MAPRE2 binds to the growing plus-ends of microtubules via its CH domain, regulating microtubule stability and dynamics . Key findings include:
Microtubule growth regulation: Knockdown in human iPSC-derived cardiomyocytes increases microtubule growth velocity (8.05 µm/min → 10.17 µm/min) and distance (5.76 µm → 7.69 µm) .
Neural crest migration: Mutations in MAPRE2 disrupt cranial neural crest migration in vitro and in chicken embryo xenotransplants, leading to craniofacial defects .
MAPRE2 influences sodium channel (Na<sub>V</sub>) localization and function:
Electrophysiological defects: mapre2 knockout in zebrafish causes slowed ventricular conduction and reduced Na<sub>V</sub> current density .
Adherens junction integrity: MAPRE2 loss disrupts N-cadherin (Ncad) localization at adherens junctions, impairing cell-cell adhesion and Na<sub>V</sub> membrane density .
Knockdown of ttl (tubulin tyrosine ligase) restores detyrosinated α-tubulin levels and rescues Na<sub>V</sub> function in mapre2-deficient models, highlighting microtubule post-translational modification as a therapeutic target .
Data from the Human Protein Atlas and experimental models reveal MAPRE2 expression in diverse tissues:
Genetic basis: Missense mutations (e.g., p.Asn68Ser, p.Gln152*) in the CH domain cause craniofacial dysmorphisms and cleft palate .
Mechanism: Altered MAPRE2-microtubule interactions disrupt focal adhesion dynamics and neural crest migration .
Genetic link: A locus near MAPRE2 (intron 2) is associated with BrS, though direct functional evidence remains limited .
MAPRE2, also known as EB2 (End-Binding protein 2), is a member of a family of three proteins (EB1, EB2, and EB3) that bind to the growing plus-ends of microtubules via their N-terminal domain and interact with other partners via their C-terminus. MAPRE2 contains a calponin homology (CH) domain responsible for tracking and interacting with the plus-end tip of growing microtubules . Unlike other family members, MAPRE2 has a unique 43 amino acid segment at its N-terminus not present in EB1 or EB3, suggesting specialized functions. The protein demonstrates 87.4% similarity between human and zebrafish homologs, indicating high evolutionary conservation .
The functional significance of MAPRE2 is evident in its regulation of microtubule dynamics. Knockdown experiments have demonstrated that MAPRE2 loss of function leads to increased microtubule growth velocity and distance, affecting microtubule stability and post-translational modifications such as tubulin detyrosination .
While the search results don't provide comprehensive information on tissue-specific expression patterns, research indicates that MAPRE2 plays critical roles in both cardiac tissue and neural crest cells. In cardiomyocytes, MAPRE2 influences sodium channel function and ventricular conduction . In cranial neural crest cells, MAPRE2 affects migration capabilities and focal adhesion formation .
The regulatory mechanisms controlling MAPRE2 expression remain an active area of investigation. Current research methodologies for studying MAPRE2 expression include RT-qPCR for mRNA quantification and Western blotting for protein expression analysis, as demonstrated in studies of induced pluripotent stem cell (iPSC) lines carrying various MAPRE2 mutations .
MAPRE2 demonstrates remarkable evolutionary conservation, particularly at functionally critical residues. Analysis of homologous proteins across diverse organisms, including those with high evolutionary distances from humans such as Arabidopsis thaliana and Saccharomyces cerevisiae, reveals 100% conservation at certain residues (e.g., position 68) .
This high degree of conservation suggests essential structural and functional importance. For research purposes, this conservation validates the use of model organisms like zebrafish for studying MAPRE2 function. The zebrafish homolog shows 87.4% similarity at the protein level compared to human MAPRE2, making it a valuable model for investigating MAPRE2-related phenotypes and mechanisms .
MAPRE2 plays a crucial role in maintaining normal cardiac electrophysiology through its effects on microtubule dynamics and subsequent impacts on sodium channel function and intercellular junctions. Research using mapre2 knockout zebrafish models has revealed that MAPRE2 influences ventricular conduction and voltage-gated sodium channel (Nav) function .
Methodologically, these findings were established through:
Voltage mapping of isolated hearts, which showed decreased ventricular maximum upstroke velocity of the action potential and reduced conduction velocity in mapre2 knockout models
Patch clamping experiments demonstrating altered sodium current properties
Immunocytochemistry showing disruption of adherens junctions
The mechanism linking MAPRE2 to cardiac function involves its regulation of microtubule dynamics, which affects both the subcellular localization of sodium channels and the integrity of adherens junctions essential for proper electrical conduction between cardiomyocytes .
Brugada syndrome (BrS) is a cardiac arrhythmia disorder traditionally associated with SCN5A mutations, which account for only about 20% of cases. A genome-wide association study by Bezzina et al. identified a novel locus within intron 2 of MAPRE2, implicating microtubule dynamics in BrS pathophysiology .
