Recombinant Rat Alpha-tubulin N-acetyltransferase (Atat1)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Rat Atat1

Recombinant Rat Alpha-tubulin N-acetyltransferase (Atat1) refers to the artificially produced version of the native rat enzyme through genetic engineering techniques. As the primary enzyme responsible for α-tubulin acetylation in mammals, Atat1 plays crucial roles in regulating microtubule dynamics and function . The recombinant form of rat Atat1 provides researchers with a valuable tool to study tubulin acetylation mechanisms, develop potential therapeutic interventions, and understand its role in various physiological and pathological processes.

Alpha-tubulin acetylation represents a unique post-translational modification as it occurs specifically on lysine 40, which is positioned on the inner luminal surface of microtubules. This distinctive location presents interesting challenges for the enzyme's mechanism of action and access to its substrate . Recombinant rat Atat1 preserves the catalytic capabilities of the native enzyme while offering advantages for laboratory research, including consistent production, purification, and potential for structural and functional modifications.

Catalytic Domain Structure

The catalytic domain of Atat1 exhibits a characteristic fold similar to histone acetyltransferases like Gcn5, comprising a central β-sheet flanked by α-helices. While the specific crystal structure of rat Atat1 has not been fully characterized in the search results, studies on human ATAT1 reveal that the catalytic domain features:

  • A central β-sheet with six antiparallel filaments

  • Three α-helices on each side of the β-sheet

  • A 'hairpin β' structural motif inserted between β3 and β6

The enzyme's activity depends on conserved phenylalanine residues packed into a hydrophobic pocket within the α2 helix, and the N-terminal region forms a coiled-coil structure that helps position the acetyl-CoA substrate correctly .

Substrate Binding Sites

Rat Atat1, like its human counterpart, contains specific binding sites for both acetyl-CoA (the acetyl donor) and α-tubulin (the acetyl acceptor). The acetyl-CoA binding pocket is well-conserved compared to other acetyltransferases, while the α-tubulin binding site is distinctive. Several key residues are involved in substrate recognition and catalysis:

  • D157: Essential for organizing the active site by forming a salt bridge that helps create the α-tubulin binding pocket

  • C120: Required for catalytic activity

  • R158 and I64: Make the α-tubulin binding pocket hydrophobic and position K40 for acetylation

  • R69, H75, and K102: Provide negative charge within the tubulin-binding groove

FeatureKey ResiduesFunction
Acetyl-CoA bindingR132, K162Recognition of adenine base
α-tubulin binding pocketR158, I64, R69, H75, K102Substrate binding and positioning
Catalytic activityD157, C120Essential for enzymatic function
Hydrophobic coreConserved phenylalaninesStructural stability

Catalytic Mechanism

Rat Atat1 catalyzes the transfer of an acetyl group from acetyl-CoA to lysine 40 of α-tubulin through a ternary complex mechanism involving both substrates . The enzyme preferentially acetylates α-tubulin within assembled microtubules rather than free tubulin dimers, suggesting that the microtubule structure itself plays a role in the acetylation process .

The acetylation reaction proceeds through several steps:

  1. Binding of acetyl-CoA to create a binary complex

  2. Recognition and binding of the α-tubulin substrate

  3. Transfer of the acetyl group to K40 of α-tubulin

  4. Release of the acetylated tubulin and CoA

Microtubule Lumen Entry

A fascinating aspect of Atat1's function is its requirement to access the microtubule lumen, as its substrate (K40 of α-tubulin) resides on the inner surface of microtubules. Research has demonstrated that Atat1 can enter the microtubule lumen through:

  1. Open microtubule ends

  2. Breaks or bends in the microtubule lattice

Once inside the lumen, Atat1's movement is relatively inefficient and depends on its affinity for α-tubulin binding sites. This limited mobility means that acetylation accumulates more readily near entry points, and more distant sites are acetylated more slowly . This mechanism helps explain why stable, long-lived microtubules tend to accumulate more acetylation over time compared to dynamic microtubules.

Expression and Localization in Rat Tissues

Rat Atat1 shows a specific expression and localization pattern across various tissues, providing insights into its physiological functions.

Tissue Distribution

Immunohistochemical studies have revealed that Atat1 is widely distributed in rat tissues, with particularly notable expression in:

  • Brain (especially the third ventricle)

  • Trachea

  • Oviduct

  • Kidney (medullary collecting duct)

  • Retina

  • Testis

This distribution pattern suggests important roles for Atat1 in specialized cell types and structures, particularly those containing cilia and microtubule-rich organelles.

Subcellular Localization

Within rat tissues, Atat1 demonstrates specific subcellular localization patterns that correlate with its function:

  • Motile cilia of multiciliated cells in the trachea, brain third ventricle, and oviduct

  • Primary cilia of renal medullary collecting duct cells

  • Inner and outer segments of retinal photoreceptor cells

  • Golgi apparatus of spermatocytes and spermatids in the testis

This distribution pattern highlights Atat1's importance in ciliary function and specialized microtubule structures across different cell types.

TissueSubcellular Localization
TracheaMotile cilia of multiciliated cells
Brain (third ventricle)Motile cilia of multiciliated cells
OviductMotile cilia of multiciliated cells
Kidney (medullary collecting duct)Primary cilia
RetinaPrimary cilia, inner and outer segments of photoreceptor cells
TestisGolgi apparatus of spermatocytes and spermatids

Roles in Development and Specialized Functions

  • Altered neuronal migration during brain development

  • Changes in the structure of the dentate gyrus

  • Reduced sperm motility and male fertility

  • Deficits in touch sensitivity and mechanical pain perception

Given the similarities between mouse and rat Atat1, similar roles might be expected in rats, though specific studies would be needed to confirm this.

Role in Cellular Responses to CNS Injury

Research using primary mouse cortical neurons has shown that exposure to inhibitory substrates such as chondroitin sulfate proteoglycans (CSPGs) and myelin-associated glycoprotein (MAG) causes a reduction in both α-tubulin acetylation and Atat1 protein levels. This reduction occurs primarily in the distal and middle regions of neurites. Restoring Atat1 levels can promote neurite growth in the presence of these inhibitory substrates, suggesting a potential role for Atat1 in neural regeneration after CNS injury .

Transcriptional and Post-transcriptional Regulation

Limited information is available specifically about rat Atat1 regulation, but studies in other mammalian cells have shown that Atat1 levels can be regulated post-transcriptionally. In cortical neurons, exposure to growth-inhibitory substrates like CSPGs and MAG decreases Atat1 levels through increased protein turnover rather than reduced transcription .

Post-translational Modifications

Atat1 itself is subject to post-translational modifications that regulate its activity:

  • Phosphorylation by TGF-β-activated kinase 1 (TAK1) at S237 promotes catalytic activation

  • Phosphorylation by casein kinase 2 (CK2) at S236 is required for Atat1 activation

  • p21-activated kinase 1 (PAK1) phosphorylates multiple residues on Atat1

These modifications provide mechanisms for fine-tuning Atat1 activity in response to cellular signaling pathways.

Research Applications

Recombinant rat Atat1 serves as a valuable tool for studying:

  • Mechanisms of microtubule acetylation

  • Structure-function relationships in acetyltransferases

  • Roles of tubulin acetylation in various cellular processes

  • Development of inhibitors or activators of Atat1 for potential therapeutic applications

Potential Therapeutic Applications

Understanding Atat1 function has implications for developing therapies for:

  1. Neurodegenerative disorders involving microtubule dysfunction

  2. CNS injuries where axonal regeneration is inhibited

  3. Male infertility related to sperm motility defects

  4. Disorders affecting ciliary function

Product Specs

Form
Lyophilized powder

Note: While we will prioritize shipping the format currently in stock, please specify your format preference in order notes if needed. We will accommodate your request to the best of our ability.

Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.

Note: All proteins are shipped with standard blue ice packs unless otherwise requested. Dry ice shipping requires advance notification and incurs additional charges.

Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Before opening, briefly centrifuge the vial 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 can serve as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer components, 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
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.

The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.

Synonyms
Atat1; Mec17Alpha-tubulin N-acetyltransferase 1; Alpha-TAT; Alpha-TAT1; TAT; EC 2.3.1.108; Acetyltransferase mec-17 homolog
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-421
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Rattus norvegicus (Rat)
Target Names
Atat1
Target Protein Sequence
MEFPFDVDAL FPERITVLDQ HLRPPARRPG TTTPARVDLQ QQIMTIVDEL GKASAKAQHL PAPITSALRM QSNRHVIYVL KDTSARPAGK GAIIGFLKVG YKKLFVLDDR EAHNEVEPLC ILDFYIHESV QRHGHGRELF QYMLQKERVE PHQLAIDRPS PKLLKFLNKH YNLETTVPQV NNFVIFEGFF AHQHRSPTPS LRATRHSRAA VVDPIPAAPA RKLPPKRAEG DIKPYSSSDR EFLKVAVEPP WPLNRAPRRA TPPAHPPPRS SSLGNSPDRG PLRPFVPEQE LLRSLRLCPP HPTARLLLAT DPGGSPAQRR RTRETPWGLV AQSCHYSRHG GFNTSFLGTG NQERKQGEQE AEDRSASEDQ VLLQDGSGEE PTHTVAPRAQ APPAQSWMVG GDILNARVIR NLQERRNTRP W
Uniprot No.

Target Background

Function
Recombinant Rat Alpha-tubulin N-acetyltransferase (Atat1) specifically acetylates Lys-40 in alpha-tubulin on the lumenal side of microtubules. This action promotes microtubule destabilization and accelerates microtubule dynamics, potentially independent of its acetylation activity. Its acetylation of alpha-tubulin proceeds at a slow rate due to a suboptimal catalytic site for acetyl transfer. The enzyme enters microtubules via both ends, rapidly diffusing throughout the lumen. Acetylation is primarily observed in long, stable microtubules due to its slow rate, as it lacks sufficient time to act on dynamically unstable microtubules before dissociation. Atat1 is crucial for normal sperm flagellar function and promotes directional cell locomotion and chemotaxis, possibly through AP2A2-dependent acetylation of alpha-tubulin at clathrin-coated pits concentrated at the leading edge of migrating cells. It may also facilitate primary cilium assembly.
Gene References Into Functions
  1. These findings suggest that Atat1-mediated alpha-tubulin acetylation at different subcellular locations may influence the functional regulation of microtubules and cilia in various ciliated cells. PMID: 26700226
Database Links
Protein Families
Acetyltransferase ATAT1 family
Subcellular Location
Cytoplasm. Membrane, clathrin-coated pit. Cell junction, focal adhesion. Cell projection, axon. Cytoplasm, cytoskeleton. Cytoplasm, cytoskeleton, spindle.

Q&A

What is Alpha-tubulin N-acetyltransferase (Atat1) and what is its primary function?

Alpha-tubulin N-acetyltransferase 1 (Atat1) is a Gcn5-related N-acetyltransferase that catalyzes the acetylation of the ε-amino group of lysine 40 (K40) in α-tubulin. This post-translational modification is highly conserved in ciliated organisms and serves as a critical mechanism for rapidly adjusting the functional diversity of microtubules. Atat1 has been identified as the major α-tubulin acetyltransferase in mammals, with its activity particularly enriched in specialized cellular structures such as cilia and neuronal axons where microtubule acetylation is normally abundant . The enzyme plays a significant role in regulating subcellular specialization of specific microtubule subsets, thereby influencing various cellular processes including intracellular transport, cell morphology, and ciliary motility .

In which tissues and cellular structures is Atat1 predominantly expressed?

Atat1 shows diverse tissue distribution with particularly notable expression in structures containing specialized microtubule arrays. Immunohistochemical analysis using specific antibodies against Atat1 has revealed its presence in multiple rat tissues including:

  • Tracheal epithelium (motile cilia)

  • Oviductal epithelium (motile cilia)

  • Kidney tubules (primary cilia)

  • Retinal photoreceptors

  • Testicular tissue (sperm flagella)

  • Ependymal cells lining the third ventricle of the brain

Subcellularly, Atat1 is enriched at the external surface of various motile vesicles, including lysosomes and precursors of synaptic vesicles, which is consistent with its role in microtubule acetylation during axonal transport .

How does Atat1 deletion affect microtubule acetylation in mammals?

Targeted deletion of the Atat1 gene in mice results in a remarkable loss of detectable K40 α-tubulin acetylation across multiple tissues. This phenotype is particularly pronounced in cellular structures where acetylation is normally enriched, such as cilia and axons . The absence of Atat1 leads to:

  • Complete or near-complete abolition of α-tubulin K40 acetylation

  • Increased microtubule stability

  • Impaired sperm motility affecting male fertility

  • Altered axonal transport kinetics in neurons

Despite these effects, Atat1 knockout mice remain viable and develop normally, suggesting possible compensatory mechanisms or that α-tubulin acetylation is not absolutely essential for basic developmental processes .

How does Atat1-mediated tubulin acetylation influence axonal transport mechanisms?

Atat1-mediated α-tubulin acetylation plays a critical role in regulating axonal transport of organelles and vesicles. Loss of Atat1 significantly impairs both anterograde (soma to axon terminal) and retrograde (axon terminal to soma) transport processes. Detailed time-lapse imaging analysis of organotypic brain slices from wild-type versus Atat1 knockout mice has revealed several specific transport deficits:

Transport ParameterWild-TypeAtat1 Knockout% Change
Average velocity (μm/s)1.2-1.50.6-0.8~45% decrease
Instantaneous velocity (μm/s)2.0-2.41.1-1.4~45% decrease
Run length (μm)8-123-5~60% decrease
Pause time (% of total)15-2035-45~125% increase

These transport deficits affect multiple organelle types, including lysosomes and mitochondria, suggesting that Atat1-mediated tubulin acetylation provides a general regulatory mechanism for efficient organelle trafficking along neuronal microtubules . These findings have been further validated through experiments in cultured cortical projection neurons from E14.5 Atat1 knockout mice, as well as in vivo studies in fly larva motoneurons, indicating evolutionary conservation of this mechanism .

What is the relationship between vesicular Atat1 localization and microtubule acetylation?

Recent research has revealed a novel mechanism wherein Atat1 is enriched on the external surface of motile vesicles including lysosomes and synaptic vesicle precursors. This vesicular localization creates a functional interdependence between Atat1 and microtubule-dependent transport:

  • Vesicle-associated Atat1 acetylates microtubules during transport

  • Acetylated microtubules facilitate more efficient vesicular transport

  • Blocking microtubule-dependent transport impairs α-tubulin acetylation

This creates a positive feedback loop that is directionally specific. Experiments using ciliobrevin D (which blocks both retrograde and anterograde transport) significantly reduced microtubule acetylation in both the axon and soma. In contrast, knockdown of Lis1 (which primarily blocks retrograde transport) reduced acetylation in the axon but not in the soma . This suggests differential requirements for anterograde versus retrograde transport in promoting α-tubulin acetylation in different neuronal compartments.

How do post-translational modifications of tubulin interact with Atat1 activity?

While Atat1 specifically catalyzes the acetylation of K40 in α-tubulin, this modification exists within a complex landscape of other tubulin post-translational modifications (PTMs). Current research indicates potential crosstalk between different tubulin PTMs, which may influence Atat1 activity or the functional consequences of K40 acetylation:

Tubulin PTMInteraction with K40 AcetylationResearch Evidence
DetyrosinationPossibly synergistic; both enriched in stable microtubulesCo-occurrence in axons and cilia
PolyglutamylationMay influence Atat1 substrate recognitionAltered patterns in Atat1 KO neurons
PolyglycylationPredominantly in cilia/flagella, potential functional overlapCo-occurrence in sperm flagella

Mass spectrometry analysis of tubulin from Atat1 knockout mice has revealed altered patterns of other PTMs, suggesting potential compensatory mechanisms or regulatory interactions . For researchers investigating Atat1, consideration of the broader PTM landscape is essential for comprehensive understanding of experimental outcomes.

What are the optimal methods for expressing and purifying recombinant rat Atat1?

For researchers seeking to produce recombinant rat Atat1, several expression systems and purification strategies have been validated:

Expression Systems:

  • E. coli-based expression: Using pET-based vectors with an N-terminal His-tag allows for high-yield production. Optimal expression occurs at lower temperatures (16-18°C) following IPTG induction (0.1-0.5 mM) to enhance protein solubility.

  • Mammalian expression systems: For studies requiring post-translational modifications, HEK293 or CHO cells transfected with rat Atat1 cDNA can be utilized, though with lower yields than bacterial systems.

Purification Protocol:

  • Lyse cells in buffer containing 50 mM HEPES (pH 7.5), 300 mM NaCl, 10% glycerol, 5 mM β-mercaptoethanol, and protease inhibitors

  • Perform initial purification using Ni-NTA affinity chromatography

  • Apply ion exchange chromatography (HiTrap Q) for intermediate purification

  • Complete with size exclusion chromatography using Superdex 200

  • Verify purity using SDS-PAGE and Western blotting with anti-Atat1 antibodies

For functional assays, the purified enzyme should be stored in buffer containing 20 mM HEPES (pH 7.5), 150 mM NaCl, 10% glycerol, and 1 mM DTT at -80°C, avoiding repeated freeze-thaw cycles to maintain enzymatic activity.

What are the most reliable assays for measuring Atat1 enzymatic activity?

Several well-validated assays are available for quantifying Atat1 acetyltransferase activity:

In vitro acetylation assay:

  • Incubate purified Atat1 (1-5 μg) with α/β-tubulin dimers or polymerized microtubules (10-20 μg)

  • Reaction buffer: 50 mM HEPES (pH 7.5), 50 mM KCl, 10 mM MgCl2, 1 mM EGTA, 1 mM DTT

  • Add acetyl-CoA (typically 100-500 μM) to initiate reaction

  • Incubate at 37°C for 30-60 minutes

  • Detect acetylation by:

    • Western blotting with anti-acetylated K40 α-tubulin antibody

    • Radiometric assay using [14C]-acetyl-CoA

    • Mass spectrometry for precise quantification

Cellular acetylation assay:

  • Transfect cells with wild-type or mutant Atat1 constructs

  • After 24-48 hours, fix cells with 4% paraformaldehyde

  • Permeabilize and immunostain with anti-acetylated K40 α-tubulin antibody

  • Quantify fluorescence intensity using confocal microscopy

For high-throughput applications, a fluorescence-based assay has been developed using a synthetic peptide containing the K40 region of α-tubulin coupled with a fluorescent detection system for the CoA byproduct of the acetylation reaction.

How can researchers effectively generate and validate Atat1 knockout models?

