ADK Human, Active

Adenosine Kinase Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Biochemical Properties of ADK Human, Active

ADK Human, Active is a monomeric protein with a molecular mass of 40.5 kDa and 362 amino acids, produced via recombinant expression in E. coli . Key features include:

PropertyDetails
Gene NameADK
IsoformsCytoplasmic (ADK-S, 38.7 kDa) and nuclear (ADK-L, 40.5 kDa)
Catalytic MechanismOrdered Bi-Bi reaction mechanism
Substrate SpecificityAdenosine, ATP, and analogs (e.g., toyocamycin, formycin A)
Stabilizing Agents20% glycerol, 1 mM DTT, 20 mM Tris-HCl buffer (pH 8.0)

Catalytic Activity and Assay Conditions

ADK catalyzes the phosphorylation of adenosine using ATP as a phosphate donor:
ATP + adenosineADP + AMP\text{ATP + adenosine} \rightarrow \text{ADP + AMP}

Key Assay Parameters

ParameterValue
Optimal pH7.5–8.0
Temperature37°C
Unit Definition1 unit = 1 µmole adenosine phosphorylated/min
Coupled EnzymesPyruvate kinase (PK) and lactate dehydrogenase (LDH)

Neurological Disorders

  • Epilepsy: ADK overexpression reduces adenosine, increasing neuronal excitability. ADK-deficient mice (Adk Δbrain) exhibit seizures and cognitive deficits .

  • Synaptic Plasticity: ADK deficiency enhances adenosine A2A receptor signaling, impairing hippocampal learning .

Metabolic and Genetic Roles

  • Hypermethioninemia: ADK mutations disrupt methylation cycles, leading to elevated S-adenosylhomocysteine (SAH) .

  • Diabetes and Cancer: ADK inhibitors promote insulin-producing β-cell replication and modulate cancer cell proliferation .

Challenges and Future Directions

  • Therapeutic Targeting: Developing isoform-specific inhibitors remains challenging due to structural homology with other kinases .

  • Clinical Translation: ADK’s dual role in adenosine regulation and methylation necessitates tissue-selective modulation .

Product Specs

Introduction

Adenosine kinase (ADK) is a key enzyme regulating levels of adenosine and adenine nucleotides. It plays a vital role in various physiological processes, particularly in the cardiovascular, nervous, respiratory, and immune systems. ADK inhibitors are of significant pharmacological interest for their potential as anti-inflammatory agents and for increasing adenosine levels in the blood vessels.

Description

Recombinant ADK, expressed in E. coli, is a single polypeptide chain devoid of glycosylation. It encompasses amino acids 22 to 362, resulting in a molecular weight of 40.5 kDa. The protein includes an N-terminal His-tag (21 amino acids) and is purified using proprietary chromatographic methods.

Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation

The ADK protein solution is provided at a concentration of 0.5 mg/ml and contains the following components: 20% glycerol, 20 mM Tris-HCl buffer (pH 8.0), 1 mM DTT, 1 mM EDTA, and 50 mM NaCl.

Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For longer-term storage, it is recommended to freeze the product at -20°C. Adding a carrier protein such as HSA or BSA (0.1%) is advisable for extended storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the ADK protein is determined to be greater than 95.0% using SDS-PAGE analysis.
Biological Activity

The specific activity of the enzyme is measured to be greater than 30 pmol/min/µg. Specific activity represents the amount of enzyme required to convert 1.0 picomole of adenosine to AMP per minute. This measurement is conducted at 37°C and pH 7.5 using a coupled enzyme assay system involving pyruvate kinase (PK) and lactate dehydrogenase (LDH).