Research using zebrafish models with mapre2 loss of function demonstrated a ventricular phenotype consistent with SCN5A loss of function associated with Brugada syndrome. Specifically, mapre2 knockout resulted in:
Ventricular conduction slowing
Decreased sodium channel function
Arrhythmogenesis
Disruption of adherens junctions
These phenotypes were associated with decreased detyrosinated α-tubulin and altered microtubule dynamics. The mechanistic connection appears to involve MAPRE2's role in regulating microtubule stability, which affects sodium channel density at the cell membrane and results in ventricular conduction defects .
Based on the search results, two complementary model systems have proven effective for studying MAPRE2 function in cardiac research:
Zebrafish models:
Both germline mapre2 knockout zebrafish (using CRISPR/Cas9) and morpholino-based knockdown approaches have been successfully employed
Advantages include cardiac electrophysiology similar to humans, optical transparency for imaging, and efficient genetic manipulation
Allow for functional studies including ECG recording, voltage mapping, and patch clamping
Human iPSC-derived cardiomyocytes (iPSC-CMs):
Enable MAPRE2 knockdown or knockout in human cells
Allow for direct measurement of microtubule dynamics through live-cell imaging with EB3-GFP
Permit patch clamping experiments to assess ion channel function
The combination of these models offers complementary strengths. Zebrafish provide an in vivo system for studying cardiac conduction and arrhythmogenesis, while iPSC-CMs allow detailed mechanistic studies of microtubule dynamics in human cells. Both systems support comprehensive functional assessments including electrophysiological measurements, microtubule tracking, and immunocytochemistry .
MAPRE2 plays a significant role in neural crest cell migration, particularly affecting cranial neural crest cells (CNCCs). Research using patient-derived induced pluripotent stem cells (iPSCs) with MAPRE2 mutations demonstrated altered migration capabilities in these cells .
Methodologically, this has been studied through:
In vitro scratch assays with live imaging: These experiments tracked single CNCCs as they migrated into an open area, revealing that cells with MAPRE2 mutations (either heterozygous or homozygous Q152* stop mutations) displayed significantly lower migration speeds compared to isogenic controls .
Xenotransplantation experiments: Neural crest progenitors were transplanted into developing chicken embryos, confirming migration defects in vivo .
The mechanism appears to involve focal adhesion (FA) dynamics. Immunohistochemistry and confocal microscopy revealed that MAPRE2-deficient CNCCs had larger vinculin-positive focal adhesion spots compared to control cells, suggesting altered cell-substrate interactions that impair efficient migration .
MAPRE2 mutations have been implicated in Congenital Symmetrical Circumferential Skin Creases type 2 (CSC-KT), a condition characterized by craniofacial dysmorphisms and cleft palate . Research in both human patients and zebrafish models has provided insights into these developmental abnormalities.
Studies in zebrafish embryos demonstrated that MAPRE2 knockdown resulted in:
Interestingly, rescue experiments suggested a "Goldilocks effect" where either overactive or insufficient MAPRE2 leads to similar clinical pathologies in branchial arch maturation . This suggests that precise regulation of MAPRE2 levels is critical for normal craniofacial development.
The connection between MAPRE2 and craniofacial development appears to be mediated through its effects on neural crest cell migration, which is essential for proper formation of craniofacial structures during embryonic development .
Based on the search results, several experimental approaches have proven effective for studying MAPRE2 in neural development:
Patient-derived iPSC models:
CRISPR/Cas9 genome editing:
Functional assays:
Molecular validation techniques:
These approaches collectively enable comprehensive investigation of MAPRE2 function in neural crest development, from molecular mechanisms to cellular behaviors and developmental outcomes.
Based on the search results, several genetic manipulation techniques have been successfully employed to study MAPRE2 function:
CRISPR/Cas9 genome editing:
For developing isogenic control lines
Key methodological considerations include:
Morpholino-based knockdown:
siRNA-mediated knockdown:
The choice between these approaches depends on research questions, with CRISPR/Cas9 preferred for stable genetic models, morpholinos for developmental studies, and siRNA for acute cellular experiments. Combined approaches provide complementary insights into MAPRE2 function across different temporal and spatial scales.
Advanced imaging techniques have been instrumental in understanding MAPRE2's role in microtubule dynamics. Based on the search results, the following approaches have proven effective:
Microtubule plus-end tracking with live-cell imaging:
Immunocytochemistry for post-translational modifications:
Confocal microscopy for focal adhesion analysis:
These methods collectively enable researchers to characterize how MAPRE2 influences microtubule dynamics, stability, and interactions with cellular structures. The results from these imaging approaches revealed that MAPRE2 loss of function increases microtubule growth velocity and distance, decreases tubulin detyrosination, and affects focal adhesion morphology .
