AK1 Human

Adenylate Kinase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in Cellular Metabolism

AK1 ensures energy homeostasis via two primary mechanisms:

  1. Nucleotide Recycling: Balances adenine nucleotide pools (ATP, ADP, AMP) during metabolic stress, enabling rapid ATP regeneration .

  2. AMP Signaling: Generates AMP during energy deficits, indirectly activating AMP-activated protein kinase (AMPK), a regulator of energy production and consumption .

Key Pathways:

  • Cardiac Energetics: AK1 deficiency in mice exacerbates ATP loss during ischemia, impairing post-ischemic recovery, while overexpression improves functional recovery but alters cardiac metabolites (e.g., ↑creatine, ↓taurine) .

  • Erythrocyte Function: AK1 mutations cause nonspherocytic hemolytic anemia due to inadequate ATP synthesis, leading to chronic anemia and hyperbilirubinemia .

Clinical and Pathological Significance

Genetic Disorders:

  • AK1 Deficiency: Rare autosomal recessive condition characterized by chronic hemolytic anemia (Hb 8–9 g/dL) and, in severe cases, psychomotor impairment . Enzyme activity in patients is <30% of normal .

  • Diagnostic Testing: Mayo Clinic Laboratories offer AK1 activity assays using kinetic spectrophotometry to confirm deficiency .

Therapeutic Implications:

  • Cardioprotection: AK1 overexpression in mice improved post-ischemic recovery but caused baseline ventricular dysfunction and arrhythmias, highlighting a trade-off between energetic adaptation and cardiac stability .

  • Neuromuscular Function: Elevated AK1 levels correlate with increased locomotor activity in animal models, suggesting roles beyond metabolism .

Recent Research Findings

Study FocusMethodologyKey FindingsImplications
Conformational Dynamics (2022)NMR spectroscopyADP binding rigidifies α3α4 and α7α8 domains, enabling catalytic conformational shifts Explains substrate-driven activation mechanism
Cardiac Overexpression (2021)Transgenic mice modelsAK1-OE hearts showed ↑AMP (20%), ↑creatine, and altered lipid profiles, with mixed functional outcomes Highlights metabolic trade-offs in AK1 modulation
Behavioral Impact (2022)Medaka fish modelsAK1 overexpression linked to hyperactivity and altered thiamine metabolism Suggests roles in neural signaling

Challenges and Future Directions

  • Therapeutic Targeting: AK1’s dual role in cytoprotection and metabolic disruption complicates drug development. Strategies to temporally regulate its activity during ischemia are under exploration .

  • Structural Insights: Cryo-EM and mutagenesis studies are needed to map residues critical for catalysis (e.g., D93, D140, D141) and refine enzyme engineering .

Product Specs

Introduction
AK1 is a small, ubiquitous enzyme crucial for cell growth and maintenance. It helps regulate adenine nucleotide levels within cells by catalyzing the reversible transfer of the terminal phosphate group between ATP and AMP. Found in the cytosol of skeletal muscle, brain, and erythrocytes, defects in the AK1 gene can lead to a form of hemolytic anemia.
Description
Recombinant human AK1, expressed in E. coli, is a single, non-glycosylated polypeptide chain. This protein, fused with a 20 amino acid His tag at the N-terminus, contains 214 amino acids (1-194 a.a.) and has a molecular weight of 23.7kDa. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
The AK1 protein solution is provided at a concentration of 1mg/ml and contains 20mM Tris-HCl (pH 7.5) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For longer storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Biological Activity
The specific activity is greater than 600 units/mg. One unit is defined as the amount of enzyme that catalyzes the conversion of 2.0 µmoles of ADP to ATP and AMP per minute at pH 7.5 and 25°C.
Synonyms
Adenylate kinase isoenzyme 1, AK 1, ATP-AMP transphosphorylase 1, Myokinase, AK1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MEEKLKKTKI IFVVGGPGSG KGTQCEKIVQ KYGYTHLSTG DLLRSEVSSG SARGKKLSEI MEKGQLVPLE TVLDMLRDAM VAKVNTSKGF LIDGYPREVQ QGEEFERRIG QPTLLLYVDA GPETMTQRLL KRGETSGRVD DNEETIKKRL ETYYKATEPV IAFYEKRGIV RKVNAEGSVD SVFSQVCTHL DALK.

