Recombinant Mouse 5'-AMP-activated protein kinase catalytic subunit alpha-2 (Prkaa2)

Shipped with Ice Packs
In Stock

Description

Functional Roles

Prkaa2 regulates metabolic pathways through isoform-specific mechanisms:

Metabolic Regulation

  • Lipid Metabolism: Phosphorylates acetyl-CoA carboxylase (ACC) at Ser79 (ACC1) and Ser212 (ACC2), inhibiting fatty acid synthesis while promoting β-oxidation .

  • Glucose Homeostasis: Activates glucose uptake in skeletal muscle and suppresses gluconeogenesis in the liver .

  • Autophagy: Differentially regulates pro-autophagy (BECN1/PIK3C3) and non-autophagy complexes (PIK3C3 alone) .

Tissue-Specific Functions

  • Retina: Critical for rod photoreceptor metabolism; Prkaa2 deficiency causes structural abnormalities and visual dysfunction linked to dysregulated inosine monophosphate dehydrogenase (IMPDH) .

  • Liver: Controls triglyceride/ketone body balance; liver-specific knockout increases triglycerides and reduces ketones .

  • Heart: Overactivation in Prkag2 cardiomyopathy models leads to glycogen accumulation and hypertrophy .

Table 2: Prkaa2-Knockout Phenotypes in Mice

Tissue/OrganPhenotypeMechanismSource
LiverElevated plasma triglyceridesReduced β-oxidation, ACC activation
RetinaPhotoreceptor outer segment defectsIMPDH hyperactivity, metabolic stress
HeartHypertrophy, glycogen accumulationα2-subunit hyperactivation

Research Applications

Prkaa2 is widely used in biochemical and genetic studies to elucidate AMPK signaling:

Recombinant Production

  • Expression Systems: Produced in E. coli or mammalian cells using vectors like pCMV6-XL4 .

  • Purification: Affinity chromatography (e.g., His-tagged proteins) or immunoprecipitation with isoform-specific antibodies (e.g., AF2850) .

  • Kinase Activity Assays: Measured via phosphorylation of ACC peptides or synthetic substrates .

Genetic Models

  • Tissue-Specific Knockouts: Rod photoreceptor-specific deletion (Prkaa2^rod-/-) reveals isoform-specific roles in vision .

  • Mutant Mice: Acc1 S79A;Acc2 S212A double knock-in mice mimic metabolic dysregulation seen in AMPK dysfunction .

Therapeutic Potential

Prkaa2 modulation offers targets for treating metabolic and degenerative diseases:

Metabolic Disorders

  • Obesity/Diabetes: AMPK activators (e.g., metformin) enhance Prkaa2-mediated suppression of ACC, improving insulin sensitivity .

  • Hepatoblastoma: Prkaa2 inhibition may reduce tumor growth by altering ferroptosis pathways .

Neurological/Retinal Diseases

  • Retinopathy: IMPDH inhibitors (e.g., mycophenolic acid) rescue visual function in Prkaa2-deficient mice .

  • Cardiomyopathy: Dominant-negative α2 subunits (e.g., TGα2 DN) attenuate hypertrophy in Prkag2 models .

Key Research Findings

Study FocusMethodologyOutcomeSource
Retinal DysfunctionRod photoreceptor-specific knockoutImpaired scotopic a/b waves, OS damage
Lipid MetabolismLiver-specific Prkaa2 deletionHypertriglyceridemia, reduced ketones
CardiomyopathyPrkag2 N488I + TGα2 DN miceReversed hypertrophy, glycogen clearance

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional fees.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate 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%, provided as a reference.
Shelf Life
Shelf life depends on several factors: 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. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
If a specific tag type is required, please inform us; we will prioritize its development.
Synonyms
Prkaa25'-AMP-activated protein kinase catalytic subunit alpha-2; AMPK subunit alpha-2; EC 2.7.11.1; Acetyl-CoA carboxylase kinase; ACACA kinase; EC 2.7.11.27; Hydroxymethylglutaryl-CoA reductase kinase; HMGCR kinase; EC 2.7.11.31
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-552
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Target Protein Sequence
MAEKQKHDGR VKIGHYVLGD TLGVGTFGKV KIGEHQLTGH KVAVKILNRQ KIRSLDVVGK IKREIQNLKL FRHPHIIKLY QVISTPTDFF MVMEYVSGGE LFDYICKHGR VEEVEARRLF QQILSAVDYC HRHMVVHRDL KPENVLLDAQ MNAKIADFGL SNMMSDGEFL RTSCGSPNYA APEVISGRLY AGPEVDIWSC GVILYALLCG TLPFDDEHVP TLFKKIRGGV FYIPDYLNRS VATLLMHMLQ VDPLKRATIK DIREHEWFKQ DLPSYLFPED PSYDANVIDD EAVKEVCEKF ECTESEVMNS LYSGDPQDQL AVAYHLIIDN RRIMNQASEF YLASSPPSGS FMDDSAMHIP PGLKPHPERM PPLIADSPKA RCPLDALNTT KPKSLAVKKA KWHLGIRSQS KACDIMAEVY RAMKQLGFEW KVVNAYHLRV RRKNPVTGNY VKMSLQLYLV DSRSYLLDFK SIDDEVVEQR SGSSTPQRSC SAAGLHRARS SFDSSTAENH SLSGSLTGSL TGSTLSSASP RLGSHTMDFF EMCASLITAL AR
Uniprot No.

