hearts to detect QRS prolongation
Use patch clamping to measure sodium current density in ventricular myocytes
iPSC-CM approaches:
Create MAPRE2 knockdown or knockout in human induced pluripotent stem cell-derived cardiomyocytes
Perform patch clamping to detect arrhythmias
Use immunocytochemistry to examine adherens junction organization
Assess N-cadherin localization and detyrosinated tubulin levels
Key findings to validate:
MAPRE2 loss typically leads to decreased ventricular maximum upstroke velocity and conduction velocity
QRS prolongation is observed in ECGs of knockout models
Sodium current density is decreased in knockout ventricular myocytes
Adherens junctions appear disorganized with mislocalization of mature N-cadherin
MAPRE2 affects microtubule dynamics in several ways that can be studied using these approaches:
Microtubule plus-end tracking:
Transfect cells with EB3-GFP to visualize microtubule plus-ends
Perform live-cell imaging to track individual microtubules
Generate kymographs for quantitative analysis
Expected results in MAPRE2 knockdown:
Increased microtubule growth velocity (1.11-1.26 fold increase)
Increased microtubule growth distance (1.22-1.34 fold increase)
Tubulin post-translational modification analysis:
Use Western blotting with antibodies specific to detyrosinated α-tubulin
Calculate the ratio of detyrosinated to total α-tubulin
Perform immunofluorescence to visualize tubulin modifications in situ
Microtubule architecture assessment:
Immunofluorescence with antibodies against α-tubulin
Confocal microscopy to assess microtubule bundling and straightness
Computational analysis of microtubule organization and dynamics
Rescue strategies:
Knockdown of tubulin tyrosine ligase (TTL) in MAPRE2 knockout models can rescue tubulin detyrosination and associated phenotypes
The structural analysis of MAPRE2 mutations, particularly the N68S mutation, requires multifaceted approaches:
Computational structure analysis:
Use homology modeling based on MAPRE1 structures (e.g., 1VKA template)
Analyze conservation scores across multiple species - residue 68 shows 100% conservation, suggesting structural/functional importance
Calculate protein stability changes induced by mutations using tools like DUET, FoldX, or Rosetta
Assess aggregation propensity changes using tools like TANGO
Expression and stability analysis:
Compare mRNA and protein levels between mutant and isogenic control lines
For N68S mutation, note that despite normal mRNA levels, protein expression may be reduced by about 70%
Structural impact predictions:
N68S mutation increases aggregation propensity of the APR from 41.8 to 44.6
Although this increase alone might not significantly affect aggregation tendency, the mutation is destabilizing
The destabilization can hamper native folding by requiring higher energy states, indirectly increasing aggregation
Microtubule binding assays:
In vitro microtubule-polymerization assays can detect changes in binding affinity
Previous studies with missense mutations showed increased affinity and dwell time at microtubules, indicating gain-of-function effects
Several contradictions and knowledge gaps exist in the current MAPRE2 literature:
Contradictory findings on tubulin detyrosination:
Mutation effects - Loss vs. Gain of function:
Some MAPRE2 mutations (Q152*) act through loss-of-function mechanisms
Other mutations (missense mutations in the CH domain) show increased microtubule binding, suggesting gain-of-function effects
The "Goldilocks effect" hypothesis suggests that both overactive and insufficient protein can lead to similar clinical pathologies
Tissue-specific effects:
How MAPRE2 functions differently in diverse tissues (cardiac, neural crest, epithelial) remains unclear
The 43 amino acid segment unique to MAPRE2 (not present in EB1 or EB3) has an unknown function
Research gaps:
Detailed 3D structure of MAPRE2 remains unknown
Complete interactome of MAPRE2 across different cell types is not established
Mechanistic understanding of how MAPRE2 variants lead to skin creases phenotype is incomplete
The connection between MAPRE2, microtubule dynamics, and sodium channel function needs further clarification
To effectively study MAPRE2 in neural crest development, optimized differentiation protocols are essential:
Neural crest differentiation protocol:
Start with induced pluripotent stem cells (iPSCs)
Adapt protocols from previous studies (e.g., references 26,27 mentioned in source material)
Validate cranial neural crest identity through gene expression analysis of specific markers:
Quality control metrics:
Flow cytometry for surface markers (p75, HNK1)
Immunofluorescence for neural crest transcription factors
RT-qPCR for quantitative assessment of marker expression
Single-cell RNA sequencing to confirm population homogeneity
Experimental considerations:
Generate isogenic control lines using CRISPR/Cas9 technology
Create both heterozygous and homozygous mutant lines
Ensure cranial identity of neural crest cells to study craniofacial development accurately
Consider timing of differentiation and maturation for phenotypic analysis
Migration assays for functional assessment: