ADK Mouse

Adenosine Kinase Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to ADK Mouse

ADK (Adenosine Kinase) Mouse refers to genetically modified mouse models used to study the molecular and physiological roles of adenosine kinase, an enzyme critical for adenosine metabolism. ADK catalyzes the phosphorylation of adenosine to AMP, regulating intracellular/extracellular adenosine levels and influencing methylation via S-adenosylhomocysteine (SAH) dynamics . These mice are engineered to overexpress, delete, or modify ADK expression in specific tissues, enabling researchers to investigate its role in neurological disorders, cardiovascular diseases, sleep regulation, and metabolic syndromes .

Genetic Models and Construction Methods

ADK Mouse models are generated using Cre-LoxP systems, viral vectors, or transgenic approaches. Key models include:

Brain-Specific ADK Knockout (Adk Δbrain)

  • Construction: Crossbreeding Adk fl/fl mice with Nestin-Cre mice .

  • Phenotype: Progressive seizures, cognitive deficits, altered synaptic plasticity, and elevated adenosine A2A receptor (A2AR) activity .

Myeloid-Specific ADK Knockout (ADK MAC-KO)

  • Construction: ADK F/F mice crossed with LysM-Cre mice .

  • Phenotype: Reduced atherosclerotic plaques, increased ABCG1-mediated cholesterol efflux, and improved plaque stability .

ADK Overexpression (Adk-tg)

  • Construction: Transgenic overexpression of cytoplasmic ADK isoform .

  • Phenotype: Reduced EEG delta power, fragmented sleep, and resistance to sleep deprivation .

Neurological Implications

  • Seizures and Cognitive Deficits: Adk Δbrain mice exhibit spontaneous convulsive seizures and impaired hippocampus-dependent memory due to elevated synaptic adenosine and A2AR-TrkB signaling .

  • Epileptogenesis: Overexpression of ADK in hippocampal neurons exacerbates kainate-induced seizures, while ADK inhibitors suppress seizure activity .

Cardiovascular Research

  • Atherosclerosis: ADK MAC-KO mice show 40% smaller aortic lesions and increased ABCG1 expression, reducing foam cell formation .

  • Cardioprotection: Cardiomyocyte-specific ADK deletion improves recovery after ischemia-reperfusion injury via adenosine-mediated pathways .

Sleep Regulation

  • Sleep Architecture: Adk-tg mice sleep 60 minutes less daily, with reduced REM sleep and consolidated NREM sleep episodes .

  • EEG Alterations: Reduced theta (6.25–11 Hz) and delta (0.75–4 Hz) power during waking and sleep states .

Metabolic Disorders

  • Hepatic Dysfunction: Global ADK knockout causes neonatal hepatic steatosis and elevated SAH levels, disrupting methylation .

  • Insulin Signaling: ADK inhibition promotes β-cell replication, suggesting therapeutic potential for diabetes .

Molecular Mechanisms and Pathways

MechanismKey FindingsReferences
Adenosine RegulationADK deficiency increases extracellular adenosine, activating A1R (neuroprotective) and A2AR (synaptic plasticity) .
Epigenetic ModulationADK deletion in myeloid cells reduces DNA methylation of ABCG1, enhancing cholesterol efflux .
Synaptic PlasticityEnhanced theta-burst LTP in Adk Δbrain mice via A2AR-TrkB signaling .

Therapeutic Insights

  • Epilepsy: A2AR antagonists restore normal learning and reduce seizures in Adk Δbrain mice .

  • Atherosclerosis: ADK inhibitors reduce plaque formation by 35% in preclinical models .

  • Ischemic Preconditioning: AAV-mediated ADK knockdown induces ischemic tolerance in stroke models .

Table 1: Phenotypic Comparison of ADK Mouse Models

ModelGenotypeKey PhenotypeTherapeutic Target
Adk ΔbrainNestin-Cre:Adk fl/flSeizures, cognitive deficitsA2AR antagonists
ADK MAC-KOLysM-Cre:ADK F/FReduced atherosclerosisADK inhibitors
Adk-tgTransgenic ADK overexpressionSleep fragmentation, EEG abnormalitiesAdenosine agonists

Implications for Human Disease

ADK Mouse studies have direct relevance to:

  • OMIM #614300: Human ADK deficiency causes hepatic encephalopathy and seizures .

  • Atherosclerosis: Myeloid ADK inhibition may stabilize plaques in high-risk patients .

  • Epilepsy: A2AR modulation could mitigate drug-resistant seizures .

Product Specs

Introduction

Adenosine Kinase is a key enzyme found in high concentrations within mammalian tissues. It plays a critical role in regulating both intra- and extracellular levels of adenosine and adenine nucleotides by catalyzing the transfer of a phosphate group from ATP to adenosine. Due to adenosine's significant impact on various systems like the cardiovascular, nervous, respiratory, and immune systems, inhibitors of Adenosine Kinase are of significant pharmacological interest. These inhibitors increase intravascular adenosine concentrations and demonstrate anti-inflammatory properties.

Description

This ADK protein is produced in E. coli and is a single, non-glycosylated polypeptide chain. It consists of 384 amino acids (with the first 361 amino acids being the active protein) and has a molecular weight of 42.5kDa. A 23 amino acid His-tag is fused to the N-terminus to facilitate purification, which is carried out using proprietary chromatographic methods.

Physical Appearance
Clear solution, sterile-filtered.
Formulation

The ADK protein is supplied in a solution at a concentration of 1mg/ml. The solution contains the following components: 20% glycerol, 20mM Tris-HCl buffer (pH 8.0), 1mM EDTA, and 50mM NaCl.

Stability
For short-term storage (up to 4 weeks), the protein solution can be stored at 4°C. For extended storage, it is recommended to store the solution frozen at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein such as HSA or BSA (at a concentration of 0.1%) is advisable. Repeated freezing and thawing of the protein solution should be minimized.
Purity
The purity of this ADK protein is greater than 95% as determined by SDS-PAGE analysis.
Biological Activity

The specific activity of this ADK protein is greater than 100 pmol/min/μg. Specific activity is defined as the amount of enzyme required to convert 1.0 picomole of adenosine to AMP per minute at a pH of 7.5 and a temperature of 37°C in a coupled enzyme assay system utilizing pyruvate kinase (PK) and lactate dehydrogenase (LDH).

Synonyms

AK, ADK, Adenosine Kinase, Adenosine 5-Phosphotransferase.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMAAADEP KPKKLKVEAP QALSENVLFG MGNPLLDISA VVDKDFLDKY SLKPNDQILA EDKHKELFDE LVKKFKVEYH AGGSTQNSMK VAQWLIQEPH KAATFFGCIG IDKFGEILKR KAADAHVDAH YYEQNEQPTG TCAACITGGN RSLVANLAAA NCYKKEKHLD LERNWVLVEK ARVYYIAGFF LTVSPESVLK VARYAAENNR VFTLNLSAPF ISQFFKEALM DVMPYVDILF GNETEAATFA REQGFETKDI KEIAKKAQAL PKVNSKRQRT VIFTQGRDDT IVAAENDVTA FPVLDQNQEE IIDTNGAGDA FVGGFLSQLV SDKPLTECIR AGHYAASVII RRTGCTFPEK PDFH.

Q&A

What is an ADK Mouse model?

ADK mouse models are genetically engineered laboratory mice with modified expression of adenosine kinase (ADK), the primary enzyme responsible for metabolizing adenosine in the brain. These models are specifically designed to study how altered ADK expression affects brain function, particularly in pathological conditions. The most common modifications include knockout models (where ADK expression is eliminated), knockdown models (where ADK expression is reduced), and overexpression models (where ADK levels are elevated beyond normal levels) . These models are particularly valuable because ADK serves as the major negative metabolic regulator of adenosine, an endogenous neuroprotectant and homeostatic bioenergetic network regulator in the brain .

What types of ADK transgenic mouse models are available?

Several specialized ADK mouse models have been developed for research purposes:

  • fb-Adk-def mice: These mice have a forebrain-selective reduction of ADK in cortical and hippocampal regions (approximately 65% of normal levels) while maintaining overexpression in the striatum (approximately 164% of normal levels) .

  • Adk-tg mice: These transgenic mice exhibit brain-wide overexpression of ADK (approximately 147% of wild-type levels in forebrain), resulting in increased adenosine metabolism and clearance .

  • AAV-mediated ADK-modification models: Using adeno-associated virus vectors expressing either ADK sense (ADK-SS) or antisense (ADK-AS) constructs, researchers can achieve localized overexpression or knockdown of ADK expression in specific brain regions .

Each model serves distinct research purposes, allowing for the investigation of region-specific effects of ADK expression on brain function and pathology.

How does adenosine kinase affect brain function and pathology?

ADK functions as the primary regulator of extracellular adenosine concentrations in the brain. By phosphorylating adenosine to adenosine monophosphate (AMP), ADK reduces the availability of adenosine for receptor activation. This regulatory function has significant implications for brain pathophysiology:

  • Neuroprotection: Adenosine levels rapidly increase in injured brain tissue (such as during ischemic stroke), serving as an acute neuroprotectant. Under normal conditions, ADK expression decreases following injury onset, potentiating this protective adenosine surge .

  • Seizure susceptibility: Transgenic mice overexpressing ADK (Adk-tg) exhibit spontaneous seizures (4.8 ± 1.5 seizures per hour), demonstrating that elevated ADK levels increase seizure susceptibility .

  • Stroke injury: ADK expression levels directly correlate with stroke injury severity. Reduced ADK expression provides substantial neuroprotection against ischemic injury, while elevated ADK increases susceptibility to damage .

  • Epileptogenesis: Upregulation of ADK during epileptogenesis appears causative for the development of spontaneous seizures, making ADK a critical link between astrogliosis and neuronal dysfunction in epilepsy .

How should I design experiments to study stroke in ADK mouse models?

When designing experiments to study stroke using ADK mouse models, consider the following methodological approaches:

  • Model selection: Choose the appropriate ADK model based on your research question:

    • fb-Adk-def mice for studying region-specific effects (cortical protection with striatal vulnerability)

    • Adk-tg mice for assessing effects of increased ADK expression

    • AAV-mediated modifications for localized ADK manipulation

  • Stroke induction protocol: The middle cerebral artery occlusion (MCAO) model has been validated for use with ADK transgenic mice. Typically, a 60-minute occlusion is sufficient to produce measurable infarcts while allowing assessment of neuroprotective effects .

  • Assessment timepoints: Plan assessments at both acute (24-48 hours) and extended timepoints (7-28 days) to capture both immediate injury and delayed effects of ADK manipulation.

  • Outcome measures: Include multiple outcome measures:

    • Histological analysis for infarct volume quantification

    • TUNEL staining for cell death assessment

    • Behavioral testing for functional outcomes

    • Molecular analysis (Western blotting) for ADK expression verification

  • Controls: Always include wild-type littermates as controls since background strain can influence stroke outcomes .

What methods are recommended to verify ADK expression in transgenic models?

Proper verification of ADK expression is critical for experimental validity. The following methods are recommended:

  • Immunohistochemistry: Use polyclonal rabbit antiserum against ADK (1:5,000 dilution) to visualize the distribution pattern of ADK expression. This technique allows assessment of regional differences and cellular localization (nuclear versus cytoplasmic) .

  • Western blot analysis: For quantitative assessment of ADK protein levels:

    • Prepare aqueous protein extractions from specific brain regions

    • Standardize to 40 μg protein per lane

    • Use polyclonal rabbit antiserum against ADK (1:5,000)

    • Normalize to α-tubulin (1:5,000) as a loading control

    • Quantify using densitometric analysis

  • Regional analysis: Always compare expression levels between brain regions (e.g., dorsal telencephalon versus brainstem/cerebellum) as transgenic manipulations may have region-specific effects .

  • Cellular specificity: Consider dual-labeling with cell-type-specific markers to determine whether altered ADK expression occurs in neurons, astrocytes, or both cell types .

Mouse ModelForebrain ADK LevelBrainstem/Cerebellum ADK Level
Wild-type100% (baseline)100% (baseline)
fb-Adk-def62% ± 27%141% ± 50%
Adk-tg147% ± 49%141% ± 30%

What controls are essential when working with ADK transgenic mice?

Proper experimental controls are critical for valid interpretation of results with ADK transgenic mice:

  • Genotype controls:

    • Wild-type littermates should always be included as primary controls

    • Heterozygous animals may provide insights into gene-dosage effects

  • Pharmacological controls:

    • Adenosine receptor antagonists (e.g., DPCPX at 1 mg/kg) should be used to confirm whether observed effects are mediated by adenosine receptor activation

    • In fb-Adk-def mice, DPCPX administration reverses neuroprotection, confirming the role of adenosine in the observed effects

  • Regional controls:

    • When studying focal effects, include analysis of both ipsilateral and contralateral brain regions

    • For fb-Adk-def mice, compare cortical (ADK-deficient) versus striatal (ADK-overexpressing) regions

  • Viral vector controls:

    • When using AAV-mediated ADK modification, include control vectors (AAV-null or AAV-GFP)

    • Co-injection of AAV-GFP with ADK constructs allows verification of viral transduction efficiency

  • Temporal controls:

    • For AAV-mediated manipulations, allow 4 weeks after injection before experimental manipulation to ensure stable transgene expression

How should I interpret conflicting data between different ADK mouse models?

When encountering seemingly conflicting results between different ADK mouse models, consider these methodological approaches:

  • Regional expression differences: The fb-Adk-def mouse shows opposite effects in different brain regions—protected cortex but increased injury in striatum—directly correlating with regional ADK expression levels. Always map findings to the specific expression pattern in each model .

  • Developmental compensation: Constitutive transgenic models may develop compensatory mechanisms that acute manipulations (like AAV-mediated approaches) do not. Compare results between constitutive and acute manipulation models to identify potential compensatory effects .

  • Cellular specificity: ADK expression in neurons versus astrocytes may have different functional consequences. The Adk-tg model shows novel neuronal expression of ADK, particularly in pyramidal neurons of the CA3 region, which may explain its severe seizure phenotype .

  • Receptor-specific effects: Use receptor antagonists (like DPCPX for A1 receptors) to determine which adenosine receptor subtypes mediate the observed effects. This approach helped confirm that neuroprotection in fb-Adk-def mice is mediated specifically through adenosine A1 receptors .

  • Quantitative relationship: Assess whether the relationship between ADK expression and the phenotype is linear or threshold-based. Research suggests that the relationship between ADK levels and injury is direct and quantitative—infarct volumes directly correlate with ADK expression levels .

What explains the regional differences in injury susceptibility in fb-Adk-def mice?

The fb-Adk-def mouse model exhibits a fascinating region-specific response to cerebral ischemia that provides important insights into ADK's role in brain injury:

  • Expression pattern correlation: These mice have reduced ADK in cortical forebrain (65% of normal levels) but increased ADK in striatum (164% of normal levels). This expression pattern directly correlates with injury outcomes: almost complete protection in cortex (27% of wild-type injury) but increased damage in striatum (126% of wild-type injury) .

  • Mechanistic explanation: This regional difference confirms that ADK levels directly determine tissue susceptibility to ischemic damage. Higher ADK levels accelerate adenosine metabolism, reducing the availability of this endogenous neuroprotectant during injury .

  • Astrocyte-neuron interplay: In wild-type mice, ADK is predominantly expressed in astrocytes, which regulate extracellular adenosine levels. In transgenic models, altered ADK expression affects the astrocyte-neuron adenosine signaling pathway differently across brain regions .

  • Methodological implications: When analyzing data from these mice, researchers should separately quantify injury in cortical and striatal regions rather than measuring total hemispheric damage, which might obscure these region-specific effects .

  • Translational significance: These regional differences suggest that targeted ADK inhibition in specific brain regions might be more effective than global ADK inhibition for neuroprotection .

How should I analyze EEG data from ADK transgenic mice?

EEG analysis in ADK transgenic mice requires special considerations due to their unique seizure phenotypes:

  • Seizure classification: Categorize seizure activity into standardized types (I-IV) based on amplitude and frequency characteristics:

    • Type I: Low amplitude, low frequency

    • Type II: High amplitude, low frequency

    • Type III: High amplitude, high frequency

    • Type IV: High amplitude with polyspikes

  • Quantitative parameters:

    • Seizure frequency (e.g., 4.8 ± 1.5 seizures per hour in Adk-tg mice)

    • Seizure duration (e.g., 26.7 ± 13.2 seconds per seizure in Adk-tg mice)

    • Time spent in each seizure type

    • Post-ictal depression periods

  • Electrode placement:

    • For Adk-tg mice, bilateral electrodes in the CA3 region are recommended due to prominent ADK overexpression in this region

    • For fb-Adk-def mice, cortical electrodes provide more relevant data on forebrain activity

  • Spontaneous versus induced seizures:

    • Adk-tg mice exhibit spontaneous seizures that should be recorded during extended monitoring periods (≥8 hours)

    • For induced seizures (e.g., post-KA injection), focus analysis on the development of type IV seizure activity, which is absent in fb-Adk-def mice but exacerbated in Adk-tg mice

  • Pharmacological validation:

    • Compare seizure patterns before and after administration of adenosine receptor antagonists (e.g., DPCPX) to confirm adenosine-dependent mechanisms

    • DPCPX administration to fb-Adk-def mice induces type IV seizures similar to those seen in wild-type mice

How can ADK mouse models be used to study epileptogenesis?

ADK mouse models offer unique insights into epileptogenesis mechanisms and potential therapeutic approaches:

  • Model selection for epilepsy research:

    • Adk-tg mice exhibit spontaneous seizures (4.8 seizures/hour) and can be used to study constitutive epilepsy

    • fb-Adk-def mice are resistant to seizure induction and can be used to study neuroprotective mechanisms

    • Wild-type mice subjected to injury develop astrogliosis with ADK upregulation, modeling acquired epilepsy

  • Experimental approaches:

    • Intraamygdaloid kainic acid (KA) injection serves as a well-established protocol to induce status epilepticus and subsequent epileptogenesis

    • Seizure activity should be monitored using cortical and hippocampal EEG recordings

    • Lorazepam administration (30 minutes post-KA) standardizes the duration of status epilepticus

  • Assessment of epileptogenesis:

    • Document the development of spontaneous recurrent seizures through continuous video-EEG monitoring

    • Analyze astrogliosis and regional ADK upregulation through immunohistochemistry

    • Quantify neuronal cell loss through TUNEL staining and cresyl violet histology

  • Mechanistic insights:

    • Adk-tg mice demonstrate that ADK overexpression is sufficient to cause spontaneous seizures

    • fb-Adk-def mice show that reduced forebrain ADK provides protection against seizure development through adenosine A1 receptor activation (confirmed by DPCPX reversal)

    • These findings support the ADK hypothesis of epileptogenesis, which posits that ADK upregulation during astrogliosis is causative for epilepsy development

What is the relationship between astrogliosis, ADK upregulation, and seizure development?

The interaction between astrogliosis, ADK upregulation, and seizure development represents a critical pathological mechanism:

  • Sequential relationship:

    • Brain injury (such as status epilepticus) triggers reactive astrogliosis

    • Reactive astrocytes exhibit upregulation of ADK expression

    • Increased ADK reduces extracellular adenosine levels

    • Reduced adenosine signaling leads to increased neuronal excitability and spontaneous seizures

  • Evidence from transgenic models:

    • Adk-tg mice with brain-wide ADK overexpression develop spontaneous seizures similar to those seen in epileptic wild-type mice

    • The seizure frequency in Adk-tg mice (4.8 ± 1.5 seizures per hour) closely resembles that of wild-type mice following epileptogenic injury (4.3 ± 1.5 seizures per hour)

  • Causative relationship:

    • Transgenic studies demonstrate that ADK upregulation is not merely correlative but causative for seizure development

    • ADK overexpression precedes seizure development in transgenic models

    • Experimental reduction of ADK expression prevents seizure development even after injury

  • Clinical implications:

    • ADK could serve as a biomarker for epileptogenesis

    • Therapeutic strategies targeting ADK or boosting adenosine signaling might prevent epileptogenesis after brain injury

    • ADK inhibitors may represent a novel class of antiepileptogenic drugs

How can ADK mouse models inform gene therapy approaches for neurological disorders?

ADK mouse models provide critical insights for developing gene therapy approaches targeting neurological disorders:

  • Proof-of-concept for ADK-based therapy:

    • AAV-mediated knockdown of ADK (using ADK-AS vectors) reduces infarct volume by 51% in wild-type mice subjected to stroke

    • Conversely, ADK overexpression (using ADK-SS vectors) increases infarct volume by 126%

    • These findings validate ADK as a viable therapeutic target for stroke neuroprotection

  • Delivery strategies:

    • AAV serotype 8 vectors with the gfaABC1D promoter achieve selective expression in astrocytes

    • Stereotactic injection allows targeted delivery to specific brain regions

    • Gene therapy effects are stable for at least 4 weeks post-injection

  • Therapeutic applications:

    • For stroke protection: ADK knockdown in regions at risk for ischemia

    • For epilepsy prevention: ADK knockdown following traumatic brain injury or status epilepticus

    • For chronic epilepsy: ADK knockdown in epileptogenic zones

  • Methodological considerations:

    • Allow 4 weeks after virus injection before experimental manipulations

    • Co-inject AAV-GFP to verify transduction efficiency

    • Confirm successful ADK manipulation through immunohistochemistry and Western blotting

    • Use pharmacological validation (e.g., adenosine receptor antagonists) to confirm mechanism

  • Limitations and considerations:

    • Global ADK inhibition may have undesired side effects in non-CNS tissues

    • Region-specific manipulations may be necessary for therapeutic efficacy

    • Combining gene therapy with pharmacological approaches might provide synergistic benefits

What are common challenges when working with ADK transgenic mice?

Researchers should be aware of several technical challenges specific to ADK transgenic models:

  • Mortality concerns:

    • Adk-tg mice subjected to KA-induced seizures have a 40% mortality rate within 24 hours, requiring careful monitoring and potentially adjusted dosing protocols

    • Global ADK knockout is lethal, necessitating tissue-specific approaches

  • Phenotypic variation:

    • ADK expression in fb-Adk-def mice shows variability (62% ± 27% of wild-type levels), which may contribute to experimental variation

    • Consider quantifying ADK levels in each experimental animal rather than relying on genotyping alone

  • Region-specific effects:

    • The opposite effects in striatum versus cortex in fb-Adk-def mice require region-specific analysis approaches

    • Whole-brain or hemisphere-level analyses may mask important regional differences

  • Cell-type specificity:

    • In fb-Adk-def mice, Cre expression (and thus ADK reduction) does not occur in GABAergic interneurons, creating cellular heterogeneity within brain regions

    • Consider cell-type specific analyses when interpreting results

  • Behavioral confounds:

    • Spontaneous seizures in Adk-tg mice may confound behavioral testing

    • EEG monitoring concurrent with behavioral testing may be necessary to identify and exclude seizure episodes from analysis

How should I design experiments to study the role of ADK in neuroprotection?

When designing experiments to investigate ADK's role in neuroprotection, consider these methodological approaches:

  • Experimental models:

    • MCAO stroke model: 60-minute occlusion followed by reperfusion provides a standardized injury model

    • KA-induced excitotoxicity: Intraamygdaloid KA injection provides a focal excitotoxic injury

    • Combined models: Preconditioning paradigms (e.g., LPS followed by MCAO) to study tolerance mechanisms

  • Comprehensive outcome measures:

    • Histological assessment: Infarct volume quantification, TUNEL staining for cell death

    • Functional testing: Neurological deficit scores, sensorimotor testing

    • Molecular analysis: Inflammatory markers, adenosine receptor expression

    • Electrophysiological measures: EEG recordings, field potentials

  • Pharmacological validation:

    • Use adenosine receptor antagonists (e.g., DPCPX for A1R) to confirm adenosine-dependent mechanisms

    • Include adenosine receptor agonists as positive controls for neuroprotection

    • Consider combining ADK inhibitors with receptor-specific compounds to dissect mechanisms

  • Temporal considerations:

    • Acute phase (24-48 hours): Focus on cell death markers, infarct volume

    • Subacute phase (3-7 days): Assess inflammatory responses, initial recovery

    • Chronic phase (14-28 days): Evaluate functional recovery, long-term tissue remodeling

  • Translational considerations:

    • Include both young and aged animals to assess age-dependence of neuroprotection

    • Consider both male and female animals to identify sex-specific effects

    • Test multiple injury severities to determine the therapeutic window for ADK manipulation

What emerging applications of ADK mouse models show promise?