Q&A

What is the primary biochemical function of AK1 in human cells?

Adenylate Kinase 1 (AK1) catalyzes the reaction 2ADP ↔ ATP + AMP, which plays a critical role in cellular energy homeostasis. This enzyme is particularly important under metabolic stress conditions, where it helps extract additional energy and promotes energetic balance. Unlike other adenylate kinases, AK1 is primarily cytosolic and demonstrates tissue-specific expression patterns, with particular prominence in brain and cardiac tissues. The enzymatic activity of AK1 serves as a phosphotransfer network that connects different cellular compartments and maintains adenine nucleotide ratios .

How does AK1 interact with other components of purine metabolism pathways?

AK1 functions within a broader network of purine metabolism enzymes. Based on research findings, AK1 is one of several genes involved specifically in adenosine metabolism, alongside adenosine kinases (Adk1, Adk2), other adenylate kinases (Ak6), and nucleoside diphosphate-kinases (awd, nmdyn-D6). These enzymes collectively regulate adenosine levels in tissues. When AK1 is knocked down, studies show increased adenosine levels, suggesting its role in converting adenosine to other adenine nucleotides. This position within the purine metabolism pathway makes AK1 particularly relevant for neurological conditions where adenosine levels may impact neuronal survival .

What are the established methods for measuring AK1 activity in human tissue samples?

Researchers typically measure AK1 activity using enzymatic assays that track the conversion between ADP, ATP, and AMP. The standard approach involves:

  • Tissue homogenization in appropriate buffer conditions

  • Spectrophotometric or fluorometric assays that couple the AK1 reaction to other enzymes whose activity can be measured

  • Comparison to total creatine kinase and citrate synthase activities as controls

In cardiac tissue studies, researchers have successfully measured a 31% increase in AK1 activity in transgenic overexpression models while confirming unchanged total creatine kinase and citrate synthase activities . For human samples, similar enzymatic approaches can be applied with appropriate normalization to total protein content or reference enzymes.

What evidence supports AK1's role in Parkinson's disease pathophysiology?

Recent genetic screening and metabolomics studies have identified glial adenosine metabolism, including AK1 function, as potentially crucial in Parkinson's disease (PD) mechanisms. Research findings demonstrate that:

  • Knockdown of AK1 rescues dopaminergic neuron loss in PD models

  • AK1 inhibition reduces α-synuclein aggregation, a hallmark pathological feature of PD

  • Manipulation of AK1 improves multiple pathologic α-synuclein phenotypes, including large aggregates, oligomers, and phosphorylated forms

  • Targeting AK1 appears to affect bioenergetic pathways relevant to neurodegeneration

These observations suggest that glial adenosine metabolism, specifically through AK1 modulation, represents a novel therapeutic target for PD. The enzyme's relatively well-tolerated knockdown profile (as evidenced by viable and fertile AK1-knockout mice with only mild stress-induced phenotypes) makes it particularly interesting as a potential intervention point .

What experimental models are most appropriate for studying AK1 in the context of neurodegeneration?

Based on current research approaches, several experimental models have proven effective for studying AK1 in neurodegeneration:

  • Drosophila models: Forward genetic screening in flies has successfully identified AK1 as a potential therapeutic target. This model allows for rapid genetic manipulation and assessment of neurodegenerative phenotypes.

  • Mouse models: AK1-knockout mice (AK1-/-) have been established and show viability with mild phenotypes, making them suitable for studying the long-term effects of AK1 modulation on neurodegeneration.

  • Cell culture systems: Glial-neuronal co-culture systems allow for the study of cell-specific effects of AK1 manipulation, particularly the impact of glial AK1 on neuronal survival.

Each model system offers different advantages, with fly models providing high-throughput genetic screening capabilities, mouse models offering mammalian physiological relevance, and cell culture systems allowing for detailed mechanistic studies .

How can researchers quantify the effects of AK1 manipulation on α-synuclein pathology?