Synonyms

Adenosine 5'-phosphotransferase, EC 2.7.1.20, AK, ADK, Adenosine Kinase, Adenosine 5-Phosphotransferase, Testicular Tissue Protein Li 14, EC 2.7.1.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MRENILFGMG NPLLDISAVV DKDFLDKYSL KPNDQILAED KHKELFDELV KKFKVEYHAG GSTQNSIKVA QWMIQQPHKA ATFFGCIGID KFGEILKRKA AEAHVDAHYY EQNEQPTGTC AACITGDNRS LIANLAAANC YKKEKHLDLE KNWMLVEKAR
VCYIAGFFLT VSPESVLKVA HHASENNRIF TLNLSAPFIS QFYKESLMKV MPYVDILFGN ETEAATFARE QGFETKDIKE IAKKTQALPK MNSKRQRIVI FTQGRDDTIM ATESEVTAFA VLDQDQKEII DTNGAGDAFV GGFLSQLVSD KPLTECIRAG HYAASIIIRR TGCTFPEKPD
FH.

Q&A

What is adenosine kinase and what is its primary biochemical function?

Adenosine kinase (ADK, EC 2.7.1.20) is a ubiquitous enzyme that catalyzes the phosphorylation of the purine nucleoside adenosine at the 5' position in an ATP-dependent manner. Biochemically, it transfers the γ-phosphate from ATP to the 5'-hydroxyl group of adenosine, producing AMP. This reaction is a key step in the adenosine salvage pathway .

ADK serves as a crucial regulator of adenosine homeostasis in various tissues, particularly in the brain. It works alongside adenosine deaminase to control both intracellular and extracellular adenosine concentrations. This regulation is physiologically significant as adenosine functions as an important modulator of central nervous system activities and as a signaling molecule involved in hypoxia, inflammation, and nociception .

What are the different isoforms of human ADK and how do they differ functionally?

In mammals including humans, ADK exists in two distinct isoforms that differ in their N-terminal regions:

  • ADK-long (nuclear isoform): Contains an additional 21 amino acids at the N-terminus that replace the first 4 amino acids of the short isoform. This isoform is specifically localized in the nucleus.

  • ADK-short (cytoplasmic isoform): Lacks the extended N-terminal sequence and is predominantly found in the cytoplasm.

While both isoforms display identical catalytic activities and share the same core structure, their differential subcellular localization suggests distinct roles in regulating compartmentalized adenosine metabolism. These isoforms are independently regulated at the transcriptional level, with the promoter for the short isoform located within the first large intron of the ADK gene .

When designing experiments to study ADK function, researchers should consider these isoform differences and their potential compartment-specific effects. Immunolocalization studies using isoform-specific antibodies can help distinguish between nuclear and cytoplasmic ADK populations in research samples.

What is the genomic organization of the human ADK gene?

The human ADK gene exhibits several unusual genomic characteristics:

  • Chromosomal location: Mapped to chromosome 10 in the 10q11-10q24 region.

  • Gene size: Despite encoding a protein of approximately 1 Kb in coding sequence length, the gene itself spans an unusually large genomic region of approximately 546 Kb.

  • Structural organization: Contains 11 exons (ranging from 36 to 173 bp in length) interspersed with 10 introns (varying from 4.2 Kb to 128.6 Kb, with an average length of ~50 Kb).

  • Coding to non-coding ratio: The ratio of non-coding to coding sequence for human ADK exceeds 550, which is among the highest known for any gene.

  • Gene arrangement: The ADK gene is arranged in a head-to-head orientation with the gene encoding the μ3A adaptor protein, and both genes are transcribed from a single bidirectional promoter. This arrangement appears to be unique to amniotes (mammals, birds, and reptiles) .

This complex genomic organization has implications for researchers studying ADK regulation, as the extensive non-coding regions may contain important regulatory elements affecting expression patterns and tissue specificity.

What are the standard methods for measuring ADK enzymatic activity in research samples?

ADK activity can be assessed through several established methodological approaches:

  • Phosphorylation assays: These typically measure the phosphorylation of adenosine to AMP using radiolabeled ATP (γ-32P-ATP) as a phosphate donor. The reaction products are separated by thin-layer chromatography or HPLC, and radioactivity is quantified to determine enzyme activity.

