Recombinant Mouse Ankyrin-2 (Ank2), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and serves as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
The tag type is determined during production. Please specify your preferred tag type for prioritized development.
Synonyms
Ank2; AnkBAnkyrin-2; ANK-2; Ankyrin-B; Brain ankyrin
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Ankyrin-2 plays a crucial role in the localization and membrane stabilization of ion transporters and channels in various cell types, including cardiomyocytes and striated muscle cells. In skeletal muscle, it's essential for the proper localization of dystrophin (DMD) and dystonin (DCTN4), and for the formation/stability of costamere-associated microtubules and neuromuscular junctions. In cardiomyocytes, it coordinates the assembly of the Na/Ca exchanger (SLC8A1/NCX1), Na/K ATPases (ATP1A1 and ATP1A2), and inositol 1,4,5-trisphosphate (InsP3) receptors at sarcoplasmic reticulum/sarcolemma sites. It's required for the expression and targeting of beta-spectrin (SPTBN1) in neonatal cardiomyocytes and regulates neonatal cardiomyocyte contraction rate. In rod photoreceptor inner segments, it coordinates the expression of Na/K ATPase, Na/Ca exchanger, and beta-2-spectrin (SPTBN1). Additionally, it participates in endocytosis, intracellular protein transport, associates with phosphatidylinositol 3-phosphate (PI3P)-positive organelles, and binds dynactin to facilitate long-range cell motility. It recruits RABGAP1L to PI3P-positive early endosomes, where RABGAP1L inactivates RAB22A, promoting polarized trafficking to the leading edge of migrating cells. It is part of the ANK2/RABGAP1L complex, essential for polarized recycling of the fibronectin receptor (ITGA5 ITGB1) to the plasma membrane, enabling directional cell migration.
Gene References Into Functions
  1. Spectrin (alpha/beta) / ankyrin B cytoskeletal and signaling proteins are key regulators of neuronal T-type calcium channels. PMID: 29720258
  2. Ankyrin-B deficiency leads to increased cell surface GLUT4 and cell-autonomous adiposity in humans and mice. PMID: 29133412
  3. AnkB binds RabGAP1L, recruiting it to PI3P-positive organelles, where it inactivates Rab22A, promoting polarized trafficking to the leading edge of migrating fibroblasts. a5b1-integrin requires an AnkB/RabGAP1L complex for polarized recycling. PMID: 27718357
  4. Increased pro-arrhythmogenic Ca(2+) sparks and waves in AnkB(+/-) hearts result from enhanced CaMKII-mediated RyR phosphorylation due to higher junctional [Ca(2+)] and local CaMKII activation. PMID: 27131508
  5. Two functionally distinct ankyrin-B isoforms in the heart demonstrate that ANK2 gene alternative splicing regulates ankyrin-B protein interactions. PMID: 26109584
  6. AnkB is necessary for Prx membrane anchoring and maintenance of lens fiber cell hexagonal geometry, membrane skeleton organization, and biomechanics. PMID: 26538089
  7. Ankyrin-B deficiency causes a metabolic syndrome combining pancreatic beta cell insufficiency and peripheral insulin resistance. PMID: 26168218
  8. PIK3C3, dynactin, and AnkB interactions promote axonal transport of organelles and are necessary for normal axon length. PMID: 25533844
  9. Ankyrin-B interacts with Cav2.1 and Cav2.2, affecting their in vivo targeting. PMID: 24394417
  10. Ankyrin-B is a component of metazoan cardioprotection in heart failure. PMID: 22778271
  11. AnkB reduction alters cardiac Na and Ca transport, enhancing coupled RyR openings, leading to more frequent Ca sparks and waves without affecting total SR Ca leak. PMID: 22406428
  12. Ankyrin-B interacts with dynactin-4 and dystrophin; dynactin-4 collaborates with dystrophin in coordinating costamere-aligned microtubules. PMID: 21186323
  13. Ankyrin-B regulates Kir6.2 membrane expression and function in the heart. PMID: 20610380
  14. AnkB's role in cardiac automaticity is demonstrated through its influence on the atrial distributed pacemaker complex. PMID: 20525877
  15. Impaired glycemic regulation due to loss of ankyrin-B-dependent IP3R stabilization is a potential type 2 diabetes risk factor. PMID: 20234002
  16. The ankyrin-B C-terminal domain determines the activity of ankyrin-B/G chimeras. PMID: 11781319
  17. Mice heterozygous for an ankyrin-B null mutation exhibit haploinsufficiency and arrhythmia similar to humans. PMID: 12571597
  18. Ankyrin-B influences Inositol 1,4,5-trisphosphate receptor localization and stability in neonatal cardiomyocytes. PMID: 14722080
  19. The ankyrin-B-based complex is a cardiomyocyte adaptation involved in cytosolic Ca2+ modulation. PMID: 16292983
  20. Ankyrin-B impacts cardiac function, cardiac death, and premature senescence. PMID: 17940615
  21. ANK2 alternative splicing produces unique polypeptides with diverse roles in cardiac function. PMID: 18782775
  22. ANK2-deficient mice show abnormal membrane expression of NCX1, Na+/K+ ATPase, IP3R, and Ca(V)1.3. Loss of ank2 promotes slow, irregular Ca2+ release and afterdepolarizations in sinoatrial node cardiomyocytes. PMID: 19098452
  23. Ankyrin-B is an adaptor for sarcolemmal localization of dystrophin and dynactin-4. PMID: 19109891
  24. Ankyrin(B) regulates axon guidance via cyclic AMP. PMID: 19110015
Database Links
Subcellular Location
Cytoplasm, cytoskeleton. Cytoplasm, myofibril, sarcomere, M line. Cell membrane. Cell junction, synapse, postsynaptic cell membrane. Early endosome. Recycling endosome. Lysosome. Mitochondrion. Cytoplasm, myofibril, sarcomere, Z line. Cell membrane, sarcolemma, T-tubule.
Tissue Specificity
Detected in eye lens fiber cells (at protein level). In the retina, expressed in the inner segments of rod photoreceptors. Expressed in cardiomyocytes, as well as in skeletal muscles. Also detected in brain and pancreas, as well as in kidney and spleen (a