Researchers have developed several quantitative approaches to assess the impact of AK1 manipulation on α-synuclein pathology:

  • Aggregation analysis: Quantification of large α-synuclein aggregates using immunohistochemistry with appropriate antibodies, followed by counting of inclusion bodies in defined brain regions

  • Oligomer quantification: Biochemical separation of oligomeric species using non-denaturing gel electrophoresis or size exclusion chromatography, combined with western blotting

  • Phosphorylation assessment: Measurement of phosphorylated α-synuclein (particularly at Ser129) using phospho-specific antibodies in immunohistochemistry or western blot analyses

  • Dopaminergic neuron survival: Stereological counting of tyrosine hydroxylase-positive neurons in the substantia nigra to correlate α-synuclein pathology with neurodegeneration

These measurements have demonstrated that glial AK1 knockdown improves multiple aspects of α-synuclein pathology, suggesting a multifaceted mechanism of protection .

What are the effects of AK1 overexpression on cardiac function and metabolism?

Research on cardiac-specific AK1 overexpression has revealed complex and sometimes unexpected effects on cardiac function and metabolism:

  • Baseline cardiac function: Male AK1-overexpressing (AK1-OE) mice exhibit mild in vivo dysfunction with lower left ventricular pressure, impaired relaxation, and reduced contractile reserve.

  • Left ventricular changes: AK1-OE males show a 19% increase in left ventricular weight due to higher tissue water content, without evidence of hypertrophy or fibrosis.

  • Metabolic alterations: AK1-OE hearts demonstrate significant metabolic changes, including:

    • Raised creatine levels

    • Unaltered total adenine nucleotides

    • 20% higher AMP levels without activation of AMP-activated protein kinase

    • Altered levels of various metabolites, including elevated aspartate, tyrosine, sphingomyelin, and cholesterol, with decreased taurine and triglycerides

These findings indicate that even modest elevation of AK1 has significant impacts on cardiac physiology under basal conditions, contrary to the initial hypothesis that increased AK1 would primarily show effects under stress conditions .

How does AK1 activity influence cardiac response to ischemia/reperfusion injury?

The relationship between AK1 activity and cardiac response to ischemia/reperfusion (I/R) injury appears complex:

  • Functional recovery: Surviving AK1-OE hearts showed improved functional recovery following I/R injury in ex vivo experiments.

  • Arrhythmia risk: Four of seven AK1-OE hearts developed terminal arrhythmia in ex vivo global no-flow I/R experiments (compared to zero in wild-type).

  • Infarct size: AK1-OE did not significantly influence infarct size in vivo models of I/R injury.

  • Ex vivo artifacts: The arrhythmias observed ex vivo may represent an artifact of adenine nucleotide loss during cannulation rather than a true physiological effect.

These findings suggest that while modest elevation of AK1 may improve functional recovery following I/R injury, it also introduces potential complications that require careful experimental design to distinguish between true physiological effects and experimental artifacts .

What methodology should be employed to accurately assess metabolic changes in AK1-modified cardiac tissue?

Based on the research approaches documented, a comprehensive metabolic assessment of AK1-modified cardiac tissue should include:

  • Enzymatic activity assays:

    • Direct measurement of AK1 activity

    • Parallel assessment of related enzymes (creatine kinase, citrate synthase) as controls

  • Nucleotide profiling:

    • Quantification of ATP, ADP, and AMP levels

    • Assessment of energy charge and adenylate pool size

  • Comprehensive metabolomics:

    • 1H-NMR analysis of tissue extracts for small molecule metabolites

    • Targeted assays for creatine and phosphocreatine

  • Signaling pathway analysis:

    • Western blotting for AMP-activated protein kinase (AMPK) activity

    • Assessment of downstream targets of metabolic signaling

  • Physiological correlates:

    • Tissue water content measurement

    • Histological assessment for hypertrophy and fibrosis

This multi-modal approach allows researchers to connect enzymatic changes to broader metabolic alterations and their physiological consequences .

What are the key considerations for designing genetic knockdown experiments targeting AK1?

When designing genetic knockdown experiments for AK1, researchers should consider:

  • Cell/tissue specificity: Target knockdown to specific cell types (e.g., glial cells for neurodegeneration studies) to distinguish cell-autonomous from non-cell-autonomous effects.

  • Knockdown efficiency verification:

    • mRNA quantification via qPCR

    • Protein level assessment via western blotting

    • Enzymatic activity measurement to confirm functional consequences

  • Control selection: Include appropriate controls such as non-targeting sequences and rescue experiments where knockdown is complemented with expression of knockdown-resistant constructs.