  • Coupled enzyme assays: These assays link ADK activity to the production or consumption of NAD(P)H, which can be monitored spectrophotometrically. For example, AMP produced by ADK can be further metabolized in a reaction sequence coupled to NAD(P)H oxidation or reduction.

  • PRECICE® ADK Phosphorylation Assay: This commercially available kit allows for rapid evaluation of substrate properties for human adenosine kinase .

A typical ADK activity experiment from research literature involves homogenizing tissue samples in an appropriate buffer, performing subcellular fractionation if necessary, and then measuring activity in clarified extracts. For instance, ADK activity measurements in studies of astrocytomas were performed on surgical specimens from tumor tissue, peritumoral cortex, and non-epileptic cortex to compare enzymatic function in different contexts .

When designing ADK activity assays, researchers should consider factors such as pH optimum (typically 6.5-7.5), requirements for divalent cations (usually Mg2+), and potential inhibitors that might be present in biological samples.

How can researchers effectively express and purify active human ADK for in vitro studies?

To obtain active human ADK for biochemical and structural studies, researchers typically employ recombinant expression systems. Key methodological considerations include:

  • Expression system selection: E. coli is the most commonly used expression system for human ADK. The ADK gene can be cloned following RT-PCR amplification of mRNA from human cells (such as hepatoma cells) .

  • Construct design:

    • For full activity, expression constructs typically include amino acids 22-362 of the human ADK sequence

    • Addition of purification tags (e.g., His6-tag) facilitates downstream purification

    • Codon optimization may improve expression in bacterial systems

  • Purification strategy:

    • Initial capture using affinity chromatography (e.g., Ni-NTA for His-tagged proteins)

    • Further purification by ion-exchange and/or size-exclusion chromatography

    • Typical purity achieved is >95% as assessed by SDS-PAGE

  • Activity verification:

    • Enzymatic assays to confirm functional activity of the purified protein

    • Thermal stability assessments to ensure proper folding

Commercially available recombinant human ADK typically consists of the 22-362 amino acid fragment expressed in E. coli with an N-terminal His-tag and maintained in appropriate storage buffer to preserve activity .

How does ADK expression change in human brain tumors, and what methodologies best capture these alterations?

Research has revealed significant alterations in ADK expression in brain tumors compared to normal brain tissue. Methodologically, these changes can be assessed through:

  • Immunohistochemistry (IHC): This approach has demonstrated primarily cytoplasmic ADK labeling in tumors and peritumoral tissue containing infiltrating tumor cells. Studies have shown significantly stronger ADK immunoreactivity in tumor and peritumoral tissue compared to normal white matter and cortex, with particularly elevated expression in WHO grade III astrocytomas .

  • Western blot analysis: This quantitative technique confirms overexpression of ADK protein in tumor tissue compared to control samples. When performing western blots for ADK, researchers should consider using isoform-specific antibodies to distinguish between the nuclear and cytoplasmic variants .

  • ADK activity measurements: Enzymatic activity assays reveal functional consequences of altered ADK expression. Higher activity has been observed in astrocytoma WHO grade III samples compared to control tissue .

  • Correlation with clinical parameters: Research has found significantly higher ADK expression in peritumoral infiltrated tissue from patients with epilepsy compared to non-epileptic patients, suggesting a potential role in tumor-associated seizures .

When designing studies to investigate ADK in brain tumors, researchers should consider including:

  • Multiple control tissues (normal white matter, normal cortex)

  • Careful histological characterization of samples (using H&E staining and markers like GFAP, NeuN, Ki67, and p53)

  • Correlation of ADK expression with clinical parameters like seizure history and tumor grade

What experimental approaches are used to investigate the role of ADK in epilepsy?

ADK has emerged as a key player in epileptogenesis, particularly in the context of tumor-associated epilepsy. Several research methodologies are employed to study this relationship:

  • Comparative expression studies: Analysis of ADK levels in epileptic versus non-epileptic tissue samples provides correlative evidence. For example, significantly higher ADK expression has been observed in peritumoral infiltrated tissue from epileptic patients compared to non-epileptic patients with brain tumors .