Several emerging applications of ADK mouse models show significant promise for advancing neurological research:

  • Combination therapies:

    • Exploring synergistic effects of ADK inhibition with other neuroprotective approaches

    • Combining ADK inhibition with anti-inflammatory therapies to enhance recovery after stroke

    • Testing ADK manipulation together with cell-based therapies

  • Biomarker development:

    • Validating ADK as a predictive biomarker for stroke outcome or epileptogenesis risk

    • Developing imaging approaches to monitor ADK expression non-invasively

    • Correlating ADK levels with treatment responsiveness

  • Precision medicine approaches:

    • Region-specific ADK manipulation based on individualized injury patterns

    • Cell-type selective ADK modification targeting specific pathological processes

    • Temporal modulation of ADK expression at different disease stages

  • Extended applications beyond epilepsy and stroke:

    • Investigating ADK's role in traumatic brain injury

    • Exploring potential applications in neurodegenerative disorders

    • Examining ADK's role in neuroinflammatory conditions

  • Advanced genetic approaches:

    • Conditional and inducible ADK knockout/knockdown models

    • Single-cell ADK manipulation technologies

    • CRISPR-based approaches for precise ADK modification

How might ADK manipulation be translated to clinical applications?

The translation of ADK manipulation strategies to clinical applications faces both challenges and opportunities:

  • Therapeutic approaches:

    • Local ADK inhibition through site-specific drug delivery systems

    • Gene therapy approaches using AAV vectors for long-term ADK modulation

    • Cell-based therapies using ADK-deficient stem cells as "adenosine factories"

  • Target conditions:

    • Acute neuroprotection: Rapid ADK inhibition following stroke or traumatic brain injury

    • Epileptogenesis prevention: Early intervention after initial seizures or status epilepticus

    • Chronic epilepsy management: Long-term ADK reduction in established epileptogenic zones

  • Delivery challenges:

    • Blood-brain barrier penetration for small molecule ADK inhibitors

    • Targeted delivery of gene therapy vectors to affected brain regions

    • Timing of intervention relative to disease progression

  • Safety considerations:

    • Potential side effects of global ADK inhibition

    • Long-term consequences of permanent genetic ADK modification

    • Immunological responses to viral vectors or cell implants

  • Regulatory pathway:

    • Initial focus on severe, treatment-resistant conditions

    • Consideration of orphan drug designation for rare epilepsy syndromes

    • Development of companion diagnostics to identify patients most likely to benefit

Product Science Overview

Structure and Function

Adenosine kinase is widely expressed in various tissues, with elevated levels in the placenta, liver, muscle, and kidney . The enzyme’s primary function is to phosphorylate adenosine, a process that is vital for maintaining cellular energy balance and nucleotide metabolism . The recombinant form of mouse adenosine kinase is typically produced using E. coli expression systems, which allows for the generation of high-purity enzyme preparations .

Biological Significance

Adenosine itself is a primordial metabolite and regulator of numerous biochemical reactions related to metabolism and genetics . By converting adenosine to AMP, ADK helps regulate the availability of adenosine for various signaling pathways. This regulation is critical because adenosine can activate specific signaling pathways by binding to adenosine receptors .

Pathological Implications

Dysfunction in ADK activity is associated with several pathologies, including diabetes, epilepsy, and cancer . For instance:

  • Diabetes: Inhibiting ADK activity has been shown to promote the replication of primary insulin-producing beta cells in diabetic mouse models .
  • Epilepsy: ADK is identified as a neuropathological marker of the epileptic brain .
  • Cancer: Altered ADK activity can influence tumor growth and progression .
Therapeutic Potential

Given its significant roles in various diseases, ADK is considered a rational therapeutic target . Researchers are exploring ways to modulate ADK activity to develop treatments for conditions like diabetes and epilepsy.

Research and Applications

Recombinant mouse adenosine kinase is used extensively in research to study its enzymatic properties and potential therapeutic applications. The enzyme’s activity is often measured using phosphatase-coupled methods, which provide insights into its kinetic properties and regulatory mechanisms .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.