Q&A

What is the basic structure of mouse Ankyrin-2 and how does it differ from other ankyrin family members?

Mouse Ankyrin-2 (encoded by the ank2 gene) is a member of the ankyrin family of spectrin-binding proteins. The full-length 440-kDa Ankyrin-2 (also known as Ankyrin-B) is encoded by a 13-kb mRNA transcript, while alternative splicing produces smaller 220-kDa and 150-kDa isoforms from 8-kb and 5-kb transcripts, respectively . The protein contains three functional domains: an N-terminal membrane-binding domain composed of 24 ankyrin repeats, a spectrin-binding domain, and a regulatory domain.

Unlike Ankyrin-1 (Ankyrin-R), which is predominantly expressed in erythrocytes, Ankyrin-2 shows high expression in the nervous system, particularly in premyelinated axons. While both ankyrin family members serve as adaptors linking membrane proteins to the spectrin-based cytoskeleton, they differ in tissue distribution and binding partners, with Ankyrin-2 showing specialized functions in neuronal tissues .

What are the major expression patterns of Ankyrin-2 during mouse development?

Ankyrin-2 exhibits specific temporal and spatial expression patterns during mouse development. The 440-kDa isoform is predominantly expressed in developing neuronal tissues, with high levels detected in premyelinated axon tracts of the developing nervous system . Notably, Ankyrin-2 is co-localized with L1 cell adhesion molecule (L1CAM) in these premyelinated axons.

Expression analysis via Northern blotting reveals that the major 13-kb transcript encoding 440-kDa Ankyrin-2 is abundant in neonatal mouse brain, along with less abundant 8-kb and 5-kb transcripts . A key characteristic of Ankyrin-2 expression is its downregulation following myelination, suggesting specific roles in early neurodevelopmental processes rather than maintenance of mature myelinated axons .

How does Ankyrin-2 deficiency affect brain development in mouse models?