  • Seizure models: In research settings, ADK overexpression models can be used to study the relationship between adenosine regulation and seizure susceptibility. Conversely, ADK inhibition results in elevated adenosine concentrations and demonstrated anti-seizure effects in experimental models .

  • Cellular studies: Investigation of astrocyte-specific ADK overexpression helps understand how glial dysfunction contributes to epileptogenesis, as ADK is primarily expressed in astrocytes in the adult brain. The overexpression of ADK leads to decreased adenosine levels and loss of inhibition of neuronal excitability by astrocytes, which has been proposed as an underlying mechanism in epilepsy progression .

  • Pharmacological interventions: Studying the effects of ADK inhibitors on seizure susceptibility provides insights into therapeutic potential. Many ADK inhibitors have shown anti-seizure properties in animal models .

When designing epilepsy-related ADK studies, researchers should carefully consider the selection of appropriate control tissues and the correlation of molecular findings with clinical seizure parameters.

How do the two ADK isoforms differ in their regulation and cellular functions?

Understanding the distinct roles of ADK isoforms requires specialized research approaches:

  • Differential subcellular localization: The long isoform (ADK-long) localizes to the nucleus, while the short isoform (ADK-short) resides in the cytoplasm. This distribution can be visualized using isoform-specific antibodies and confocal microscopy .

  • Transcriptional regulation: The two isoforms are independently regulated at the transcriptional level, with the promoter for the short isoform located within the first large ADK intron. Researchers can use promoter-reporter constructs to study the differential regulation of these isoforms .

  • Functional implications: While both isoforms catalyze the same reaction, their different subcellular localizations suggest distinct roles:

    • Cytoplasmic ADK-short likely regulates cytosolic adenosine levels and adenosine flux across the cell membrane

    • Nuclear ADK-long may influence nuclear adenosine pools, potentially affecting processes like mRNA methylation

  • Isoform-specific knockdown/overexpression: Selectively manipulating individual isoforms through molecular techniques (siRNA, CRISPR, isoform-specific expression constructs) can help elucidate their specific functions.

Researchers should note that the relative expression of these isoforms may vary across tissue types and pathological conditions, potentially contributing to tissue-specific adenosine metabolism patterns.

What methodological approaches are used to develop and evaluate ADK inhibitors as potential therapeutics?

ADK inhibitors have therapeutic potential in various conditions, including epilepsy, pain, and inflammation. Research methodologies for their development include:

  • Structure-based drug design: The availability of human ADK crystal structure provides a structural basis for rational design of inhibitors. Virtual screening, molecular docking, and structure-activity relationship studies guide compound optimization .

  • High-throughput screening (HTS): Systems like the PRECICE® ADK Assay Kit facilitate rapid screening of compound libraries to identify novel ADK inhibitors .

  • Enzymatic inhibition assays: Measurement of ADK activity in the presence of candidate inhibitors determines IC50 values and inhibition kinetics. These assays typically use purified recombinant ADK and monitor adenosine phosphorylation .

  • Cell-based assays: Testing compounds in cellular systems evaluates membrane permeability, intracellular target engagement, and effects on downstream adenosine signaling.

  • Animal models: Evaluation in disease models assesses in vivo efficacy:

    • Seizure models for antiepileptic potential

    • Inflammatory pain models for analgesic effects

    • Ischemia models for neuroprotective properties

  • Selectivity profiling: Assessing activity against related enzymes and adenosine receptors ensures target specificity and helps predict potential side effects.

When developing ADK inhibitors, researchers should consider the differential tissue expression of ADK and aim for compounds with appropriate tissue distribution profiles for their intended therapeutic application.

What techniques are used to study the role of ADK in regulating adenosine-dependent signaling pathways?