Target Background

Function
The catalytic subunit of AMP-activated protein kinase (AMPK) is an energy-sensing protein kinase crucial in regulating cellular energy metabolism. In response to reduced intracellular ATP, AMPK activates energy-producing pathways and inhibits energy-consuming processes, including protein, carbohydrate, and lipid biosynthesis, as well as cell growth and proliferation. AMPK achieves this through direct phosphorylation of metabolic enzymes and longer-term effects via phosphorylation of transcriptional regulators. It also regulates cellular polarity by remodeling the actin cytoskeleton, likely through indirect myosin activation. AMPK regulates lipid synthesis by phosphorylating and inactivating enzymes such as ACACA, ACACB, GYS1, HMGCR, and LIPE; it controls fatty acid and cholesterol synthesis via phosphorylation of acetyl-CoA carboxylase (ACACA and ACACB) and hormone-sensitive lipase (LIPE), respectively. It further regulates insulin signaling and glycolysis by phosphorylating IRS1, PFKFB2, and PFKFB3 and participates in insulin receptor (INSR) internalization. AMPK stimulates glucose uptake in muscle by promoting the translocation of the glucose transporter SLC2A4/GLUT4 to the plasma membrane, possibly by phosphorylating TBC1D4/AS160. It regulates transcription and chromatin structure through phosphorylation of transcriptional regulators involved in energy metabolism, including CRTC2/TORC2, FOXO3, histone H2B, HDAC5, MEF2C, MLXIPL/ChREBP, EP300, HNF4A, p53/TP53, SREBF1, SREBF2, and PPARGC1A. AMPK acts as a key regulator of hepatic glucose homeostasis, phosphorylating CRTC2/TORC2, leading to its cytoplasmic sequestration. Under stress, it phosphorylates histone H2B at Ser-36 (H2BS36ph), promoting transcription. As a key regulator of cell growth and proliferation, it phosphorylates TSC2, RPTOR, and ATG1/ULK1; in response to nutrient limitation, it negatively regulates the mTORC1 complex by phosphorylating its component RPTOR and by activating TSC2. Under nutrient limitation, AMPK promotes autophagy by activating ATG1/ULK1, also activating WDR45 in the process. It also regulates circadian rhythm by phosphorylating and destabilizing CRY1. AMPK may regulate the Wnt signaling pathway via phosphorylation and stabilization of CTNNB1. It further phosphorylates CFTR, EEF2K, KLC1, NOS3, and SLC12A1. AMPK plays a significant role in differentially regulating pro-autophagy (PIK3C3, BECN1, PIK3R4, and UVRAG or ATG14) and non-autophagy (PIK3C3, BECN1, and PIK3R4) complexes in response to glucose starvation. It can inhibit the non-autophagy complex by phosphorylating PIK3C3 and activate the pro-autophagy complex by phosphorylating BECN1.
Gene References Into Functions
  1. AMPK mediates muscle atrophy during unloading and regrowth following reloading, without affecting myosin heavy chain isoform transitions. PMID: 30262782
  2. This research provides insights into AMPKα2-dependent regulation of GLUT4 transcription post-exercise. PMID: 28688716
  3. Data suggest AMPK acts as an intermediary in endocannabinoid-mediated exercise-induced antinociception. PMID: 28479394
  4. The rs2746342 polymorphism is significantly associated with type 2 diabetes mellitus (T2DM) susceptibility and interacts with rs2143754 in modulating fasting plasma glucose (FPG) in Han Chinese populations. PMID: 27427333
  5. AMPKα2 activation prevents cardiac hypertrophy primarily by inhibiting O-GlcNAcylation. PMID: 29371602
  6. AMPK is required for exercise-induced mitophagy in muscle. PMID: 28916822
  7. This mechanism explains how CDK4 promotes anabolism by inhibiting AMPK-activated catabolic processes (FAO). PMID: 29053957
  8. High AMPKα2 phosphorylation is associated with abdominal aortic aneurysm. PMID: 28179583
  9. TNF-α treatment of colonic ρ0 cells increased IL-8 expression ninefold (P < 0.01) via NF-κB compared to controls. Reduced mitochondrial function facilitated TNF-α-mediated NF-κB luciferase promoter activity due to decreased inhibitory IκBα, leading to elevated NF-κB. ... PMID: 28183804
  10. Rac1 and AMPK together account for nearly all ex vivo contraction response in muscle glucose transport; only Rac1, not α2 AMPK, regulates muscle glucose uptake during submaximal exercise in vivo. PMID: 28389470
  11. Using the Cre-loxP system to knockdown AMPKα2 in renal epithelial cells, combined with systemic AMPKα1 deletion, reduced renal AMPK activity, leading to a moderate water- and salt-wasting phenotype, confirming AMPK's physiological role in the kidney. PMID: 28179232
  12. AMPK phosphorylation of cortactin, followed by SIRT1 deacetylation, modulates their interaction with cortical-actin in response to shear stress. This AMPK/SIRT1-regulated cortactin-F-actin dynamics is necessary for endothelial nitric oxide synthase subcellular translocation/activation and is atheroprotective. PMID: 27758765
  13. Decreased muscle AMPK activity leads to reduced voluntary activity, unrelated to dopamine levels in the ventral striatum or cocaine sensitivity. PMID: 27306083
  14. Hypoxia reduces HNF4α/MODY1 protein expression in pancreatic β-cells by activating AMPK. PMID: 28364040
  15. CYP2J2 and epoxyeicosatrienoic acids enhance Akt1 nuclear translocation through interaction with AMPKα2β2γ1, protecting against cardiac hypertrophy. PMID: 27416746
  16. Loss of AMPKα2 impairs control of vesicular stomatitis virus infection. PMID: 27879319
  17. AMPK-mediated HSPB1 expression enhances skeletal muscle insulin sensitivity. PMID: 27928820
  18. AMPKα2 activation contributes to denervation-induced atrophy. PMID: 27136709
  19. AMPKα2 activation in neutrophils is crucial for initiating vascular repair post-ischemia. PMID: 27777247
  20. AMPKα2 deletion induces vascular smooth muscle cell phenotype switching and promotes atherosclerotic plaque features. PMID: 27439892
  21. E2 treatment ameliorates estrogen deficiency-induced changes in cardiac contractile function, possibly through an AMPK-dependent mechanism. PMID: 25448287
  22. AMPKα2 deletion and high-fat feeding do not additively affect heart function or myocardial ischemic tolerance. PMID: 26596312
  23. AMPKα2 isoform plays a key role in antioxidant stress and anti-senescence. PMID: 26718972
  24. AMPKα2 is involved in post-transcriptional, not transcriptional, regulation of PDK4 in muscle. PMID: 26359931
  25. PKD1 phosphorylates AMPKα2 at Ser485/491, decreasing AMPK activity. PMID: 26797128
  26. Activating AMPK in the CNS attenuates inflammatory responses and acute lung injury in sepsis. PMID: 26252187
  27. AMPKα signaling suppresses EMT and chemokine secretion in renal tubular epithelia via interaction with CK2β, attenuating renal injury. PMID: 26108355
  28. AMPK signaling activation is sufficient but not required for exercise-induced mitochondrial FAT/CD36 accumulation. PMID: 25965390
  29. The increase in nuclear PPARα protein after four weeks of intermittent hypoxia exercise training depends on AMPKα2 activation. PMID: 25923694
  30. Low AMPK-α2 signaling disrupts exercise training-induced adaptations. PMID: 25103967
  31. High CO2 activates skeletal muscle atrophy via AMPKα2-FoxO3a-MuRF1, relevant to lung disease and hypercapnia. PMID: 25691571
  32. AMPKα2 is an essential mediator of nicotine-induced whole-body insulin resistance despite reduced adiposity. PMID: 25799226
  33. AMPKα2 ablation in muscle did not worsen high-fat diet-induced obesity; instead, it improved glucose tolerance, insulin sensitivity, and reduced skeletal muscle triglyceride content. PMID: 25637528
  34. AMPKα2 translocates to the nucleus via phosphorylation, interacts with and phosphorylates hnRNP H, modulating metformin-mediated glucose uptake. PMID: 24686086
  35. AMPKα2 is crucial for maintaining skeletal muscle function, integrity, and compensation for chronic metabolic stress from mitochondrial uncoupling. PMID: 24732703
  36. Dietary wolfberry increased xanthophyll concentrations, enhanced BCO2 and HSP60 expression, activated AMPKα2, potentiated mitophagy and mitochondrial biogenesis, and enhanced hepatic lipid oxidation and secretion in mice. PMID: 24449471
  37. Leptin regulates cardiomyocyte contraction (or dysfunction) via Prkaa2 phosphorylation/activation and autophagy. PMID: 23688013
  38. AMPKα2 might be a therapeutic target for colon cancer metastasis-induced liver injury. PMID: 24515110
  39. AMPKα2 contributes to doxorubicin-mediated E2F1 stabilization and activation, forming a positive feedback loop. PMID: 24398673
  40. AMPKα2 deletion causes aberrant VSMC migration with accelerated neointima formation through Skp2 upregulation and E-cadherin downregulation. PMID: 24115035
  41. Ageing and high-fat diet do not worsen insulin action in mice lacking functional muscle α2AMPK. PMID: 23671593
  42. AMPK is the first identified physiological substrate for CHIP chaperone activity, linking cardiac proteolytic and metabolic pathways. PMID: 23863712
  43. A functional AMPKα2 subunit is required for insulin-stimulated muscle glycogen synthesis. PMID: 23224579
  44. AMPKα(2) regulates substrate uptake in a time-dependent manner in contracting muscle but is not essential for fatty acid uptake and oxidation during caffeine treatment. PMID: 21551008
  45. AMPKα2 controls cardiac p70S6K under normoxia and regulates eEF-2, but not the mTOR-p70S6K pathway, during ischemia. PMID: 23466593
  46. LKB1 acts independently of AMPKa2 in regulating skeletal muscle free fatty acid oxidation during exercise; LKB1 knockdown decreases muscle AMPKa2. PMID: 23349504
  47. AMPK suppression or knockdown produces opposite effects. This demonstrates an anti-inflammatory pathway linking AMPK, PARP-1, and Bcl-6 in endothelial cells. PMID: 23382195
  48. The microtubule cytoskeleton is a sensitive target of AMPK activity; AMPK limits microtubule accumulation and densification in response to hypertrophic stress. PMID: 23316058
  49. Endoplasmic reticulum stress increases vascular smooth muscle contractility, resulting in hypertension; AMPK activation mitigates hypertension by suppressing ER stress in vivo. PMID: 23288166
  50. p70S6 kinase forms a complex with AMPK's α-2 subunit, resulting in phosphorylation at serine(491). PMID: 22727014
Database Links
Protein Families
Protein kinase superfamily, CAMK Ser/Thr protein kinase family, SNF1 subfamily
Subcellular Location
Cytoplasm. Nucleus. Note=In response to stress, recruited by p53/TP53 to specific promoters.

Q&A

What is the basic structure and function of recombinant mouse PRKAA2?

Recombinant mouse PRKAA2 (5'-AMP-activated protein kinase catalytic subunit alpha-2) is one of two isoforms of the catalytic α subunit of AMPK, a heterotrimeric protein consisting of α, β, and γ subunits. The protein functions as an energy sensor that maintains cellular energy homeostasis by responding to changes in AMP:ATP and ADP:ATP ratios. PRKAA2 possesses serine/threonine kinase activity and becomes fully activated when phosphorylated at Thr172. When expressed recombinantly, the protein is typically produced in systems such as baculovirus-infected Sf9 cells to maintain proper folding and post-translational modifications. The full-length mouse PRKAA2 consists of 330 amino acids and contains several functional domains including a kinase domain and regulatory regions that mediate interactions with β and γ subunits, as well as with upstream kinases and downstream substrates .

How does PRKAA2 expression differ across mouse tissues compared to human tissues?