Ankyrin-2 knockout (Ank2−/−) mice exhibit severe neurological abnormalities, providing crucial insights into its role in brain development. Three-dimensional diffusion-weighted magnetic resonance microscopy (DWM) of fixed brains from 1-day-old Ank2−/− mice revealed:

  • Sevenfold enlargement of lateral ventricles compared to controls

  • Hypoplasia of the corpus callosum and pyramidal tracts

  • Normal total brain and cerebellar volumes

  • Patent aqueduct of Sylvius (unlike some hydrocephalus models)

The neurological impairments intensify as development progresses, with surviving mutant mice (only about 5% survive beyond two weeks) showing abnormal locomotion and balance, weighing approximately 25% less than their littermates . These findings establish Ankyrin-2 as essential for normal brain development and function.

What is the relationship between Ankyrin-2 and L1 cell adhesion molecule in the developing nervous system?

Ankyrin-2 and L1 cell adhesion molecule (L1CAM) exhibit functional coupling in the developing nervous system, particularly in premyelinated axons. Evidence for this interaction includes:

  • Co-localization of 440-kDa Ankyrin-2 and L1CAM in premyelinated axon tracts

  • Concurrent downregulation of both proteins after myelination

  • Similar (though more severe) phenotypes in Ank2−/− mice compared to L1−/− mice

  • Reduced L1CAM levels in premyelinated axons of long fiber tracts in Ank2−/− mice

Particularly striking is the reduction of L1CAM in specific neuroanatomical structures of Ank2−/− mice, including the corpus callosum, fimbria, internal capsule, pyramidal tracts, and lateral columns of the spinal cord . In the optic nerve, L1CAM is initially present at postnatal day 1 but disappears by postnatal day 7 in mutant mice, while other neural cell adhesion molecules like NCAM remain unchanged .

This relationship suggests that Ankyrin-2 stabilizes or maintains L1CAM in developing axons, providing a critical link between the extracellular environment and the neuronal cytoskeleton.

How does Ankyrin deficiency contribute to hemolytic anemia in mouse models?

While Ankyrin-1 (Ankyrin-R) is the predominant ankyrin in erythrocytes, research on the normoblastosis (nb/nb) mouse model provides valuable insights into ankyrin function in erythrocyte membranes that may inform Ankyrin-2 research. The nb/nb mice exhibit severe hemolytic anemia characterized by extreme fragility and shortened lifespan of erythrocytes .

The primary defect in nb/nb mice involves ankyrin deficiency, with consequent spectrin reduction (approximately 50% of normal levels). This deficiency disrupts the erythrocyte membrane skeleton's integrity in several ways:

  • Compromised linkage between spectrin and band 3 transmembrane protein

  • Disruption of the two-dimensional net-like structure of the membrane skeleton

  • Reduced stability of the erythrocyte membrane

Biochemical analyses reveal that nb/nb erythrocytes lack normal 210-kDa ankyrin but contain a 150-kDa ankyrin immunoreactive protein that likely represents a truncated or abnormally processed form of ankyrin . The partial functionality of this 150-kDa protein, together with alternative membrane-stabilizing mechanisms (such as the band 4.1-glycophorin A complex), appears to enable approximately 50% of normal spectrin incorporation into the membrane skeleton .

What methodologies are most effective for studying Ankyrin-2 in erythrocyte membrane dynamics?

To effectively study Ankyrin-2 or related ankyrins in erythrocyte membrane dynamics, researchers should consider a multi-faceted approach combining genetic, biochemical, and immunological techniques:

  • Genetic mapping and linkage analysis: Using interstrain backcross mice and recombinant inbred strains to locate ankyrin genes, as demonstrated with the mapping of nb and Ank-1 loci to mouse chromosome 8 .

  • Biochemical fractionation: Separating membrane components through techniques such as:

    • Preparation of erythrocyte ghosts using hypotonic lysis

    • Spectrin extraction using low ionic strength buffers

    • Isolation of inside-out vesicles (IOVs) to study ankyrin-membrane interactions

  • Immunological detection: Using monospecific antibodies (such as anti-human erythrocyte ankyrin IgG) to detect ankyrin and ankyrin-related proteins via:

    • Western blotting/immunoblot analysis

    • Immunoprecipitation

    • Immunohistochemistry

  • Functional assays:

    • Osmotic fragility testing to assess erythrocyte membrane integrity

    • Spectrin binding assays to quantify ankyrin-spectrin interactions

    • Analysis of ankyrin association with spectrin-depleted IOVs

These methodologies, applied individually or in combination, can provide comprehensive insights into the role of ankyrins in maintaining erythrocyte membrane stability and function.