Understanding how ADK influences adenosine signaling requires integrated methodological approaches:

  • Adenosine measurement techniques:

    • Microdialysis for in vivo extracellular adenosine monitoring

    • HPLC or LC-MS/MS for tissue adenosine quantification

    • Biosensor approaches for real-time adenosine detection

  • Receptor pharmacology:

    • Radioligand binding assays to quantify adenosine receptor occupancy

    • cAMP/calcium assays to measure receptor activation

    • Selective adenosine receptor agonists/antagonists to dissect pathway contributions

  • Genetic manipulation approaches:

    • ADK knockout/knockdown to increase adenosine tone

    • ADK overexpression to decrease adenosine availability

    • Inducible and tissue-specific genetic models to control spatiotemporal ADK expression

  • Signal transduction analysis:

    • Western blotting for downstream signaling components (e.g., MAPK, AKT)

    • Phosphoproteomic approaches to identify signaling networks

    • Reporter gene assays for transcriptional readouts

  • Integrated physiological readouts:

    • Electrophysiology to measure neuronal activity

    • Metabolic assays for adenosine's effects on energy metabolism

    • Functional assays specific to the system under study (e.g., vascular tone, inflammatory mediator production)

When designing experiments to investigate ADK's role in adenosine signaling, researchers should consider the presence of four different adenosine receptor subtypes (A1, A2A, A2B, A3) with distinct signaling mechanisms and the complex interplay between adenosine production, metabolism, and transport processes.

What are the methodological considerations when studying ADK mutations and variants?

Investigating ADK mutations presents several technical challenges. Researchers should consider these methodological approaches:

  • Mutation identification and characterization:

    • Next-generation sequencing to identify novel variants

    • Site-directed mutagenesis to introduce specific mutations for functional studies

    • Bioinformatic prediction tools to assess potential functional impacts

  • Functional consequence assessment:

    • Activity assays comparing wild-type and mutant proteins

    • Thermal stability measurements to detect folding defects

    • Subcellular localization studies to identify trafficking abnormalities

  • Cell-based models:

    • Heterologous expression systems to study isolated mutant effects

    • CRISPR/Cas9 gene editing to introduce mutations into endogenous loci

    • Patient-derived cell lines to study mutations in native contexts

  • Spontaneous mutation models:

    • CHO cell models have yielded interesting ADK mutants with different genetic and biochemical properties

    • Some mutants show unusually high spontaneous mutation frequency (10^-3-10^-4) with large deletions in the ADK gene, leading to loss of several introns and exons

    • Other mutants show selective effects on the expression of specific ADK isoforms

When studying ADK variants, researchers should consider that mutations may affect enzyme activity, stability, regulation, or subcellular localization, potentially leading to diverse physiological consequences.

How can researchers effectively study the tissue-specific expression patterns of ADK?

ADK expression varies across tissues and cell types, requiring specialized approaches for comprehensive characterization:

  • Transcriptional profiling:

    • qRT-PCR with isoform-specific primers to quantify ADK-long vs. ADK-short mRNA levels

    • RNA-seq to examine tissue-specific expression patterns

    • Single-cell RNA-seq to resolve cell type-specific expression

  • Protein detection methods:

    • Western blotting with isoform-specific antibodies for quantitative analysis

    • Immunohistochemistry for spatial distribution in tissue sections

    • Immunofluorescence with co-staining for cell type markers to identify ADK-expressing cells

  • Promoter analysis:

    • Reporter gene assays to study tissue-specific promoter activity

    • ChIP-seq to identify transcription factor binding patterns

    • DNA methylation analysis to examine epigenetic regulation

  • Translational considerations:

    • Different tissues may exhibit varying ratios of ADK isoforms

    • Astrocytes typically show high ADK expression in the adult brain

    • Pathological conditions can dramatically alter ADK expression patterns

When studying tissue-specific ADK expression, researchers should carefully validate antibody specificity and include appropriate controls, as cross-reactivity can lead to misinterpretation of expression patterns.

What emerging technologies might advance our understanding of ADK biology?