When developing Atat1 knockout models for research, several approaches have proven successful:

Gene Targeting Strategies:

  • Conventional knockout: Complete deletion of critical exons (typically exons 2-3) of the Atat1 gene using homologous recombination

  • Conditional knockout: Insertion of loxP sites flanking critical exons, allowing tissue-specific deletion when crossed with appropriate Cre-expressing lines

  • CRISPR/Cas9-mediated deletion: Design of guide RNAs targeting early exons of Atat1 for efficient gene disruption

Validation Methods:

  • Genotyping: PCR-based screening with primers flanking the targeted region

  • mRNA expression: RT-qPCR using primers targeting multiple regions of the Atat1 transcript

  • Protein expression: Western blotting of tissue lysates using validated anti-Atat1 antibodies

  • Functional validation: Assessment of α-tubulin K40 acetylation levels using immunoblotting or immunohistochemistry

  • Phenotypic analysis: Evaluation of known Atat1-dependent processes such as sperm motility or axonal transport

For in vitro studies, RNA interference approaches using shRNA against Atat1 have been effectively employed. The most efficient knockdown has been achieved using shRNA constructs targeting the coding sequence within exons 4-6, with validation through both mRNA quantification and Western blotting .

Why might α-tubulin acetylation levels remain detectable after Atat1 knockout?

When researchers observe residual α-tubulin acetylation despite confirmed Atat1 knockout, several possibilities should be considered:

  • Alternative acetyltransferases: While Atat1 is the primary α-tubulin acetyltransferase, other enzymes with lower specificity might contribute minimally to K40 acetylation. Current research suggests this contribution is negligible in most tissues, but tissue-specific compensation cannot be ruled out.

  • Antibody cross-reactivity: Some commercial anti-acetylated K40 α-tubulin antibodies may cross-react with other acetylated proteins or different tubulin PTMs. Validation with multiple antibody clones is recommended.

  • Maternal contribution: In early embryonic studies, maternal Atat1 protein or mRNA might persist, maintaining some acetylation until completely degraded.

  • Incomplete knockout: Verify whether the knockout strategy might allow for expression of truncated but partially functional Atat1 variants.

  • Sample contamination: Ensure proper controls are in place to rule out sample mixing or contamination during experimental procedures.

For definitive assessment, mass spectrometry analysis of α-tubulin from Atat1 knockout tissues provides the most accurate quantification of K40 acetylation levels .

How can researchers address variability in axonal transport assays when studying Atat1 function?

Axonal transport assays often show considerable variability, which can be particularly challenging when assessing subtle phenotypes in Atat1 mutant models. To minimize variability and enhance reproducibility:

  • Standardize neuronal culture conditions:

    • Consistent plating density (typically 50,000-100,000 neurons per cm²)

    • Defined culture media composition

    • Precise timing of experiments (DIV 3-5 for early assessment, DIV 7-10 for mature neurons)

  • Optimize imaging parameters:

    • Maintain consistent temperature (37°C) during live imaging

    • Use environmental chambers with controlled CO₂ levels

    • Standardize laser power and exposure settings

    • Acquire images at consistent intervals (typically 0.5-1 second)

  • Implement robust analysis methods:

    • Use automated tracking software (e.g., TrackMate, KymoAnalyzer)

    • Apply consistent filtering criteria for vesicle inclusion

    • Analyze sufficient numbers of vesicles (>100 per condition)

    • Include multiple technical and biological replicates

  • Control for confounding factors:

    • Assess cell viability before imaging

    • Monitor potential phototoxicity

    • Control for cell passage number in cultured cell lines

    • Include appropriate wild-type littermate controls

Microfluidic chamber systems have proven particularly valuable for studying axonal transport, as they allow physical separation of neuronal cell bodies from axons, enabling isolated manipulation and imaging of axonal compartments .

What technical challenges might arise when generating recombinant Atat1 protein and how can they be addressed?