To measure the functional consequences of MAPRE2 alterations, researchers have employed several sophisticated techniques targeting different cellular and physiological processes:
Cardiac electrophysiology assessments:
Cell migration and adhesion analyses:
Junction integrity assessments:
Rescue experiments:
These methodologies provide complementary insights into how MAPRE2 affects cellular functions across different biological contexts, from cardiac conduction to neural crest migration.
A significant challenge in MAPRE2 research involves contradictory findings regarding its effects on microtubule post-translational modifications. The search results highlight one such contradiction:
In MAPRE2 loss-of-function models: Research showed a decrease in α-tubulin detyrosination .
In mouse models of cardiomyopathy: Studies found an increase in detyrosinated α-tubulin .
To address such contradictions, researchers should consider:
Context-specific effects: MAPRE2 may function differently depending on cell type, developmental stage, or disease context. Systematic comparison across multiple models and conditions is essential.
Compensatory mechanisms: Other microtubule-associated proteins or regulatory pathways may compensate for MAPRE2 loss in different ways across models.
Methodological approach:
Use multiple, complementary methods to measure microtubule modifications
Include appropriate controls and isogenic lines to minimize confounding factors
Consider the timing of measurements (acute versus chronic effects)
Evaluate multiple post-translational modifications simultaneously
Functional validation: Correlate changes in microtubule modifications with functional outcomes to determine physiological relevance.
The search results suggest a possible resolution through mechanistic research, demonstrating that MAPRE2 loss of function increases microtubule dynamics, potentially leading to decreased microtubule stability and subsequent reduction in detyrosination . This mechanistic insight helps reconcile apparently contradictory observations.
MAPRE2 research utilizes diverse model systems, each with strengths and limitations. Integrating findings across these systems presents several challenges:
Species-specific differences:
Despite 87.4% protein similarity between human and zebrafish MAPRE2 , species-specific differences in expression patterns, interacting partners, or regulatory mechanisms may exist.
Methods for addressing this include: using multiple model organisms, focusing on highly conserved domains, and validating key findings across species.
In vitro versus in vivo contexts:
Cell culture models may not recapitulate the complex three-dimensional environment and multicellular interactions present in vivo.
Researchers should combine in vitro mechanistic studies with in vivo functional validation when possible.
Developmental timing:
MAPRE2 functions may vary across developmental stages, making direct comparisons challenging.
Careful staging and temporal analyses are essential for meaningful comparisons.
Genetic background effects:
Genetic modifiers may influence MAPRE2-associated phenotypes.
Use of isogenic controls and multiple genetic backgrounds can help address this challenge.
Methodological differences:
Variations in experimental protocols, measurement techniques, and data analysis approaches may contribute to apparent discrepancies.
Standardization of key methodologies and detailed reporting of experimental conditions are important.
The search results demonstrate successful integration through complementary approaches, such as combining zebrafish models for in vivo cardiac function with human iPSC-derived cardiomyocytes for detailed microtubule dynamics studies , or validating findings from patient-derived iPSCs through xenotransplantation experiments .
While the search results don't explicitly discuss therapeutic interventions targeting MAPRE2, they suggest several potential approaches based on mechanistic insights:
Modulation of microtubule post-translational modifications:
The rescue experiments knocking down ttl (tubulin tyrosine ligase) restored normal detyrosinated tubulin levels, adherens junction formation, and sodium channel function in mapre2 loss-of-function models .
This suggests that targeting enzymes involved in tubulin post-translational modifications could represent a novel therapeutic strategy for MAPRE2-related cardiac conduction disorders.
Stabilization of adherens junctions:
Focal adhesion modulation:
Precision gene editing or gene therapy:
Ion channel targeting:
These approaches represent potential "novel therapeutic approaches for treating conduction disorders" and developmental conditions associated with MAPRE2 dysfunction, though significant preclinical research would be needed before clinical translation.
Table 1: Comparison of Key MAPRE2 Findings Across Research Models
This comparative analysis reveals complementary findings across different model systems, with each providing unique insights into MAPRE2 function. The zebrafish model provides the most comprehensive functional characterization in vivo, while human cell models offer detailed insights into specific cellular processes. The consistency in phenotypes related to cellular adhesion, migration, and cytoskeletal dynamics across models supports conserved functions of MAPRE2 across species and cell types.
Based on the search results, MAPRE2 dysfunction contributes to distinct disease phenotypes through several molecular mechanisms:
Table 2: Molecular Mechanisms Linking MAPRE2 to Disease Phenotypes
These mechanisms highlight how MAPRE2, through its fundamental role in microtubule regulation, can affect diverse cellular processes depending on tissue context. In cardiac tissue, the impact on ion channel function and intercellular junctions primarily affects electrophysiology, while in neural crest cells, the effects on focal adhesions and cell motility disrupt developmental migration patterns.