Several cutting-edge approaches show promise for deepening our understanding of ADK:

  • Cryo-EM for structural studies:

    • Higher-resolution structures of ADK complexes with substrates and inhibitors

    • Visualization of conformational changes during catalysis

    • Structural insights into isoform-specific protein interactions

  • Advanced genome editing:

    • Base editing and prime editing for precise ADK modifications

    • Tissue-specific and inducible CRISPR systems for spatiotemporal control

    • Humanized animal models to better recapitulate human ADK biology

  • Spatially resolved transcriptomics and proteomics:

    • Mapping ADK expression patterns with subcellular resolution

    • Correlating ADK levels with tissue microenvironments

    • Understanding regional variability in brain ADK expression

  • Metabolomic approaches:

    • Tracking adenosine flux through metabolic pathways

    • Identifying novel ADK substrates and products

    • Examining metabolic consequences of ADK modulation

  • Translational imaging techniques:

    • PET ligands for ADK to enable in vivo imaging

    • Functional imaging to correlate ADK activity with physiological parameters

    • Patient stratification based on ADK expression/activity profiles

These emerging technologies could provide unprecedented insights into ADK function and regulation, potentially revealing new therapeutic opportunities.

How can ADK research findings be effectively translated into clinical applications?

Bridging the gap between basic ADK research and clinical applications requires strategic approaches:

  • Biomarker development:

    • ADK expression analysis in patient samples as potential diagnostic or prognostic tools

    • Correlation of ADK levels with disease progression or treatment response

    • Development of non-invasive methods to assess ADK activity

  • Therapeutic targeting strategies:

    • Optimization of ADK inhibitors with improved pharmacokinetics and specificity

    • Exploration of isoform-selective inhibitors to minimize side effects

    • Development of targeted delivery systems for tissue-specific ADK modulation

  • Disease-specific considerations:

    • Epilepsy: Focal delivery of ADK inhibitors to seizure foci

    • Cancer: Exploitation of altered ADK activity in tumors for targeted therapies

    • Inflammation: Development of peripheral ADK inhibitors for inflammatory conditions

  • Companion diagnostics:

    • Identification of patient populations most likely to benefit from ADK-targeted therapies

    • Development of assays to monitor treatment efficacy

    • Integration of genetic and molecular profiling to guide personalized approaches

Research into adenosine-regulating drugs has already shown promise for analgesic and anti-inflammatory applications, treatment of schizophrenia, and limiting brain injury after ischemic stroke . Continued investigation of ADK's role in these and other conditions will likely yield additional therapeutic opportunities.

Product Science Overview

Structure and Function

Human recombinant adenosine kinase is typically produced in Escherichia coli (E. coli) and is available in a purified, active form. The enzyme consists of 362 amino acids and has a molecular mass of approximately 40.5 kDa . It is often tagged with a His tag at the N-terminus to facilitate purification through standard chromatography techniques .

Adenosine kinase is involved in the regulation of intra- and extracellular adenosine concentrations. Adenosine itself is an important modulator of central nervous system functions and acts as a signaling molecule in various physiological processes, including hypoxia, inflammation, and nociception . By phosphorylating adenosine, ADK helps regulate these processes and maintain cellular homeostasis.

Biological Significance

The activity of adenosine kinase is critical for several biological functions. Inhibition of ADK can lead to an increase in local adenosine concentrations, which has been shown to reduce seizure susceptibility and nociception in vivo . Dysregulation of ADK activity is associated with various pathologies, including diabetes, epilepsy, and cancer . As a result, ADK is considered a potential therapeutic target for drug discovery, with adenosine-regulating drugs being tested as new analgesic and anti-inflammatory agents .

Therapeutic Applications

Adenosine kinase is responsible for the phosphorylation and subsequent clinical activity of several therapeutically useful nucleosides. These include the antiviral drug ribavirin, the immunosuppressive drug mizoribine, and the anticancer C-nucleoside tiazofurin . The enzyme’s role in these processes highlights its importance in the development of new therapeutic agents.

Research and Development

The X-ray crystallographic structure of human adenosine kinase has been described, providing a structural basis for the rational design and optimization of new ADK inhibitors . This structural information is invaluable for researchers aiming to develop novel drugs that target ADK and modulate its activity for therapeutic purposes.

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.