  • Phenotypic assessment timeline: Establish appropriate timepoints for phenotype evaluation, considering the kinetics of knockdown and the development of downstream effects.

  • Downstream mechanism validation: Confirm that knockdown produces the expected biochemical changes (e.g., increased adenosine levels in the case of AK1 knockdown).

These considerations have been successfully applied in studies demonstrating that glial AK1 knockdown rescues neurodegeneration and α-synuclein pathology, likely through modulation of adenosine levels .

How should researchers control for potential off-target effects when manipulating AK1 expression?

To control for potential off-target effects in AK1 manipulation studies, researchers should implement:

  • Multiple knockdown/knockout strategies:

    • Use different shRNA/siRNA sequences targeting distinct regions of AK1

    • Compare CRISPR-Cas9 knockout with RNAi approaches

    • Employ conditional knockout systems to control timing and tissue specificity

  • Rescue experiments:

    • Re-express wild-type AK1 resistant to knockdown

    • Test enzymatically inactive AK1 mutants to distinguish structural from enzymatic roles

  • Pathway validation:

    • Manipulate other enzymes in the same pathway to verify consistent outcomes

    • Directly modulate presumed downstream mediators (e.g., adenosine levels)

  • Comprehensive pathway analysis:

    • Measure levels of multiple metabolites in the purine metabolism pathway

    • Assess expression of other adenosine-metabolizing enzymes to detect compensatory changes

  • Cross-species validation:

    • Compare results across different model systems (e.g., fly, mouse, human cells)

    • Validate findings with both genetic and pharmacological approaches

Following these controls helps establish the specificity of observed effects to AK1 modulation rather than off-target consequences of experimental manipulation .

What experimental design approaches are most appropriate for translating AK1 findings from animal models to human applications?

Translational research for AK1 findings should incorporate:

  • Comparative expression analysis:

    • Assess AK1 expression patterns across species in relevant tissues

    • Compare AK1 sequence homology and functional conservation

  • Human tissue validation:

    • Analyze AK1 expression and activity in post-mortem human tissues

    • Compare metabolite profiles between animal models and human samples

  • Human genetic association studies:

    • Investigate AK1 polymorphisms in relevant patient populations

    • Correlate genetic variants with disease risk or progression

  • Human-derived cellular models:

    • Test AK1 manipulation in patient-derived cells (e.g., iPSCs, fibroblasts)

    • Develop humanized animal models expressing human AK1 variants

  • Biomarker development:

    • Identify measurable indicators of AK1 activity in accessible human samples

    • Correlate biomarkers with disease state or treatment response

This translational pipeline has been partially implemented in Parkinson's disease research, where studies have moved from fly models to analyses of adenosine metabolism in human PD patients, aiming to identify potential biomarkers of dysfunctional adenosine metabolism that could guide therapeutic development .

How do changes in AK1 expression or activity impact the broader metabolic network beyond adenosine metabolism?

The impact of AK1 modulation extends beyond immediate adenosine metabolism, affecting:

  • Energy homeostasis networks:

    • AK1 influences the broader phosphotransfer network that maintains cellular energetic balance

    • Changes in AK1 activity alter AMP:ATP and ADP:ATP ratios, potentially affecting numerous AMP- and ATP-sensing processes

  • Metabolite crosstalk:

    • Research has identified unexpected changes in diverse metabolites following AK1 overexpression:

Metabolite ClassDirection of ChangePotential Mechanism
AspartateIncreasedAltered amino acid metabolism linked to nucleotide synthesis
TyrosineIncreasedChanged aromatic amino acid metabolism
SphingomyelinIncreasedModified membrane lipid composition
CholesterolIncreasedAltered sterol metabolism
TaurineDecreasedShifted sulfur amino acid metabolism
TriglyceridesDecreasedModified lipid storage/utilization
  • Signaling pathway integration:

    • Despite elevated AMP levels, AK1 overexpression did not activate AMPK signaling

    • This suggests complex integration of metabolic signals beyond simple substrate availability

These broad metabolic consequences indicate that AK1 sits at a critical node in cellular metabolism, with ripple effects extending to amino acid, lipid, and energy homeostasis pathways .

What are the potential mechanisms by which AK1 modulation affects α-synuclein aggregation in neurodegenerative disease models?