PRKAA2 exhibits distinct tissue-specific expression patterns that differ between mice and humans. In mice, PRKAA2 (α2 isoform) shows preferential expression in neuronal tissues including the retina and brain, where both α1 and α2 isoforms are expressed but with a bias toward α2. This is particularly evident in rod photoreceptors, which serve as an excellent model for studying isoform-specific functions. Conversely, in non-neuronal tissues such as immune cells and hepatocytes, there is often a skewed expression profile with the α1 isoform (PRKAA1) being predominant. Human tissues show similar patterns in neuronal cells, but human peripheral blood mononuclear cells (PBMCs) demonstrate near-complete restriction to PRKAA1 expression with minimal PRKAA2. This differential expression pattern suggests evolutionary divergence in AMPK isoform utilization across species and highlights the importance of considering species differences when translating mouse-based PRKAA2 research to human applications .

What are the major activation mechanisms of recombinant PRKAA2?

Recombinant PRKAA2 is activated through several distinct but interconnected mechanisms:

  • Phosphorylation at Thr172: The primary activation mechanism involves phosphorylation of Thr172 in the activation loop by upstream kinases, predominantly the LKB1 complex (consisting of LKB1, STRAD, and MO25) or calcium/calmodulin-dependent protein kinase kinase β (CaMKKβ).

  • Nucleotide binding and allosteric regulation: Binding of AMP or ADP to the γ subunit of the AMPK complex containing PRKAA2 promotes activation via three mechanisms:

    • Promotion of Thr172 phosphorylation

    • Inhibition of Thr172 dephosphorylation

    • Allosteric activation of already phosphorylated PRKAA2 (specific to AMP)

  • Calcium-dependent activation: In neuronal tissues where PRKAA2 is highly expressed, elevation in cytosolic calcium can activate PRKAA2 via CaMKKβ, independent of changes in adenine nucleotide ratios.

These mechanisms allow PRKAA2 to respond to a wide range of cellular stresses, including glucose deprivation, hypoxia, ischemia, and increased energy demands. When all activation mechanisms operate simultaneously under severe metabolic stress, PRKAA2 can achieve >2000-fold activation over its basal state .

What are the optimal expression systems for producing active recombinant mouse PRKAA2?

For producing functionally active recombinant mouse PRKAA2, the baculovirus-infected Sf9 insect cell system has proven most effective for several reasons:

  • Complex formation capability: To achieve full functionality, PRKAA2 must form a heterotrimeric complex with appropriate β and γ subunits. The baculovirus-Sf9 system allows co-expression of multiple subunits (PRKAA2, PRKAB2, PRKAG1) to generate the complete active complex with >85% purity.

  • Post-translational modifications: This system supports proper folding and critical post-translational modifications, particularly phosphorylation at Thr172 which is essential for catalytic activity.

  • Protein yield and solubility: Sf9 cells provide superior yields of soluble, correctly folded PRKAA2 compared to bacterial expression systems, which often produce misfolded or insoluble protein.

  • Functional validation: When expressed in this system, recombinant PRKAA2 demonstrates expected kinase activity toward canonical substrates like acetyl-CoA carboxylase (ACACA) and hydroxymethylglutaryl-CoA reductase (HMGCR).

For experimental applications requiring highly purified material, the recombinant protein should be purified using affinity chromatography followed by size-exclusion chromatography to ensure homogeneity and removal of insect cell contaminants .

How can researchers accurately measure PRKAA2 activation in experimental systems?

Accurate measurement of PRKAA2 activation requires a multi-parameter approach that addresses both phosphorylation status and functional activity:

  • Phosphorylation status assessment:

    • Western blotting: Using phospho-specific antibodies against Thr172 normalized to total PRKAA2 protein levels.

    • Mass spectrometry: For precise quantification of phosphorylation stoichiometry at Thr172 and detection of other regulatory modifications.

  • Kinase activity assays:

    • In vitro kinase assays: Using purified recombinant PRKAA2 with synthetic AMPK substrates (SAMS peptide) or physiological substrates (ACC, HMGCR).

    • Cellular target phosphorylation: Monitoring phosphorylation of downstream targets such as ACC (Ser79) as a proxy for PRKAA2 activity in cellular contexts.

  • Metabolic readouts:

    • LC-MS/MS metabolomics: Quantifying changes in key metabolites regulated by PRKAA2 activity, including ATP, GTP, and cGMP levels as demonstrated in PRKAA2-deficient mouse models.

    • Metabolic flux analysis: Measuring rates of glucose uptake, fatty acid oxidation, or protein synthesis, all processes regulated by AMPK.

  • AMP/ADP/ATP ratio determination:

    • Using HPLC or LC-MS techniques to accurately measure adenine nucleotide ratios, which correlate with AMPK activation status.

For distinguishing PRKAA2-specific activation from PRKAA1, research protocols should include appropriate controls such as isoform-specific knockouts or selective inhibitors to isolate isoform-specific contributions to the observed phenotypes .

What are the key considerations for designing PRKAA2 knockout or knockdown experiments?

Designing effective PRKAA2 knockout or knockdown experiments requires careful consideration of several factors:

  • Isoform specificity and compensation:

    • PRKAA1 may compensate for PRKAA2 deficiency in certain tissues, potentially masking phenotypes.

    • Design experiments that compare single knockouts (PRKAA2-only) with double knockouts (PRKAA1/PRKAA2) to distinguish isoform-specific functions from redundant roles.

  • Tissue/cell type selection:

    • Target tissues with high PRKAA2:PRKAA1 expression ratios (e.g., neuronal tissues, retinal photoreceptors) for clearer phenotypes.

    • For tissues with balanced expression of both isoforms, conditional tissue-specific knockout approaches may be more informative than global knockouts.

  • Temporal considerations:

    • Use inducible knockout systems (e.g., tamoxifen-inducible Cre-loxP) to distinguish developmental versus adult functions of PRKAA2.

    • Consider that acute knockdown (e.g., via siRNA) may yield different results from chronic knockout due to adaptive responses.

  • Validation strategies:

    • Confirm knockout at protein level (not just mRNA) as post-transcriptional regulation can maintain protein expression.

    • Verify functional loss by measuring phosphorylation of PRKAA2-specific substrates.

    • Include rescue experiments with wild-type PRKAA2 to confirm phenotype specificity.

  • Physiological readouts:

    • Include comprehensive metabolic profiling (ATP, GTP, cGMP levels) as PRKAA2 deficiency leads to significant alterations in energy nucleotides.

    • Assess tissue-specific functional outcomes (e.g., electrophysiological measurements in neurons, contractility in muscle cells).

A successful example of this approach is seen in studies of rod photoreceptor-specific PRKAA2 knockout mice (Prkaa2^-Rhod/-Rhod), which revealed isoform-specific regulation of visual function and metabolism without significant effects on photoreceptor survival .

How do the functions of PRKAA2 differ from PRKAA1 in neuronal tissues?

PRKAA2 exhibits distinct functions from PRKAA1 in neuronal tissues, particularly in energy metabolism regulation and neuronal signaling:

  • Metabolic regulation:

    • PRKAA2, but not PRKAA1, regulates key nucleotide levels in photoreceptors, with PRKAA2-deficient retinas showing significantly increased ATP (1.723-fold) and GTP (1.927-fold) levels, and trending increases in cGMP (1.42-fold).

    • These metabolic changes occur without corresponding alterations in AMP, ADP, GMP, GDP, or IMP levels, suggesting PRKAA2-specific pathways for high-energy phosphate regulation.

  • Functional impacts:

    • PRKAA2-deficient photoreceptors display functional deficits on electroretinography and structural abnormalities in outer segments detected by transmission electron microscopy.

    • These changes represent neuronal dysfunction rather than neurodegeneration, indicating PRKAA2's role in maintaining normal neuronal function.

  • Expression patterns:

    • While both isoforms are expressed in neuronal tissues, quantitative analysis shows a preference for PRKAA2 in mouse and human retinal cell types.

    • In contrast, non-neuronal and immune cells show strong preference for PRKAA1 with minimal PRKAA2 expression.

  • Substrate specificity:

    • PRKAA2 appears to have unique substrate preferences in neuronal contexts, regulating pathways not controlled by PRKAA1 despite their structural similarity.

    • This functional specialization likely arises from differences in subcellular localization, binding partners, or subtle variations in substrate recognition motifs.

These findings establish neuronal tissues, particularly photoreceptors, as excellent models for studying PRKAA2-specific functions and suggest that PRKAA2-targeted approaches may be particularly relevant for neurological and neurosensory disorders .

What post-translational modifications regulate PRKAA2 activity beyond phosphorylation?

Beyond the well-established regulation by phosphorylation at Thr172, PRKAA2 activity is modulated by several other post-translational modifications that create additional layers of regulatory control:

  • Succinylation:

    • Recent research has identified succinylation as a critical modification of PRKAA2, particularly at lysine residues K69 and K260.

    • Elevated PRKAA2 succinylation levels have been observed in placental tissues of patients with hypertensive disorder complicating pregnancy (HDCP).

    • Sirtuin 5 (SIRT5) interacts with PRKAA2 to induce desuccinylation at these specific lysine residues, modulating PRKAA2 activity.