What are the optimal methods for producing and purifying recombinant partial mouse Ankyrin-2?

For producing and purifying recombinant partial mouse Ankyrin-2, researchers should consider domain-specific expression based on experimental needs. An optimal approach includes:

  • Vector selection and construct design:

    • Select expression systems based on the specific Ankyrin-2 domain of interest

    • For membrane-binding domain (24 ankyrin repeats): pET or pGEX vectors

    • For spectrin-binding domain: mammalian expression systems with physiological post-translational modifications

    • Include affinity tags (His6, GST, or FLAG) for purification while ensuring they don't interfere with protein folding

  • Expression optimization:

    • For prokaryotic systems: Test multiple E. coli strains (BL21(DE3), Rosetta, Arctic Express) at various induction temperatures (16-37°C)

    • For mammalian systems: Consider HEK293 or CHO cells with inducible promoters

    • Co-expression with chaperones may improve folding of complex domains

  • Purification strategy:

    • Initial capture: Affinity chromatography using tag-specific resins

    • Intermediate purification: Ion exchange chromatography

    • Polishing: Size exclusion chromatography to ensure homogeneity

    • Consider including stabilizing agents (reducing agents, specific ions) in buffers based on the domain being purified

  • Quality control assessments:

    • SDS-PAGE for purity evaluation

    • Mass spectrometry for identity confirmation

    • Circular dichroism to assess secondary structure

    • Functional binding assays with known interaction partners (spectrin, L1CAM)

Optimized expression and purification protocols must be tailored to the specific Ankyrin-2 domain being studied, as the three major domains (membrane-binding, spectrin-binding, and regulatory) display distinct biochemical properties and folding requirements.

What experimental approaches are most effective for studying Ankyrin-2 interactions with L1CAM in neuronal tissues?

To effectively study Ankyrin-2 interactions with L1CAM in neuronal tissues, researchers should employ complementary in vitro and in vivo approaches:

  • Co-localization studies in neuronal tissues:

    • Immunohistochemistry of brain sections from different developmental stages

    • Confocal microscopy to visualize spatial relationships between Ankyrin-2 and L1CAM

    • Super-resolution microscopy (STORM, PALM) for nanoscale interaction analysis

  • Biochemical interaction analyses:

    • Co-immunoprecipitation from brain lysates at different developmental stages

    • Pull-down assays using recombinant domains of both proteins

    • Surface plasmon resonance or isothermal titration calorimetry to determine binding kinetics and affinities

    • Cross-linking mass spectrometry to map interaction interfaces

  • Functional studies in cellular models:

    • Primary neuronal cultures from wild-type and Ank2−/− mice

    • Knockdown/knockout approaches using siRNA or CRISPR-Cas9

    • Rescue experiments with wild-type or mutant Ankyrin-2 constructs

    • Live-cell imaging to track dynamics of Ankyrin-2/L1CAM interactions

  • In vivo validation approaches:

    • Conditional knockout models targeting specific neuronal populations

    • In utero electroporation to manipulate Ankyrin-2 expression in developing neurons

    • Analysis of L1CAM trafficking and localization in Ankyrin-2-deficient neurons

Based on research with Ank2−/− mice, these approaches could focus on premyelinated axons in specific brain regions, including the corpus callosum, fimbria, internal capsule, and pyramidal tracts, where significant reductions in L1CAM have been observed in the absence of Ankyrin-2 .

How do the effects of partial versus complete Ankyrin-2 deficiency differ in neuronal development and function?

The differential effects of partial versus complete Ankyrin-2 deficiency represent an important research focus, as they may reveal dosage-dependent functions and potential compensatory mechanisms. Based on available data from Ank2−/− models and other ankyrin research, several key considerations emerge:

  • Genetic approaches to generate partial deficiency models:

    • Heterozygous Ank2+/− mice for studying haploinsufficiency

    • Hypomorphic alleles generated through specific mutations

    • Conditional knockout models with incomplete recombination

    • Domain-specific deletions targeting individual functional modules

  • Developmental timeline analyses:

    • Complete deficiency results in early postnatal lethality (by day 21)

    • Partial deficiency may permit longer survival but with progressive neurological deficits

    • Analysis of specific developmental windows when Ankyrin-2 function is most critical