Several potential mechanisms may explain how AK1 modulation influences α-synuclein aggregation:

  • Direct bioenergetic effects:

    • AK1 knockdown may improve neuronal energy availability, reducing α-synuclein aggregation triggered by metabolic stress

    • Changes in ATP/ADP ratios could affect protein quality control systems that regulate α-synuclein turnover

  • Adenosine receptor signaling:

    • Increased adenosine resulting from AK1 knockdown may activate neuroprotective adenosine receptor pathways

    • A1 or A2A adenosine receptor signaling could influence autophagy or proteasome function

  • Inflammation modulation:

    • Adenosine has known anti-inflammatory properties

    • Reduced neuroinflammation through increased adenosine signaling could decrease α-synuclein aggregation

  • Oxidative stress reduction:

    • Adenosine signaling may improve antioxidant defenses

    • Reduced oxidative damage to α-synuclein could decrease its propensity to aggregate

  • Protein phosphorylation regulation:

    • Changes in kinase activity resulting from altered energy metabolism may affect α-synuclein phosphorylation

    • Decreased phosphorylation at Ser129 could reduce α-synuclein aggregation tendency

These mechanisms are not mutually exclusive and likely operate in concert to produce the observed effects of AK1 knockdown on multiple aspects of α-synuclein pathology .

How do tissue-specific differences in AK1 expression patterns influence the functional consequences of AK1 modulation?

Tissue-specific consequences of AK1 modulation reflect its varied expression and function across different cell types:

  • Neural tissue:

    • In the brain, AK1 shows cell-type specific functions, with glial knockdown producing neuroprotective effects

    • The adenosine-mediated communication between glia and neurons appears particularly sensitive to AK1 activity levels

  • Cardiac tissue:

    • Cardiac-specific AK1 overexpression produces complex phenotypes including altered left ventricular weight and impaired function

    • Cardiac metabolism shows specific sensitivity to AK1 levels, with changes in multiple metabolite classes

  • Stress response differences:

    • AK1-deficient mice show mild phenotypes under normal conditions but exhibit stress-specific responses

    • Different tissues may rely on AK1 to varying degrees depending on their metabolic characteristics and energy demands

  • Compensatory mechanisms:

    • The presence of multiple adenylate kinase isoforms (AK1-AK6) with tissue-specific expression patterns

    • Tissues may differ in their ability to compensate for AK1 modulation through alternative enzymes

Understanding these tissue-specific differences is crucial for therapeutic targeting of AK1, as intervention may need to be tailored to specific tissues or cell types to achieve desired effects while minimizing adverse consequences .

What statistical approaches are most appropriate for analyzing complex metabolomic data in AK1 studies?

Analyzing metabolomic data in AK1 research requires sophisticated statistical approaches:

  • Multivariate analysis methods:

    • Principal Component Analysis (PCA) to identify major sources of variation

    • Partial Least Squares-Discriminant Analysis (PLS-DA) to maximize separation between experimental groups

    • Hierarchical clustering to identify metabolite patterns

  • Pathway enrichment analysis:

    • Metabolite Set Enrichment Analysis (MSEA) to identify pathways affected by AK1 modulation

    • Network analysis to map interactions between altered metabolites

  • Time-series analysis:

    • Mixed-effects models for longitudinal metabolomic data

    • Trajectory analysis to identify temporal patterns in metabolite changes

  • Integration with other data types:

    • Correlation analysis between metabolites and physiological parameters

    • Multi-omics integration combining metabolomics with transcriptomics or proteomics

  • Multiple testing correction:

    • False Discovery Rate (FDR) control using Benjamini-Hochberg procedure

    • Permutation testing to establish significance thresholds

These approaches have revealed significant alterations in multiple metabolites in AK1-overexpressing cardiac tissue, including changes in aspartate, tyrosine, sphingomyelin, cholesterol, taurine, and triglycerides, suggesting broad metabolic consequences of AK1 modulation .

How can researchers resolve contradictory findings regarding AK1 function across different experimental systems?