    • This SIRT5-mediated desuccinylation appears to regulate placental cell apoptosis, suggesting a role in pregnancy-related disorders.

  • Ubiquitination:

    • Ubiquitination can target PRKAA2 for proteasomal degradation, affecting its protein stability and abundance.

    • Various cellular stresses can alter the ubiquitination status of PRKAA2, providing another mechanism for regulating its activity in response to environmental changes.

  • Myristoylation and palmitoylation:

    • While these modifications primarily affect the β subunits of AMPK complexes, they indirectly influence PRKAA2 activity by regulating subcellular localization and membrane association of the entire complex.

  • Acetylation:

    • Acetylation of specific lysine residues can modulate PRKAA2 activity by affecting its interaction with regulatory proteins or altering its conformation.

These diverse post-translational modifications create a complex regulatory network that fine-tunes PRKAA2 activity in response to various cellular signals and metabolic states. The succinylation pathway, in particular, represents a novel therapeutic target for conditions like HDCP where aberrant PRKAA2 activity contributes to pathology .

How does PRKAA2 contribute to tissue-specific metabolic regulation in physiological and pathological states?

PRKAA2 exhibits remarkable tissue-specific functions in metabolic regulation across both normal physiological conditions and disease states:

  • Neuronal tissues:

    • In retinal photoreceptors, PRKAA2 regulates energy nucleotide homeostasis (ATP, GTP, cGMP), with its deficiency leading to increased nucleotide levels and functional visual deficits.

    • PRKAA2-specific pathways appear critical for maintaining proper neural function without affecting cell survival, suggesting specialized roles in neuronal energy management.

    • The preference for PRKAA2 expression in neuronal cells across multiple brain regions suggests conserved neuronal functions beyond the retina.

  • Placental tissue and pregnancy disorders:

    • PRKAA2 expression is enhanced in placental tissues of patients with hypertensive disorder complicating pregnancy (HDCP).

    • Experimental evidence shows that PRKAA2 overexpression accelerates primary placental cell apoptosis, while its knockdown attenuates cell death.

    • SIRT5-mediated PRKAA2 succinylation modulates this process, with animal studies confirming that PRKAA2 elevates systolic blood pressure in HDCP rat models.

    • This suggests PRKAA2 as a potential therapeutic target in pregnancy-related hypertensive disorders.

  • Metabolic sensors and effectors:

    • PRKAA2 responds to cellular energy status by sensing AMP:ATP and ADP:ATP ratios, activating in response to metabolic stresses like glucose deprivation, hypoxia, or increased energy demands.

    • Upon activation, PRKAA2 phosphorylates key metabolic enzymes including acetyl-CoA carboxylase (ACACA), hormone-sensitive lipase (LIPE), and hydroxymethylglutaryl-CoA reductase (HMGCR).

    • This phosphorylation inhibits energy-consuming anabolic processes (lipid and protein synthesis) while promoting catabolic ATP-generating pathways.

  • Insulin signaling and glucose homeostasis:

    • PRKAA2 regulates insulin signaling by phosphorylating insulin receptor substrate 1 (IRS1) and key glycolytic enzymes like phosphofructokinase 2 (PFK2).

    • This creates a complex relationship with insulin sensitivity that varies across tissues and metabolic states.

The tissue-specific functions of PRKAA2 make it a promising target for treating metabolic disorders, with the potential for developing therapies that selectively modulate its activity in specific tissues to minimize off-target effects .

What are the current challenges in developing isoform-specific activators or inhibitors of PRKAA2?

Developing truly isoform-specific modulators of PRKAA2 faces several significant challenges that researchers must address:

  • Structural similarity between isoforms:

    • PRKAA1 and PRKAA2 share high sequence homology (>75%) in their catalytic domains, making selective targeting challenging.

    • The ATP-binding pockets, which are common targets for kinase inhibitors, are particularly well-conserved between the isoforms.

    • Researchers must identify and exploit subtle structural differences in less conserved regions or regulatory domains.

  • Complex formation considerations:

    • PRKAA2 functions as part of heterotrimeric complexes with β and γ subunits, which influence its conformation and activity.

    • The composition of these complexes varies across tissues, creating different potential binding interfaces.

    • Effective modulators must account for these complex interactions rather than targeting the isolated catalytic subunit.

  • Validation challenges:

    • Confirming isoform specificity requires testing in systems with knockout or knockdown of individual isoforms.

    • Appropriate experimental models must express physiologically relevant levels of both isoforms to accurately assess selectivity.

    • Researchers should employ both in vitro kinase assays and cell-based functional assays with isoform-specific readouts.

  • Tissue penetration and distribution:

    • Given the differential expression of PRKAA2 across tissues (higher in neuronal tissues), compounds must achieve appropriate tissue distribution.

    • For neuronal applications, blood-brain barrier penetration becomes a critical consideration.

    • Targeted delivery approaches may be necessary to achieve tissue-specific modulation.

  • Allosteric vs. catalytic site targeting:

    • Allosteric modulators that bind outside the conserved catalytic site may offer greater isoform selectivity.

    • Researchers should explore compounds that influence interactions with isoform-specific binding partners or regulatory proteins.

    • Screening approaches should incorporate assays that can detect allosteric modulation distinct from direct catalytic inhibition.

Current research directions include the development of small molecules that exploit unique post-translational modification sites (such as the succinylation sites at K69 and K260) or that selectively influence PRKAA2's interaction with specific upstream regulators or downstream substrates .

How can researchers effectively study PRKAA2 in the context of neurodegenerative diseases?

Studying PRKAA2 in neurodegenerative disease contexts requires specialized approaches that address the unique features of neuronal tissues and disease processes:

  • Model selection and development:

    • Cell models: Use primary neurons or iPSC-derived neurons with confirmed PRKAA2 expression rather than standard cell lines.

    • Animal models: Employ tissue-specific conditional knockouts (e.g., Prkaa2^-Rhod/-Rhod for photoreceptor studies) to isolate neuronal effects.

    • Human samples: Analyze post-mortem brain tissue or neuronal organoids from patients with neurodegenerative conditions to assess PRKAA2 expression and modification patterns.

  • Functional assessment techniques:

    • Electrophysiology: Measure neuronal activity to detect functional deficits that precede structural changes, as seen in PRKAA2-deficient retinal cells.

    • Advanced imaging: Use transmission electron microscopy and super-resolution microscopy to detect subtle structural changes in neurons, particularly in subcellular compartments.

    • Metabolic profiling: Apply LC-MS/MS metabolomics to quantify changes in nucleotide levels (ATP, GTP, cGMP) and other metabolites regulated by PRKAA2.

  • Disease-specific considerations:

    • Protein aggregation: Investigate PRKAA2's role in regulating autophagy and protein degradation pathways relevant to diseases like Alzheimer's and Parkinson's.

    • Mitochondrial dysfunction: Examine how PRKAA2 influences mitochondrial biogenesis and function, which are often compromised in neurodegenerative conditions.

    • Neuroinflammation: Assess PRKAA2's impact on neuroinflammatory processes that contribute to disease progression.

  • Intervention strategies for testing:

    • Genetic approaches: Use AAV-mediated gene delivery to rescue or modulate PRKAA2 expression in specific neuronal populations.

    • Pharmacological approaches: Test PRKAA2 activators in disease models, particularly those that can cross the blood-brain barrier.

    • Metabolic interventions: Evaluate dietary or exercise interventions that activate AMPK pathways for neuroprotective effects.

  • Translational considerations:

    • Validate findings across multiple models, from cells to animal models to human samples.

    • Consider sex differences in PRKAA2 function and response to interventions.

    • Develop biomarkers of PRKAA2 activity that can be measured non-invasively for clinical translation.

The rod photoreceptor system provides an excellent model for studying neuronal PRKAA2 function due to the high expression of both isoforms and the accessibility of functional readouts (electroretinography), making it valuable for initial screening of neuroprotective strategies .

What techniques are most effective for studying PRKAA2 substrate specificity and identifying novel substrates?

Elucidating PRKAA2 substrate specificity and identifying novel substrates requires a multi-faceted approach combining biochemical, proteomic, and computational techniques:

  • In vitro kinase assays with substrate profiling:

    • Peptide library screening: Use positional scanning peptide libraries containing the AMPK consensus motif (ΦX(B,X)XX(S/T)XXXΦ, where Φ is a hydrophobic residue and B is a basic residue) to identify sequence preferences specific to PRKAA2.

    • Protein microarrays: Screen purified recombinant PRKAA2 against arrays containing thousands of potential substrate proteins to identify direct interactions.

    • Comparative analysis: Directly compare PRKAA1 and PRKAA2 substrate preferences using identical experimental conditions to identify isoform-specific targets.