  • Compensatory mechanisms assessment:

    • Upregulation of other ankyrin family members (Ankyrin-1, Ankyrin-3)

    • Alternative linkages between membrane proteins and cytoskeleton

    • Differential compensation in various neuronal populations

  • Functional outcomes comparison:

    • Electrophysiological properties of neurons with varying Ankyrin-2 levels

    • Axonal transport dynamics and organelle distribution

    • Synaptic development and plasticity

    • Behavioral assessments in viable models

The research from completely deficient Ank2−/− mice shows severe phenotypes including hypoplasia of the corpus callosum and pyramidal tracts, ventricular enlargement, and optic nerve degeneration . Partial deficiency models would allow investigation of threshold effects and potentially reveal subtler functions masked by the severe phenotype and early lethality of complete knockout models.

What are the molecular mechanisms underlying the differential expression of Ankyrin-2 isoforms in various neural cell types?

The molecular mechanisms regulating differential expression of Ankyrin-2 isoforms (440-kDa, 220-kDa, and 150-kDa) across neural cell types represent a sophisticated level of post-transcriptional regulation. Advanced research approaches to elucidate these mechanisms include:

  • Transcriptional regulation analysis:

    • Identification of cell type-specific promoters and enhancers using ChIP-seq

    • ATAC-seq to identify accessible chromatin regions in different neural populations

    • Single-cell RNA-seq to correlate transcription factor expression with Ankyrin-2 isoform patterns

    • Promoter-reporter assays to validate regulatory elements

  • Alternative splicing mechanisms:

    • RNA-seq with long-read technologies to identify full-length isoform transcripts

    • Minigene assays to define exonic and intronic splicing enhancers/silencers

    • CLIP-seq to identify RNA-binding proteins regulating Ankyrin-2 splicing

    • Manipulation of splicing factors to modify isoform ratios

  • Post-transcriptional regulation:

    • Analysis of mRNA stability differences between isoforms

    • Identification of microRNA regulation using CLIP and functional assays

    • Assessment of RNA localization mechanisms for spatial control of translation

    • Polysome profiling to evaluate translational efficiency of different isoforms

  • Developmental switches:

    • Temporal analysis of isoform switching during neurodevelopment

    • Investigation of activity-dependent regulation of isoform expression

    • Epigenetic modifications associated with developmental isoform regulation

Northern blot analysis has demonstrated that the major 13-kb mRNA transcript encoding 440-kDa Ankyrin-2 is nearly completely lost in Ank2−/− mice brains, along with the loss of less abundant 8-kb and 5-kb transcripts encoding 220-kDa and 150-kDa Ankyrin-2 polypeptides . This suggests a complex regulatory landscape controlling the expression of these various isoforms, which may be differentially regulated across neural cell types and developmental stages.

How do the functions of Ankyrin-2 compare with Ankyrin-1 in membrane stability across different cell types?

Ankyrin-1 (Ankyrin-R) and Ankyrin-2 (Ankyrin-B) serve as key membrane-stabilizing proteins but with distinct cell type specializations and molecular mechanisms:

FeatureAnkyrin-1 (Ankyrin-R)Ankyrin-2 (Ankyrin-B)
Primary expressionErythrocytesNervous system (neurons)
Major isoform size210 kDa440 kDa
Key binding partnersBand 3, spectrinL1CAM, spectrin
Deficiency phenotypeHemolytic anemia in nb/nb mice Neurological defects, early lethality in Ank2−/− mice
Membrane localizationUniform distribution in erythrocyte membraneAxonal distribution in premyelinated neurons
Human disease associationHereditary spherocytosisLong QT syndrome, autism spectrum disorders
Developmental regulationStable expression in mature erythrocytesDownregulated after axonal myelination

In erythrocytes, Ankyrin-1 forms a critical bridge between the transmembrane protein band 3 and the spectrin-based membrane skeleton, with deficiency resulting in spectrin reduction to approximately 50% of normal levels . This leads to membrane instability and hemolytic anemia in nb/nb mice.

In contrast, Ankyrin-2 plays a crucial role in neuronal membrane stability by linking L1CAM (and potentially other cell adhesion molecules) to the spectrin cytoskeleton in premyelinated axons. Ank2−/− mice exhibit severe neurological abnormalities including hypoplasia of major axon tracts and progressive axonal degeneration .