Resolving contradictory findings about AK1 requires systematic investigation of potential sources of variation:

  • Experimental context differences:

    • Compare in vivo versus ex vivo systems (e.g., ex vivo arrhythmias in AK1-OE hearts not observed in vivo)

    • Evaluate acute versus chronic manipulations (genetic modification versus pharmacological inhibition)

    • Consider baseline state versus stress conditions (normal versus ischemia/reperfusion)

  • Dose-response relationships:

    • Analyze whether contradictions reflect different levels of AK1 modulation

    • Consider U-shaped or non-linear response curves where both too little and too much AK1 activity may be detrimental

  • Species and strain differences:

    • Compare findings across model organisms (flies, mice, human cells)

    • Consider genetic background effects within species

  • Cell/tissue type variations:

    • Distinguish cell-autonomous from non-cell-autonomous effects

    • Compare findings across different tissues with varying metabolic demands

  • Mechanistic dissection:

    • Test specific hypotheses about mechanisms underlying contradictory findings

    • Use combination approaches (e.g., AK1 modulation plus adenosine receptor blockade)

This systematic approach can resolve apparent contradictions, as illustrated by studies showing that arrhythmias observed in ex vivo AK1-OE hearts may reflect an artifact of adenine nucleotide loss during cannulation rather than a true physiological effect .

What considerations are important when developing biomarkers of AK1 activity for clinical applications?

Development of AK1-related biomarkers for clinical applications should address:

  • Biomarker accessibility:

    • Prioritize markers measurable in accessible biofluids (blood, urine, CSF)

    • Consider surrogate markers that reflect AK1 activity indirectly

  • Specificity and sensitivity:

    • Determine whether markers specifically reflect AK1 activity versus broader purine metabolism changes

    • Establish sensitivity to detect clinically relevant variations in AK1 function

  • Analytical validation:

    • Develop standardized assays with appropriate reference materials

    • Determine assay precision, accuracy, and reproducibility across laboratories

  • Clinical validation:

    • Establish normal ranges in healthy populations

    • Determine marker changes in relevant patient populations

  • Utility assessment:

    • Evaluate marker value for diagnosis, prognosis, or treatment monitoring

    • Determine whether intervention-induced changes correlate with clinical outcomes

Research has begun to characterize changes in purine metabolism in Parkinson's disease patients, which could lead to biomarker development for dysfunctional adenosine metabolism that might guide therapeutic interventions targeting the AK1 pathway .

Product Science Overview

Structure and Function

AK1 is a small enzyme with a molecular weight of approximately 23.7 kDa. It consists of three functional domains:

  1. CORE Domain: The catalytic and oligomerization regulatory enzyme domain.
  2. LID Domain: The ligand interaction domain.
  3. Substrate Binding Site: The site where the substrate binds and the reaction occurs .

The enzyme is predominantly found in the cytosol of skeletal muscle, brain, and erythrocytes. It plays a vital role in regulating the adenine nucleotide composition within these cells .

Genetic Information

The gene encoding AK1 is located on human chromosome 9 at the position 9q34.11. There are nine distinct isoenzymes of adenylate kinase in human tissues, named AK1 to AK9. Each isoenzyme has a specific role and localization within the cell .

Recombinant Expression

Human recombinant AK1 is typically expressed in Escherichia coli and purified using conventional chromatography techniques. The recombinant protein is often fused to a His-tag at the N-terminus to facilitate purification .

Clinical Significance

AK1 is involved in various physiological processes and its dysregulation can lead to several medical conditions. For instance, defects in the AK1 gene are associated with a form of hemolytic anemia. Additionally, low levels of AK1 can cause an imbalance in adenine nucleotide pools, which may contribute to other metabolic disorders .

In the context of neurodegenerative diseases, AK1 is upregulated in the brains of Alzheimer’s disease patients and downregulated in Duchenne muscular dystrophy. It is responsible for abnormal phosphorylation of tau via the AMPK-GSK3β pathway, leading to tau-mediated neurodegeneration .

Research and Applications

Due to its critical role in cellular energy metabolism, AK1 is a target for research in various fields, including cancer, neurodegenerative diseases, and metabolic disorders. Understanding the structure and function of AK1 can aid in the development of new therapeutic strategies for these conditions .

In summary, Adenylate Kinase 1 is a vital enzyme with significant roles in cellular energy homeostasis and various physiological processes. Its recombinant form, expressed in E. coli, provides a valuable tool for research and potential therapeutic applications.

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.