  • Phosphoproteomic approaches:

    • Quantitative phosphoproteomics: Compare phosphorylation patterns in wild-type versus PRKAA2-knockout cells/tissues using stable isotope labeling (SILAC) or tandem mass tag (TMT) labeling.

    • Proximity-based phosphoproteomics: Combine BioID or APEX2 proximity labeling with phosphoproteomics to identify substrates in specific subcellular compartments.

    • Substrate-trapping mutants: Use catalytically inactive PRKAA2 mutants that can bind but not phosphorylate substrates to trap interacting proteins.

  • Genetic and pharmacological validation:

    • Genetic approaches: Confirm putative substrates using PRKAA2-specific knockout or knockdown models, with rescue by wild-type but not kinase-dead PRKAA2.

    • Phosphosite mutants: Generate non-phosphorylatable mutants (S/T to A) of candidate substrates to confirm functional significance.

    • Chemical genetics: Use analog-sensitive PRKAA2 mutants that accept bulky ATP analogs to selectively label direct substrates.

  • Bioinformatic integration:

    • Motif analysis: Apply machine learning algorithms to identify refined PRKAA2-specific phosphorylation motifs from experimentally validated substrates.

    • Network analysis: Integrate phosphoproteomic data with protein-protein interaction networks to identify substrate clusters and pathways.

    • Evolutionary conservation: Analyze conservation of putative phosphosites across species to prioritize functionally significant targets.

  • Physiological validation:

    • Tissue-specific analysis: Compare substrate profiles across tissues with different PRKAA1:PRKAA2 ratios to identify context-dependent targets.

    • Metabolic stress responses: Analyze how acute versus chronic metabolic stresses affect PRKAA2 substrate selection.

    • Post-translational modification interplay: Investigate how PRKAA2 succinylation status affects its substrate specificity.

This comprehensive approach has successfully identified tissue-specific PRKAA2 substrates, such as those involved in placental cell apoptosis regulation in the context of hypertensive disorders of pregnancy .

How does PRKAA2 contribute to hypertensive disorders of pregnancy, and what therapeutic approaches are being explored?

PRKAA2 plays a significant role in hypertensive disorders of pregnancy (HDCP), with recent research illuminating its mechanisms and therapeutic potential:

  • Altered expression and modification in HDCP:

    • PRKAA2 expression is significantly enhanced in placental tissues of HDCP patients compared to normal controls.

    • The level of PRKAA2 succinylation is elevated in placental tissue from HDCP patients, suggesting dysregulation of this post-translational modification pathway.

    • These alterations correlate with clinical features of HDCP, which affects 5-10% of pregnancies and presents with hypertension, edema, proteinuria, and potential organ damage.

  • Cellular mechanisms in placental pathology:

    • PRKAA2 overexpression accelerates primary placental cell apoptosis, a key pathological feature of HDCP.

    • Conversely, PRKAA2 knockdown attenuates placental cell apoptosis, supporting its causal role in disease pathogenesis.

    • Sirtuin 5 (SIRT5) interacts with PRKAA2 at specific lysine residues (K69 and K260) to induce desuccinylation, regulating its activity in placental cells.

  • Animal model validation:

    • In HDCP rat models, PRKAA2 elevation leads to increased systolic blood pressure, recapitulating a key clinical feature of the human condition.

    • This provides in vivo validation of PRKAA2's pathogenic role and establishes a preclinical model for testing therapeutic interventions.

  • Emerging therapeutic approaches:

    • SIRT5 activators: Compounds that enhance SIRT5 activity may promote PRKAA2 desuccinylation, potentially normalizing its function in HDCP.

    • PRKAA2 inhibitors: Selective inhibitors of PRKAA2 could reduce placental cell apoptosis and improve outcomes in HDCP.

    • Succinylation modulators: Targeting the enzymes that regulate protein succinylation/desuccinylation more broadly may offer therapeutic benefits.

    • Combined approaches: Strategies that normalize both PRKAA2 expression levels and its post-translational modification status may provide synergistic benefits.

  • Clinical translation considerations:

    • Biomarker development: PRKAA2 expression and succinylation levels in placental tissue or circulating microvesicles could serve as diagnostic or prognostic markers.

    • Timing of intervention: Early intervention before clinical manifestation of HDCP may be crucial for efficacy.

    • Safety in pregnancy: Any therapeutic approach must consider the unique safety requirements of medications used during pregnancy.

These findings establish PRKAA2 as a promising therapeutic target for HDCP, a condition with significant maternal and fetal morbidity and mortality that currently lacks specific treatments beyond blood pressure management and timely delivery .

What is the current understanding of PRKAA2's role in metabolic disorders and potential for therapeutic targeting?

PRKAA2's distinct functions in metabolic regulation position it as a promising target for metabolic disorders:

  • Tissue-specific metabolic regulation:

    • PRKAA2 regulates energy metabolism through phosphorylation of key enzymes involved in lipid synthesis, including acetyl-CoA carboxylase (ACACA and ACACB) and hydroxymethylglutaryl-CoA reductase (HMGCR).

    • It influences glucose metabolism by phosphorylating insulin receptor substrate 1 (IRS1) and enzymes like phosphofructokinase (PFK).

    • In neuronal tissues, PRKAA2 specifically regulates levels of high-energy phosphates (ATP, GTP) and signaling molecules (cGMP).

  • Metabolic disorder relevance:

    • Diabetes and insulin resistance: PRKAA2 activation generally improves insulin sensitivity, suggesting potential benefits in type 2 diabetes.

    • Fatty liver disease: By inhibiting lipid synthesis and promoting fatty acid oxidation, PRKAA2 activation may reduce hepatic steatosis.

    • Neurometabolic disorders: Given its role in neuronal energy homeostasis, PRKAA2 modulation may benefit conditions with altered brain metabolism.

  • Comparative data on PRKAA1 vs. PRKAA2 targeting:

    Tissue TypePRKAA1:PRKAA2 RatioMetabolic Effect of ActivationTargeting Priority
    NeuronalLow (PRKAA2 dominant)Regulates ATP/GTP/cGMPPRKAA2-selective
    HepaticHigh (PRKAA1 dominant)Inhibits lipogenesisPRKAA1-selective
    MuscleBalancedIncreases glucose uptakeDual targeting
    AdiposeHigh (PRKAA1 dominant)Reduces lipogenesisPRKAA1-selective
    PancreaticBalancedRegulates insulin secretionContext-dependent
  • Current therapeutic approaches:

    • Direct activators: Compounds that directly bind and activate AMPK complexes containing PRKAA2.

    • Indirect activators: Agents that increase AMP:ATP ratio, such as metformin, thereby activating AMPK through canonical energy-sensing mechanisms.

    • Metabolic modulators: Interventions like exercise or caloric restriction that physiologically activate AMPK pathways.

  • Challenges and future directions:

    • Isoform selectivity: Developing compounds that selectively target PRKAA2 over PRKAA1 to minimize off-target effects.

    • Tissue specificity: Creating delivery strategies that target specific tissues with metabolic dysfunction.

    • Context-dependent activation: Understanding when PRKAA2 activation versus inhibition is beneficial in different metabolic states and diseases.

    • Combination approaches: Identifying synergistic combinations with other metabolic regulators for maximal therapeutic benefit.

The tissue-specific expression patterns and functions of PRKAA2 suggest that selectively targeting this isoform may offer advantages for treating neurometabolic disorders while minimizing effects in tissues where PRKAA1 predominates .

How can researchers design experiments to evaluate PRKAA2 as a therapeutic target in neurological conditions?

Designing robust experiments to evaluate PRKAA2 as a therapeutic target in neurological conditions requires comprehensive approaches that address both mechanistic understanding and translational potential:

  • Target validation studies:

    • Expression profiling: Quantify PRKAA2:PRKAA1 ratios across brain regions and cell types relevant to specific neurological conditions using single-cell RNA-seq and protein quantification.

    • Human tissue analysis: Compare PRKAA2 expression, phosphorylation, and succinylation patterns in post-mortem brain samples from patients versus controls.

    • Genetic association: Analyze PRKAA2 variants in patient cohorts to identify potential associations with disease risk or progression.

  • Mechanistic investigation:

    • Conditional knockout models: Generate neuron-specific PRKAA2 knockouts in disease models (e.g., Alzheimer's, Parkinson's, stroke) to determine effects on disease progression.

    • Substrate identification: Use phosphoproteomic approaches to identify neuronal substrates specifically regulated by PRKAA2 but not PRKAA1.

    • Metabolic analysis: Perform comprehensive metabolic profiling of PRKAA2-deficient neuronal tissues to identify alterations in energy nucleotides and neurotransmitter precursors.