Despite these differences, both ankyrins share the fundamental function of stabilizing the plasma membrane through linkage of integral membrane proteins to the spectrin-based cytoskeleton, demonstrating evolutionary conservation of this critical cellular mechanism across different cell types.

What innovative experimental approaches can be used to differentiate the specific roles of Ankyrin family members in complex tissues?

Differentiating the specific roles of Ankyrin family members in complex tissues requires sophisticated experimental approaches that can overcome challenges of functional redundancy and tissue complexity:

  • CRISPR-based combinatorial manipulations:

    • Multiplexed CRISPR-Cas9 to target multiple ankyrin family members simultaneously

    • CRISPR activation/inhibition systems for isoform-specific modulation

    • Base editing or prime editing for precise introduction of disease-relevant mutations

    • Inducible CRISPR systems for temporal control of gene disruption

  • Domain-swapping approaches:

    • Generation of chimeric proteins containing domains from different ankyrin family members

    • Knock-in models expressing domain-swapped ankyrins to assess functional redundancy

    • Structure-guided mutagenesis of specific interaction interfaces

  • Advanced imaging techniques:

    • Expansion microscopy for improved spatial resolution in complex tissues

    • Multiplexed imaging using oligonucleotide-based methods (MERFISH, seqFISH)

    • Live-imaging of fluorescently tagged ankyrins in organotypic cultures

    • Correlative light and electron microscopy to connect molecular localization with ultrastructure

  • Single-cell multi-omics:

    • Single-cell RNA-seq combined with spatial transcriptomics

    • Single-cell proteomics to quantify ankyrin protein levels

    • Cellular indexing of transcriptomes and epitopes (CITE-seq) to correlate protein and RNA

    • Trajectory analysis to map developmental roles of different ankyrins

  • Interactome profiling:

    • BioID or APEX2 proximity labeling with different ankyrin family members

    • Quantitative interaction proteomics across developmental timepoints

    • Comparative interaction mapping in wild-type and mutant tissues

    • Protein correlation profiling to identify novel ankyrin-containing complexes

These innovative approaches would help delineate the specific roles of Ankyrin-2 versus other family members like Ankyrin-1 and Ankyrin-3 (Ankyrin-G), particularly in tissues where multiple ankyrins are co-expressed or in developmental contexts where compensatory mechanisms may obscure primary functions.

How can insights from mouse Ankyrin-2 research inform understanding of human neurological disorders?

Research on mouse Ankyrin-2 provides valuable translational insights for human neurological disorders through several key relationships:

  • Genetic homology and disease associations:

    • Mouse Ank2 shows significant homology to human ANK2

    • Human ANK2 mutations have been linked to:

      • Long QT syndrome type 4 (cardiac arrhythmia with neurological manifestations)

      • Autism spectrum disorders

      • Cognitive disabilities

  • Phenotypic parallels with human disorders:

    • Ventricular enlargement in Ank2−/− mice mimics hydrocephalus phenotypes in humans

    • Corpus callosum hypoplasia observed in Ank2−/− mice parallels similar findings in various human neurodevelopmental disorders

    • The neurodevelopmental abnormalities in Ank2−/− mice resemble (though more severe than) those seen in humans with L1CAM mutations, which cause CRASH syndrome (Corpus callosum hypoplasia, Retardation, Adducted thumbs, Spastic paraplegia, and Hydrocephalus)

  • Mechanistic insights for therapeutic development:

    • The functional coupling between Ankyrin-2 and L1CAM suggests potential therapeutic approaches targeting this interaction

    • Understanding the role of Ankyrin-2 in axonal stability and maintenance offers pathways for developing treatments for axonal degeneration disorders

    • The progressive nature of optic nerve degeneration in Ank2−/− mice (initially normal at postnatal day 1, but dilated and degenerating by day 20) provides a model for studying neurodegenerative processes

  • Biomarker potential:

    • Ankyrin-2 expression patterns or breakdown products could serve as biomarkers for axonal integrity

    • Isoform ratios might indicate specific disease states or progression

The similarities between the Ankyrin-2 deficient mouse phenotype and human neurological disorders highlight the translational value of this research, particularly for conditions involving abnormal brain development, ventricular enlargement, and axonal degeneration.