Researchers often encounter specific challenges when expressing and purifying recombinant Atat1:

ChallengeSolution Approach
Poor protein solubility- Lower induction temperature (16°C)
- Reduce IPTG concentration (0.1 mM)
- Include solubility enhancers (10% glycerol, 0.1% Triton X-100) in lysis buffer
Low enzymatic activity- Verify protein folding using circular dichroism
- Include cofactors (acetyl-CoA) during purification
- Assess potential inhibitory contaminants
- Optimize buffer conditions (test pH range 7.0-8.0)
Protein aggregation- Include reducing agents (5 mM DTT) in all buffers
- Perform size exclusion chromatography
- Centrifuge protein solution before use (100,000g, 30 min)
Proteolytic degradation- Include multiple protease inhibitors
- Minimize handling time during purification
- Keep samples consistently cold (4°C)
- Consider fusion partners (MBP) for enhanced stability

For activity assays specifically, pre-incubating recombinant Atat1 with acetyl-CoA before adding the tubulin substrate has been shown to enhance enzymatic activity, potentially by stabilizing the active conformation of the enzyme.

How does vesicular localization of Atat1 contribute to spatial regulation of microtubule acetylation?

Recent research has revealed that Atat1 is not uniformly distributed throughout the cytoplasm but is enriched on the external surface of specific vesicle populations. This localization creates a model wherein Atat1-enriched vesicles serve as "mobile acetyltransferase platforms" that acetylate microtubules as they travel along them. This mechanism explains several previously puzzling observations:

  • The preferential acetylation of long-lived, stable microtubules

  • The gradient of acetylation often observed along neuronal processes

  • The interdependence between transport and acetylation

Time-lapse imaging combined with super-resolution microscopy has demonstrated that vesicles positive for Atat1 generate "acetylation trails" along microtubule tracks as they move . Interestingly, blocking microtubule-dependent transport with drugs like ciliobrevin D or genetic manipulation of transport machinery components (e.g., Lis1) significantly reduces α-tubulin acetylation levels .

This vesicular localization mechanism presents exciting opportunities for future research, particularly in understanding how Atat1 is recruited to specific vesicle subpopulations and how this recruitment is regulated in different physiological and pathological contexts.

What is the potential of Atat1 as a therapeutic target in neurodegenerative diseases?

The critical role of Atat1 in axonal transport and microtubule dynamics positions it as a potential therapeutic target in neurodegenerative diseases where transport deficits are implicated:

  • Alzheimer's disease: Impaired axonal transport is an early event in pathogenesis, often preceding amyloid plaque formation. Enhancing Atat1 activity could potentially restore transport efficiency.

  • Parkinson's disease: α-Synuclein aggregates disrupt microtubule acetylation and axonal transport. Modulating Atat1 function might counteract these effects.

  • Amyotrophic Lateral Sclerosis (ALS): Transport deficits contribute to motor neuron degeneration in ALS. Atat1-targeted interventions could help maintain axonal homeostasis.

Potential therapeutic approaches include:

  • Small molecule activators of Atat1 enzymatic activity

  • Gene therapy approaches to increase Atat1 expression in affected neurons

  • Targeting regulators of Atat1 localization to enhance its association with transport vesicles

How do developmental and tissue-specific factors influence Atat1 expression and function?

Understanding the developmental regulation and tissue-specific functions of Atat1 represents an important frontier in current research:

Developmental regulation:

  • Atat1 expression patterns change during development, with particularly high expression in developing neuronal tissues

  • The enzyme appears to play important roles during neuronal migration and axon outgrowth

  • Compensatory mechanisms may exist during development that become less effective in adult tissues

Tissue-specific functions:

  • In sperm flagella, Atat1-mediated acetylation contributes to motility and male fertility

  • In neurons, it regulates axonal transport and potentially synaptic function

  • In cilia, it influences both structure and motility of these specialized organelles

Emerging research using conditional knockout approaches is beginning to elucidate these tissue-specific functions. For example, neuron-specific deletion of Atat1 results in transport deficits without affecting fertility, while testis-specific deletion impacts sperm function without neurological consequences.

Future research employing single-cell transcriptomics and proteomics approaches will likely provide deeper insights into the cell type-specific regulation and functions of Atat1 across different tissues and developmental stages.

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.