  • Therapeutic intervention design:

    • Compound screening strategy:

      Screening PhaseAssay TypeEndpoint MeasurementSelection Criteria
      Primary ScreenIn vitro kinasePRKAA2 activation>5x selectivity for PRKAA2 vs. PRKAA1
      Secondary ScreenNeuronal cell cultureTarget engagement, viabilityActivity at ≤1 μM, >80% viability
      Tertiary ScreenBrain slice modelsElectrophysiological functionPreservation/restoration of normal activity
      In vivo TestingDisease modelsBehavior, histopathologyDose-dependent improvement in outcomes
    • Delivery optimization: Develop strategies for brain-targeted delivery, such as nanoparticle formulations or prodrug approaches to enhance BBB penetration.

    • Therapeutic window determination: Establish dose-response relationships and identify the therapeutic window between efficacy and potential side effects.

  • Functional outcome assessment:

    • Electrophysiology: Measure neuronal activity patterns using multi-electrode arrays or patch-clamp techniques.

    • Advanced imaging: Use two-photon microscopy to assess dendritic spine dynamics and synaptic function in vivo.

    • Behavioral testing: Apply comprehensive behavioral test batteries relevant to the neurological condition being studied.

    • Biomarker monitoring: Develop and validate fluid biomarkers (CSF, plasma) that reflect PRKAA2 activity and target engagement.

  • Translational considerations:

    • Pharmacokinetic/pharmacodynamic modeling: Determine brain penetration, target engagement duration, and dosing regimens.

    • Safety assessment: Evaluate potential off-target effects, particularly in tissues with high PRKAA1 expression.

    • Combination approaches: Test PRKAA2 modulators in combination with standard-of-care treatments for potential synergistic effects.

    • Patient stratification strategy: Identify potential biomarkers that could indicate which patients might best respond to PRKAA2-targeted therapy.

This experimental roadmap would provide comprehensive evaluation of PRKAA2 as a therapeutic target, building on the established role of PRKAA2 in neuronal energy regulation and the promising results from photoreceptor-specific studies demonstrating its importance in neuronal function .

What are common pitfalls in PRKAA2 activity assays and how can researchers avoid them?

Researchers commonly encounter several challenges when assessing PRKAA2 activity that can lead to misleading results. Here are the major pitfalls and strategies to avoid them:

  • Isoform cross-reactivity issues:

    • Pitfall: Many commercial antibodies and activity assays fail to distinguish between PRKAA1 and PRKAA2 isoforms.

    • Solution: Validate antibody specificity using appropriate knockout controls; consider using isoform-specific immunoprecipitation before activity assays; when possible, use mass spectrometry-based approaches for definitive isoform identification.

  • Complex integrity problems:

    • Pitfall: PRKAA2 functions as part of a heterotrimeric complex, and dissociation during sample preparation can alter activity measurements.

    • Solution: Use gentle lysis conditions that preserve protein complexes; consider native PAGE or blue native PAGE to verify complex integrity; include detergents that maintain protein-protein interactions.

  • Phosphatase contamination:

    • Pitfall: Endogenous phosphatases can rapidly dephosphorylate Thr172 during sample preparation, leading to underestimation of PRKAA2 activity.

    • Solution: Include multiple phosphatase inhibitors in all buffers; perform sample preparation at 4°C; minimize time between tissue/cell collection and assay.

  • ATP concentration effects:

    • Pitfall: High ATP concentrations in in vitro kinase assays can mask allosteric activation by AMP and lead to underestimation of physiological activity.

    • Solution: Perform assays across a range of ATP concentrations, including those that approximate physiological levels; include AMP in assays to assess allosteric regulation.

  • Non-specific kinase activity:

    • Pitfall: Other kinases may phosphorylate common AMPK substrates (e.g., ACC), confounding measurement of PRKAA2-specific activity.

    • Solution: Include specific AMPK inhibitors (e.g., Compound C) as controls; use PRKAA2 knockout/knockdown controls; employ highly specific AMPK substrates like the SAMS peptide.

  • Post-translational modification effects:

    • Pitfall: PRKAA2 succinylation status affects its activity, but standard activity assays do not account for this modification.

    • Solution: Consider measuring succinylation levels in parallel with activity assays; when studying specific conditions (e.g., HDCP), assess both parameters for proper interpretation.

  • Tissue-specific cofactor requirements:

    • Pitfall: PRKAA2 may require tissue-specific cofactors or binding partners for full activity that are lost during purification.

    • Solution: Compare results from cell-free systems with cellular assays; consider adding back tissue extracts to purified enzyme preparations; use tissue-specific expression systems.

How can researchers accurately distinguish between direct and indirect effects of PRKAA2 modulation in complex biological systems?

Distinguishing direct from indirect effects of PRKAA2 modulation represents a significant challenge in complex biological systems. Researchers can implement several complementary strategies to address this challenge:

  • Temporal analysis approaches:

    • Acute vs. chronic interventions: Compare rapid responses (minutes to hours) following PRKAA2 modulation, which likely represent direct effects, with longer-term responses (days to weeks) that may include compensatory mechanisms.

    • Time-course experiments: Track the sequence of molecular events following PRKAA2 activation or inhibition to establish a temporal hierarchy of responses.

    • Inducible systems: Use rapidly inducible genetic systems (e.g., tet-on/off) to control the timing of PRKAA2 expression changes with precision.

  • Substrate validation techniques:

    • Phosphorylation site mapping: Use mass spectrometry to identify sites directly phosphorylated by PRKAA2 following acute activation.

    • Consensus motif analysis: Confirm that putative direct targets contain the AMPK consensus phosphorylation motif (ΦX(B,X)XX(S/T)XXXΦ).

    • In vitro confirmation: Validate direct phosphorylation using purified components in cell-free kinase assays.

    • Phosphosite mutants: Generate non-phosphorylatable mutants (S/T→A) of putative direct targets to confirm functional significance.

  • Genetic complementation strategies:

    • Kinase-dead controls: Compare effects of wild-type PRKAA2 with catalytically inactive mutants (e.g., K45R) to distinguish kinase-dependent from scaffold-dependent functions.

    • Isoform-specific rescue: Determine whether PRKAA1 can rescue PRKAA2 knockout phenotypes, indicating shared direct targets versus isoform-specific effects.

    • Analog-sensitive approaches: Use PRKAA2 mutants engineered to accept bulky ATP analogs that selectively label direct substrates.

  • Pathway dissection approaches:

    • Combinatorial interventions: Combine PRKAA2 modulation with inhibitors of downstream pathways to isolate direct effects.

    • Epistasis analysis: Determine whether knockout/inhibition of putative downstream targets blocks the effects of PRKAA2 activation.

    • Dynamic pathway modeling: Develop mathematical models incorporating known direct PRKAA2 targets and validate with experimental data.

  • Advanced multi-omics integration:

    • Multi-timepoint proteomics: Combine phosphoproteomics, acetylomics, and succinylomics at multiple timepoints to distinguish primary from secondary modifications.

    • Transcriptomics integration: Compare rapid changes in the phosphoproteome with delayed transcriptional responses to separate direct signaling from gene expression effects.

    • Metabolic flux analysis: Use isotope tracing to determine which metabolic pathways are directly altered by PRKAA2 activation versus those changed through secondary adaptations.

This multi-faceted approach has proven effective in distinguishing direct PRKAA2 effects, such as its immediate regulation of placental cell apoptosis through direct protein interactions and phosphorylation events, from indirect consequences that emerge through transcriptional or metabolic adaptations .

What are the best practices for ensuring reproducibility when working with recombinant PRKAA2 in different experimental systems?

Ensuring reproducibility when working with recombinant PRKAA2 across different experimental systems requires stringent attention to multiple factors that affect protein quality, activity, and consistency:

Implementing these practices has been shown to dramatically improve inter-laboratory reproducibility in studies using recombinant kinases, reducing variation from as high as 100-fold to less than 2-fold in controlled multi-laboratory studies .

What emerging technologies could advance our understanding of PRKAA2 function in complex biological systems?

Several cutting-edge technologies are poised to transform our understanding of PRKAA2 function in complex biological systems:

  • Advanced imaging technologies:

    • Super-resolution microscopy: Techniques like STORM, PALM, and STED can visualize PRKAA2 subcellular localization with nanometer precision, revealing previously undetectable complexes and interactions.

    • Live-cell FRET sensors: Genetically encoded FRET-based sensors for PRKAA2 activity enable real-time, spatiotemporal monitoring of kinase activation in living cells and tissues.

    • Expansion microscopy: This technique physically expands biological specimens, allowing conventional microscopes to resolve structures below the diffraction limit, potentially revealing PRKAA2 nanoclusters and signaling hubs.

    • Label-free imaging: Methods like stimulated Raman scattering microscopy could detect metabolic changes downstream of PRKAA2 activation without introducing potentially perturbative labels.

  • Advanced genetic and genomic tools:

    • CRISPR-based screening: Genome-wide or targeted CRISPR screens in relevant cell types can identify novel regulators and effectors of PRKAA2 signaling.