What are the primary technical challenges in generating and analyzing Ankyrin-2 conditional knockout models?

Generating and analyzing Ankyrin-2 conditional knockout models presents several significant technical challenges, each requiring specific solutions:

  • Gene targeting complexity:

    • Challenge: The Ank2 gene is large with multiple splice variants encoding different isoforms (440-kDa, 220-kDa, and 150-kDa)

    • Solution: Design conditional targeting strategies focusing on exons common to all major isoforms, or target specific exons for isoform-selective inactivation

  • Early lethality circumvention:

    • Challenge: Complete Ank2−/− mice die by postnatal day 21, limiting studies of adult functions

    • Solution: Implement temporal control using inducible Cre-recombinase systems (e.g., tamoxifen-inducible CreERT2) to delete Ankyrin-2 at specific developmental stages

  • Cell type-specific deletion:

    • Challenge: Ankyrin-2 functions in multiple neural cell types

    • Solution: Utilize cell type-specific Cre lines (e.g., Thy1-Cre for neurons, Olig2-Cre for oligodendrocytes) to dissect cell-autonomous versus non-cell-autonomous functions

  • Phenotypic analysis complexity:

    • Challenge: Multiple overlapping phenotypes affecting various organ systems

    • Solution: Develop comprehensive phenotyping pipelines including:

      • Advanced neuroimaging (DWM, tractography)

      • Electrophysiological analyses

      • Behavioral testing batteries

      • Molecular profiling of affected tissues

  • Compensation by other ankyrin family members:

    • Challenge: Functional redundancy may mask phenotypes

    • Solution: Generate compound conditional mutants targeting multiple ankyrin family members, or perform acute depletion to minimize compensatory mechanisms

  • Quantitative assessment of subtle phenotypes:

    • Challenge: Partial deletion may produce subtle effects difficult to quantify

    • Solution: Implement automated image analysis, machine learning approaches, and sensitive functional assays to detect minor alterations

The existing Ank2−/− mouse model demonstrates severe neurological phenotypes including hypoplasia of the corpus callosum and pyramidal tracts, dilated ventricles, and extensive degeneration of the optic nerve . Conditional knockout approaches would allow more refined analysis of these phenotypes by controlling the timing and location of Ankyrin-2 deletion.

What are the most promising future research directions for understanding Ankyrin-2 function in neuronal development and disease?

Future research on Ankyrin-2 should address critical knowledge gaps while leveraging emerging technologies. The most promising directions include:

  • Single-cell resolution analyses:

    • Single-cell transcriptomics to identify cell populations most dependent on Ankyrin-2

    • Spatial transcriptomics to map Ankyrin-2 expression patterns in developing brain regions

    • Single-cell proteomics to quantify Ankyrin-2 isoform distributions across neural populations

  • Structure-function relationships:

    • Cryo-EM studies of Ankyrin-2 complexes with binding partners (L1CAM, spectrin)

    • In silico molecular dynamics simulations to predict effects of disease-associated mutations

    • Domain-specific functional studies using CRISPR-engineered mouse models

  • Developmental regulation mechanisms:

    • Investigation of transcriptional and post-transcriptional regulation of Ankyrin-2 expression

    • Analysis of Ankyrin-2 downregulation mechanisms following axonal myelination

    • Identification of signaling pathways controlling Ankyrin-2 function during neurodevelopment

  • Human disease modeling:

    • Generation of patient-specific iPSC-derived neurons carrying ANK2 mutations

    • Development of humanized mouse models expressing human ANK2 variants

    • Exploration of Ankyrin-2's role in neurodevelopmental and neurodegenerative conditions

  • Therapeutic targeting strategies:

    • Small molecule screening to identify compounds stabilizing Ankyrin-2/L1CAM interactions

    • Antisense oligonucleotide approaches to modulate Ankyrin-2 isoform expression

    • Gene therapy approaches for ANK2-related disorders

The significant neurological abnormalities observed in Ankyrin-2 deficient mice—including ventricular enlargement, corpus callosum hypoplasia, and progressive optic nerve degeneration —highlight the critical importance of Ankyrin-2 in neural development and suggest its potential role in human neurological disorders. Future research should aim to translate these fundamental insights into clinical applications.