The search results suggest a "Goldilocks effect" whereby either overactive or insufficient MAPRE2 can lead to similar pathologies , indicating that precise regulation of this protein is critical for normal function across different tissues. This mechanistic understanding provides potential targets for tissue-specific therapeutic interventions.
Based on current research approaches identified in the search results, several emerging technologies could significantly advance MAPRE2 research:
Advanced live-cell imaging techniques:
Super-resolution microscopy to visualize microtubule dynamics and interactions at nanoscale resolution
Correlative light and electron microscopy (CLEM) to connect molecular-level changes with ultrastructural alterations
Lattice light-sheet microscopy for extended live imaging with reduced phototoxicity
Multi-omics approaches:
Spatial transcriptomics to map MAPRE2-dependent gene expression changes within tissue contexts
Proteomics to identify the complete interactome of MAPRE2 in different cell types
Metabolomics to assess downstream metabolic consequences of altered microtubule dynamics
Advanced genetic engineering:
Base editing or prime editing for precise modification of specific MAPRE2 residues
Optogenetic or chemogenetic control of MAPRE2 activity or localization
Tissue-specific and inducible MAPRE2 manipulation in animal models
Organ-on-chip and organoid technologies:
Cardiac organoids to study MAPRE2 in three-dimensional tissue architecture
Multi-cell type organ-on-chip models to investigate MAPRE2 in complex tissue interactions
Patient-specific organoids to model individual disease variants
Computational approaches:
Machine learning for automated analysis of microtubule dynamics and cell migration
Molecular dynamics simulations to predict effects of MAPRE2 mutations
Network modeling to integrate MAPRE2 into broader cytoskeletal and signaling networks
These technologies would address current limitations in understanding MAPRE2 function across spatial and temporal scales, from molecular interactions to tissue-level phenotypes.
Analysis of the search results reveals several critical knowledge gaps in MAPRE2 research that warrant further investigation:
Tissue-specific functions:
How does MAPRE2 function differ across cell types and tissues?
What explains the tissue-specific manifestations of MAPRE2 mutations despite its ubiquitous expression?
Regulatory mechanisms:
How is MAPRE2 expression and activity regulated under normal and pathological conditions?
What signaling pathways modulate MAPRE2 function?
Interaction with other microtubule-associated proteins:
Mechanism of ion channel regulation:
How precisely does MAPRE2 influence sodium channel localization and function in cardiomyocytes?
Are other ion channels similarly affected by MAPRE2 dysfunction?
Developmental dynamics:
How does MAPRE2 function change during development and differentiation?
What developmental processes beyond cardiac conduction and neural crest migration depend on MAPRE2?
Therapeutic targeting:
Human disease associations:
Beyond Brugada syndrome and CSC-KT, what other human conditions might involve MAPRE2 dysfunction?
How do common genetic variants in MAPRE2 influence disease susceptibility?
The MAPRE2 gene is located on chromosome 18 and encodes a protein that shares significant homology with the adenomatous polyposis coli (APC) protein-binding EB1 gene family . The protein consists of several domains that facilitate its interaction with microtubules and other cellular components. Alternative splicing of the MAPRE2 gene results in multiple transcript variants, which may lead to the production of different protein isoforms .
MAPRE2 is essential for maintaining spindle symmetry during mitosis, a critical process for accurate chromosome segregation and cell division . It stabilizes microtubules by binding to their plus-ends, thereby promoting microtubule polymerization and anchoring them at centrosomes . This stabilization is vital for the proper formation and function of the mitotic spindle, ensuring that chromosomes are evenly distributed to daughter cells during cell division.
Additionally, MAPRE2 is involved in cell migration by regulating the dynamics of the microtubule cytoskeleton . It interacts with other proteins to facilitate the organization and elongation of microtubules, contributing to the polarized movement of cells .
MAPRE2 has been implicated in various diseases and conditions. It is thought to play a role in the tumorigenesis of colorectal cancers and the proliferative control of normal cells . Abnormal expression or mutations in the MAPRE2 gene may disrupt microtubule dynamics, leading to defects in cell division and contributing to cancer development.
Moreover, diseases associated with MAPRE2 include congenital symmetric circumferential skin creases and multiple benign circumferential skin creases on limbs . These conditions are characterized by the presence of multiple skin folds, which may be linked to underlying defects in microtubule function and cell proliferation.
Human recombinant MAPRE2 is widely used in research to study its role in microtubule dynamics and cell division. By using recombinant proteins, scientists can investigate the specific functions and interactions of MAPRE2 in a controlled environment, providing insights into its mechanisms and potential therapeutic targets.