    • Base editing and prime editing: These technologies allow precise introduction of specific mutations in PRKAA2 or its substrates without double-strand breaks, enabling detailed structure-function studies in endogenous contexts.

    • Single-cell multi-omics: Combined single-cell transcriptomics, proteomics, and metabolomics can reveal cell-specific PRKAA2 functions and heterogeneity within tissues.

    • Spatial transcriptomics and proteomics: These approaches provide spatial context to gene and protein expression patterns, potentially revealing localized PRKAA2 signaling domains within tissues.

  • Protein engineering and chemical biology approaches:

    • Optogenetic and chemogenetic tools: Light-activated or small molecule-activated versions of PRKAA2 enable precise spatiotemporal control of kinase activity.

    • Proximity labeling: BioID or APEX2 fusion proteins can identify proteins in close proximity to PRKAA2 in different subcellular compartments under various conditions.

    • Activity-based protein profiling: Chemical probes that report on PRKAA2 activation state could enable high-throughput screening and in vivo monitoring.

    • Protein condensate analysis: Techniques to study biomolecular condensates could reveal how PRKAA2 participates in membraneless organelles that concentrate signaling components.

  • Advanced computational and systems biology approaches:

    • Deep learning for image analysis: AI-powered image analysis tools can extract subtle patterns from microscopy data that might reveal new aspects of PRKAA2 regulation.

    • Multi-scale modeling: Computational models integrating molecular, cellular, and tissue-level data can predict emergent properties of PRKAA2 networks.

    • Network pharmacology: Systems-level analysis of drug effects can identify optimal points for therapeutic intervention in PRKAA2 pathways.

    • Digital twins: Computational models of individual patients incorporating PRKAA2 pathway variations could enable personalized therapeutic strategies.

These emerging technologies collectively promise to transform our understanding of PRKAA2 by providing unprecedented resolution, specificity, and integrative insights that bridge molecular mechanisms with physiological functions .

How might PRKAA2 research inform the development of personalized medicine approaches for metabolic and neurological disorders?

PRKAA2 research is poised to contribute significantly to personalized medicine approaches for metabolic and neurological disorders through several promising pathways:

  • Genetic variation and patient stratification:

    • Current understanding: Genetic variations in PRKAA2 and its regulatory pathways likely contribute to individual differences in disease susceptibility and treatment response.

    • Personalization opportunity: Genetic profiling could identify patients most likely to benefit from PRKAA2-targeted therapies based on their specific variants.

    • Implementation approach: Develop a panel of PRKAA2 pathway variants that predict treatment response; validate in prospective clinical trials; create algorithmic treatment decision tools incorporating genetic data.

  • Tissue-specific biomarker development:

    • Current understanding: PRKAA2 exhibits tissue-specific functions and is differentially regulated across tissues, suggesting the need for tissue-specific biomarkers.

    • Personalization opportunity: Non-invasive assessment of tissue-specific PRKAA2 activity could guide personalized dosing and treatment selection.

    • Implementation approach: Identify circulating markers that correlate with tissue-specific PRKAA2 activity; develop imaging probes for non-invasive monitoring; establish biomarker-guided treatment algorithms.

  • Post-translational modification profiles:

    • Current understanding: The function of PRKAA2 is regulated by multiple post-translational modifications, including phosphorylation and succinylation, which may vary between individuals and disease states.

    • Personalization opportunity: Profiles of PRKAA2 modifications could serve as predictive biomarkers for patient-specific treatment strategies.

    • Implementation approach: Develop assays for measuring PRKAA2 modification patterns in accessible samples (blood, CSF); correlate patterns with disease progression and treatment response; design therapies targeting specific modification pathways.

  • Integration with other personalized approaches:

    Precision Medicine ApproachPRKAA2 Research ContributionPersonalization Potential
    PharmacogenomicsIdentification of genetic variants affecting PRKAA2 drug responsesPatient-specific dosing of AMPK modulators
    MetabolomicsCharacterization of PRKAA2-regulated metabolite networksMetabolic phenotyping to guide intervention timing
    Digital Health MonitoringCorrelation of activity/diet with PRKAA2 activationLifestyle recommendations tailored to genetic profile
    Multimodal BiomarkersIntegration of PRKAA2 activity markers with other disease indicatorsComprehensive disease risk and progression monitoring
  • Neurological disorder applications:

    • Current understanding: PRKAA2 regulates neuronal energy homeostasis and shows preferential expression in neuronal tissues.

    • Personalization opportunity: Neurological patients could be stratified based on PRKAA2 pathway integrity and metabolic profiles.

    • Implementation approach: Develop neuron-specific PRKAA2 imaging markers; correlate neuronal bioenergetics with disease progression; tailor interventions to individual neurometabolic profiles.

  • Pregnancy complication prevention:

    • Current understanding: PRKAA2 expression and modification patterns influence placental function and risk of hypertensive disorders.

    • Personalization opportunity: Early screening of PRKAA2-related markers could identify high-risk pregnancies for preventive intervention.

    • Implementation approach: Develop placenta-specific non-invasive biomarkers; create risk prediction models incorporating PRKAA2 pathway data; design early intervention strategies for high-risk individuals.

By integrating PRKAA2 research with other precision medicine approaches, researchers can develop multi-faceted personalized strategies that account for individual variations in PRKAA2 biology, ultimately leading to more effective and targeted treatments for metabolic and neurological disorders .

What are the key takeaways for researchers beginning work with recombinant mouse PRKAA2?

Researchers initiating work with recombinant mouse PRKAA2 should consider these essential guidelines for successful experimental design and implementation:

  • Expression and preparation considerations:

    • Utilize baculovirus-infected Sf9 insect cell systems for optimal expression of functional PRKAA2, as this system provides proper folding and post-translational modifications.

    • Co-express PRKAA2 with appropriate β and γ subunits (typically PRKAB2 and PRKAG1) to generate the complete heterotrimeric complex necessary for physiological activity.

    • Verify protein quality through rigorous purity assessment (>85% purity via SDS-PAGE), phosphorylation status (Thr172 phosphorylation), and functional activity testing.

    • Prepare single-use aliquots stored at -80°C with appropriate stabilizing agents to avoid activity loss from freeze-thaw cycles.

  • Experimental design fundamentals:

    • Include appropriate isoform controls in all experiments to distinguish PRKAA2-specific effects from those common to both AMPK catalytic subunits.

    • Consider tissue-specific expression patterns when designing experiments, with particular attention to neuronal tissues where PRKAA2 shows preferential expression.

    • Incorporate multiple activation readouts (phosphorylation status, substrate phosphorylation, metabolite changes) for comprehensive activity assessment.

    • Design experiments that account for the three distinct mechanisms of AMPK activation: Thr172 phosphorylation, protection from dephosphorylation, and allosteric activation.

  • Biological context awareness:

    • Recognize that PRKAA2 demonstrates isoform-specific functions in neuronal tissues, with unique roles in regulating nucleotide levels (ATP, GTP, cGMP) that are not shared with PRKAA1.

    • Consider the regulatory influence of post-translational modifications beyond phosphorylation, particularly succinylation at K69 and K260 residues.

    • Account for the dynamic interplay between PRKAA2 and energy status, with activation occurring in response to increased AMP:ATP and ADP:ATP ratios during metabolic stress.

    • Acknowledge the emerging role of PRKAA2 in conditions like hypertensive disorders of pregnancy when interpreting results in reproductive biology contexts.

  • Methodological adaptations:

    • Employ physiologically relevant ATP concentrations in kinase assays to avoid masking allosteric effects of AMP.

    • Include appropriate phosphatase inhibitors in all buffers when measuring phosphorylation status.

    • Consider both acute (minutes to hours) and chronic (days to weeks) effects when studying PRKAA2 modulation.

    • Validate antibody specificity rigorously, as many commercial antibodies fail to distinguish between PRKAA1 and PRKAA2.

By integrating these key considerations into research designs, investigators new to PRKAA2 research can establish robust experimental systems that yield reproducible and physiologically relevant results, positioning their work to make meaningful contributions to our understanding of this important metabolic regulator .

How should researchers approach the integration of PRKAA2 studies across different model systems and disease contexts?

Integrating PRKAA2 research across diverse model systems and disease contexts requires a systematic approach that accounts for biological variations while maintaining methodological consistency:

  • Cross-species considerations:

    • Sequence and structure comparison: Begin by analyzing conservation of PRKAA2 sequence, structure, and regulatory elements across species of interest (mouse, human, other models).

    • Expression pattern mapping: Compare tissue-specific expression patterns of PRKAA2 versus PRKAA1 across species using comprehensive databases and direct measurements.

    • Functional validation: Validate key findings across species using consistent methodologies to identify conserved versus species-specific mechanisms.

    • Translational indicators: Establish clear criteria for determining when mouse PRKAA2 findings can be reliably translated to human applications.