How might systems biology approaches enhance understanding of Ankyrin-2's role in coordinating cytoskeletal-membrane interactions?

Systems biology approaches offer powerful frameworks for understanding Ankyrin-2's role within the complex network of cytoskeletal-membrane interactions:

  • Network modeling and analysis:

    • Construction of protein-protein interaction networks centered on Ankyrin-2

    • Identification of network motifs and regulatory hubs in Ankyrin-2-dependent pathways

    • Dynamic modeling of cytoskeletal-membrane interactions under normal and pathological conditions

    • Bayesian network analysis to predict indirect consequences of Ankyrin-2 dysfunction

  • Multi-omics integration:

    • Integration of transcriptomic, proteomic, and phosphoproteomic data from Ankyrin-2 models

    • Temporal profiling across developmental stages to capture dynamic changes

    • Correlation of molecular changes with structural and functional phenotypes

    • Meta-analysis across multiple model systems (cell lines, primary neurons, mouse models)

  • Computational prediction and validation:

    • Machine learning approaches to predict novel Ankyrin-2 functions and interactions

    • Virtual screening for compounds that modulate Ankyrin-2 interactions

    • In silico prediction of the effects of post-translational modifications on Ankyrin-2 function

    • Development of predictive models for disease progression based on Ankyrin-2 status

  • Quantitative imaging and biophysical methods:

    • Super-resolution imaging combined with computational image analysis

    • Fluorescence correlation spectroscopy to measure binding dynamics in living cells

    • Atomic force microscopy to assess membrane-cytoskeleton mechanical properties

    • Optical tweezers or magnetic tweezers to measure forces in Ankyrin-2-mediated linkages

  • In silico modeling of structural dynamics:

    • Molecular dynamics simulations of Ankyrin-2's membrane-binding domain interactions

    • Coarse-grained simulations of large-scale cytoskeletal-membrane organization

    • Multi-scale modeling connecting molecular interactions to cellular properties

These systems biology approaches would provide comprehensive insights into how Ankyrin-2 orchestrates interactions between the plasma membrane and the spectrin-based cytoskeleton in neurons, similar to but distinct from the well-characterized role of Ankyrin-1 in erythrocytes .

What are the key implications of Ankyrin-2 research for broader understanding of cytoskeletal organization in specialized cell types?

Research on Ankyrin-2 has profound implications for understanding fundamental aspects of cytoskeletal organization, particularly in specialized cell types such as neurons:

  • Evolutionary conservation of membrane-cytoskeleton linkage mechanisms:

    • Ankyrin-mediated linkages represent conserved strategies across diverse cell types

    • The specialization of Ankyrin-2 for neuronal functions versus Ankyrin-1 for erythrocytes illustrates how evolution has adapted a common structural mechanism for cell type-specific requirements

  • Developmental regulation of membrane-cytoskeleton interactions:

    • The temporal regulation of Ankyrin-2 during development, particularly its downregulation after myelination, reveals sophisticated control mechanisms governing cytoskeletal organization during cellular maturation

    • This suggests that membrane-cytoskeleton interactions are not static but dynamically regulated during development

  • Cell type-specific adaptor functions:

    • Ankyrin-2's interaction with L1CAM in neurons parallels Ankyrin-1's interaction with Band 3 in erythrocytes, demonstrating how ankyrins serve as adaptable linkers for different membrane proteins in different cellular contexts

    • This adaptability allows for specialized membrane domains with distinct functional properties

  • Hierarchical organization of membrane-associated complexes:

    • Ankyrin-2 deficiency impacts multiple downstream components, including L1CAM localization and stability, illustrating the hierarchical nature of membrane-cytoskeleton organization

    • This hierarchy may represent a common organizational principle across specialized cell types

  • Pathological implications of cytoskeletal disruption:

    • The severe phenotypes in Ankyrin-2 deficient mice highlight the critical importance of proper membrane-cytoskeleton linkages for cellular function and survival

    • Similar principles may apply across diverse cell types and may underlie multiple human diseases

These insights from Ankyrin-2 research contribute to a broader understanding of how cells establish and maintain specialized membrane domains through cytoskeletal interactions, with implications extending beyond neurobiology to fundamental cell biology principles.

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 2025 TheBiotek. All Rights Reserved.