  • Cross-disease integration framework:

    • Pathway-centric analysis: Focus on core PRKAA2 pathways that may be dysregulated across multiple conditions rather than disease-specific endpoints.

    • Common mechanism identification: Identify shared molecular mechanisms (e.g., altered succinylation, disrupted energy sensing) that operate across different disease contexts.

    • Modification profile comparison: Compare patterns of PRKAA2 post-translational modifications across conditions to identify disease-specific signatures.

    • Network perturbation mapping: Use systems biology approaches to map how PRKAA2 network perturbations manifest differently across disease contexts.

  • Methodological standardization:

    • Core protocol establishment: Develop standardized protocols for key measurements (activity assays, complex formation, substrate phosphorylation) that can be applied across models.

    • Reference standards creation: Establish common reference standards and positive controls that enable direct comparison of results across laboratories and disease models.

    • Data reporting templates: Implement comprehensive data reporting formats that capture all relevant experimental parameters to facilitate meta-analysis.

    • Community resources development: Create shared resources such as validated antibodies, cell lines, and animal models that can be used across research groups.

  • Integrated data analysis approaches:

    • Multi-omics integration: Combine data from genomics, transcriptomics, proteomics, metabolomics, and phenotypic assays to build comprehensive models of PRKAA2 function.

    • Meta-analysis frameworks: Develop statistical approaches for integrating findings across studies with different designs and endpoints.

    • Machine learning applications: Apply machine learning to identify patterns in PRKAA2 regulation and function that may not be apparent through conventional analysis.

    • Knowledge graph construction: Build comprehensive knowledge graphs that connect PRKAA2 findings across model systems and disease contexts to identify gaps and opportunities.

  • Collaborative research structures:

    • Multi-disciplinary teams: Form collaborative groups that include experts in metabolism, neurobiology, reproductive biology, and other relevant fields.

    • Data sharing platforms: Utilize open science platforms to share raw data, protocols, and resources across research groups.

    • Standardized model development: Collectively develop and characterize standardized models (cell lines, animal models) that can be used across research groups.

    • Coordinated clinical integration: Establish biobanks and patient registries with standardized PRKAA2 pathway assessment to facilitate clinical correlation.

This integrative approach can accelerate translation of fundamental PRKAA2 biology into clinical applications by identifying robust mechanisms that operate across species and disease contexts while accounting for important biological variations .

What key experimental protocols should be in every PRKAA2 researcher's toolkit?

Every researcher working with recombinant mouse PRKAA2 should master these essential protocols to ensure robust and reproducible results:

  • Recombinant PRKAA2 complex production and validation:

    • Expression protocol: Baculovirus-mediated co-expression of PRKAA2, PRKAB2, and PRKAG1 in Sf9 insect cells using optimized viral titers and infection conditions.

    • Purification workflow: Two-step purification using affinity chromatography (typically His-tag or GST-tag) followed by size-exclusion chromatography to ensure complex integrity.

    • Quality control assays: SDS-PAGE with Coomassie staining to verify purity (>85%) and subunit stoichiometry; Western blotting for phospho-Thr172 to confirm activation status; activity assay using SAMS peptide to verify functional integrity.

    • Storage protocol: Preparation of single-use aliquots in stabilizing buffer (typically containing 20% glycerol, 1 mM DTT, and protease inhibitors) stored at -80°C.

  • PRKAA2 activity assessment:

    • In vitro kinase assay: Quantification of PRKAA2 activity using the SAMS peptide substrate with 32P-ATP or a non-radioactive alternative like the ADPGlo Kinase Assay.

    • Cellular activation monitoring: Western blotting for phospho-ACC (Ser79) as a proxy for cellular AMPK activity, with isoform-specific immunoprecipitation to isolate PRKAA2 contribution.

    • Nucleotide ratio determination: LC-MS/MS-based quantification of AMP, ADP, and ATP to calculate energy charge and correlate with PRKAA2 activation.

    • Phospho-specific flow cytometry: For high-throughput assessment of PRKAA2 activation in heterogeneous cell populations.

  • Genetic manipulation approaches:

    • Isoform-specific knockout generation: CRISPR-Cas9 protocol targeting PRKAA2-specific exons to generate clean knockouts without affecting PRKAA1.

    • Conditional knockout strategy: Tissue-specific deletion using Cre-loxP system, with particular focus on neuronal tissues where PRKAA2 shows preferential functions.

    • Rescue experiment design: Re-expression of wild-type or mutant PRKAA2 in knockout backgrounds to establish structure-function relationships.

    • RNA interference protocol: Validated siRNA or shRNA designs with confirmed specificity for PRKAA2 over PRKAA1 for acute knockdown experiments.

  • Post-translational modification analysis:

    • Phosphorylation mapping: Enrichment of phosphopeptides followed by LC-MS/MS to quantify Thr172 phosphorylation and identify other regulatory phosphosites.

    • Succinylation assessment: Immunoprecipitation with anti-succinyllysine antibodies followed by PRKAA2 detection, or targeted MS/MS for K69 and K260 succinylation.

    • PTM cross-talk analysis: Sequential immunoprecipitation protocol to assess how different modifications co-occur or influence each other on individual PRKAA2 molecules.

  • Functional readout assays:

    • Metabolomic profiling: Targeted LC-MS/MS protocol for quantifying ATP, GTP, cGMP, and other nucleotides regulated by PRKAA2 in neuronal tissues.

    • Cell-specific phenotypic assays: Standardized protocols for measuring apoptosis in placental cells, electrophysiological function in neurons, or metabolic flux in various cell types.

    • In vivo functional assessment: Tissue-specific readouts such as electroretinography for visual function in retinal models or blood pressure monitoring in HDCP models.

These core protocols, implemented with rigorous controls and consistent methodology, provide a foundation for productive PRKAA2 research across various applications from basic mechanistic studies to therapeutic development .

What collaborative approaches could accelerate progress in PRKAA2 research?

Strategic collaborative initiatives could substantially accelerate progress in PRKAA2 research by leveraging complementary expertise, resources, and perspectives:

  • Interdisciplinary research consortia:

    • Structure and composition: Form networks connecting researchers from metabolism, neuroscience, reproductive biology, structural biology, and clinical specialties.

    • Operational framework: Establish regular knowledge exchange platforms, shared resource development, and coordinated research priorities.

    • Implementation strategy: Develop focused working groups addressing specific aspects (e.g., neuronal PRKAA2 function, post-translational regulation, therapeutic development).

    • Outcome measures: Joint publications, resource development, and accelerated translation of findings.

  • Standardized resource development:

    • Reagent validation initiative: Collaboratively validate antibodies, recombinant proteins, and small molecule modulators across multiple laboratories.

    • Model system repository: Establish centralized repositories for PRKAA2 mouse models, cell lines, and expression constructs with comprehensive characterization.

    • Protocol standardization: Develop consensus protocols for key PRKAA2 assays with detailed documentation of methodological nuances.

    • Reference data collection: Generate benchmark datasets that can serve as quality control standards for new studies.

  • Integrated data platforms:

    • PRKAA2 knowledge base: Create a specialized database integrating findings across species, tissues, and disease contexts.

    • Multi-omics data repository: Establish centralized storage for raw data from genomics, proteomics, metabolomics, and functional studies.

    • Interactive visualization tools: Develop user-friendly interfaces for exploring PRKAA2 regulatory networks across tissues and conditions.

    • Predictive modeling resources: Share computational models and simulation tools for predicting PRKAA2 pathway responses.

  • Coordinated clinical translation efforts:

    Collaborative ElementKey ActivitiesExpected Impact
    Biomarker ConsortiumValidation of PRKAA2 activity markers across clinical sitesStandardized diagnostic approaches
    Tissue Biobank NetworkCollection of samples with standardized PRKAA2 pathway assessmentRobust human validation of animal findings
    Clinical Trial ConsortiumCoordinated testing of PRKAA2 modulators in multiple conditionsAccelerated therapeutic development
    Patient RegistryLongitudinal data collection from patients with PRKAA2-related disordersImproved understanding of natural history
  • Industry-academia partnerships:

    • Target validation collaborations: Partner with pharmaceutical companies to rigorously validate PRKAA2 as a therapeutic target.

    • Tool compound development: Jointly develop selective PRKAA2 modulators as research tools and potential therapeutic leads.

    • Translational research funding: Establish industry-sponsored challenges or funding mechanisms focused on bridging basic PRKAA2 biology with clinical applications.

    • Technology transfer initiatives: Create streamlined pathways for commercializing academic discoveries related to PRKAA2.

  • Education and workforce development:

    • Specialized training programs: Develop courses and workshops focused on PRKAA2 biology and methodology.

    • Cross-disciplinary exchange programs: Facilitate research rotations between laboratories with complementary expertise.

    • Early career researcher networks: Create mentoring and collaboration opportunities specifically for young investigators in the field.

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.