Phospho-PRKAA2 (Thr172) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Phospho-PRKAA2 (Thr172) Recombinant Monoclonal Antibody: Definition and Mechanism

Phospho-PRKAA2 (Thr172) Recombinant Monoclonal Antibody is a highly specific immunological reagent designed to detect phosphorylation at threonine residue 172 (Thr172) of PRKAA2, the alpha-2 catalytic subunit of AMP-activated protein kinase (AMPK). AMPK is a key energy sensor that regulates metabolic pathways, including fatty acid oxidation, glucose uptake, and cholesterol synthesis, in response to cellular stress or energy depletion .

Key Features:

  • Target: PRKAA2 (AMPKα2) phosphorylated at Thr172.

  • Antibody Type: Recombinant monoclonal antibody, produced via in vitro cloning of antibody genes into mammalian cell lines (e.g., HEK293 or CHO cells) .

  • Purification: Affinity chromatography ensures high specificity and low cross-reactivity .

  • Function: Detects activated AMPK, which is critical for studying metabolic regulation, diabetes, and cancer .

Structure

The antibody binds exclusively to the phosphorylated Thr172 residue, distinguishing it from non-phosphorylated AMPK. This specificity enables precise detection of AMPK activation in experimental models.

Role in AMPK Activation

Phosphorylation at Thr172 is essential for AMPK activation:

  1. Kinase Activation: Thr172 phosphorylation by upstream kinases (e.g., LKB1, CaMKKβ) confers catalytic activity to AMPK .

  2. Allosteric Regulation: AMP binds to the γ subunit, enhancing AMPK activity and protecting Thr172 from dephosphorylation by phosphatases like PP2Cα .

Table 1: AMPK Activation Mechanisms

MechanismRole of Thr172 PhosphorylationKey Regulators
Allosteric ActivationAMP binding stabilizes active conformationAMP, ATP, ADP
PhosphorylationLKB1/CaMKKβ phosphorylate Thr172LKB1, CaMKKβ
DephosphorylationPP2Cα removes phosphate groupPP2Cα, ATP depletion

Common Applications

ApplicationRecommended DilutionSource
Western Blot (WB)1:500–1:5000
Immunofluorescence (IF)1:20–1:200
Immunohistochemistry1:100–1:1000
ELISA1:500–1:1000

Notes:

  • WB Protocol: Incubate membranes with primary antibody (4°C overnight) followed by HRP-conjugated secondary antibody and ECL detection .

  • IF/IHC: Use blocking agents (e.g., 5% BSA) to reduce background; visualize with fluorescent or chromogenic substrates .

Research Findings

  1. Metabolic Regulation: AMPK activation via Thr172 phosphorylation inhibits acetyl-CoA carboxylase (ACC) and HMG-CoA reductase, reducing fatty acid and cholesterol synthesis .

  2. AMP-Dependent Activation: AMP stabilizes the phosphorylated state of Thr172 by inhibiting dephosphorylation, as shown in studies using recombinant AMPK and PP2Cα .

  3. Disease Relevance: Dysregulated Thr172 phosphorylation is implicated in obesity, diabetes, and cancer .

Supplier Overview

SupplierCloneReactivityApplicationsPrice (USD)
CUSABIOCSB-RA805325HumanWB, IF, ELISA210
Cell Signaling#2531, #2535H, M, R, MkWB, IP, IHC39 delivery
Sigma-Aldrich (ZooMAb)5J23H, M, R, MkIHC, ICC, WB230
Thermo Fisher (10H2L20)10H2L20H, M, R, MkWB, ICC280

Notes:

  • Reactivity: Most antibodies cross-react with human, mouse, rat, and monkey .

  • Storage: Typically stored at 4°C (short-term) or -20°C (long-term) .

Critical Considerations

  1. Specificity Validation: Confirm antibody specificity using non-phosphorylated controls or phosphatase-treated samples .

  2. Cross-Reactivity: Ensure no binding to non-target phosphorylated proteins (e.g., AMPKα1 Thr183) .

  3. Experimental Controls: Use total AMPK antibodies (e.g., Cell Signaling #2795) to normalize phosphorylation signals .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

This recombinant PRKAA2 antibody is a monoclonal antibody generated through a sophisticated process. PRKAA2 antibody genes were cloned into plasma vectors, and these vector clones were transfected into stable cell lines for antibody production. Commonly employed mammalian cell lines for this purpose include CHO and HEK293 cells. The recombinant PRKAA2 antibody was then purified using affinity chromatography. Its efficacy has been verified in ELISA, Western blotting, and immunofluorescence assays for the detection of PRKAA2 protein in human samples.

The Thr172 Phospho-PRKAA2 antibody is specifically designed to detect the PRKAA2 protein phosphorylated at the Thr172 residue. PRKAA2, also known as the AMPK alpha 2 subunit, plays a critical role in cellular energy metabolism. Its phosphorylation at threonine 172 by upstream AMPK kinase (AMPKK) is essential for fully activating AMPK. AMPK acts as a cellular energy sensor, regulating energy-producing metabolic and biosynthetic pathways during physiological and pathological cellular stress. AMPK activation promotes energy production by stimulating fatty acid oxidation, glucose transport, and glycolysis, while simultaneously inhibiting energy-consuming processes such as triglyceride and protein synthesis.

Form
Liquid
Lead Time
We typically ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery time estimates.
Synonyms
5'-AMP-activated protein kinase catalytic subunit alpha-2 antibody; AAPK2_HUMAN antibody; ACACA kinase antibody; Acetyl CoA carboxylase kinase antibody; Acetyl-CoA carboxylase kinase antibody; AMPK alpha 2 chain antibody; AMPK subunit alpha-2 antibody; AMPK2 antibody; AMPKa2 antibody; AMPKalpha2 antibody; HMGCR kinase antibody; Hydroxymethylglutaryl CoA reductase kinase antibody; Hydroxymethylglutaryl-CoA reductase kinase antibody; PRKAA antibody; PRKAA2 antibody; Protein kinase AMP activated alpha 2 catalytic subunit antibody; Protein kinase AMP activated catalytic subunit alpha 2 antibody
Target Names
Uniprot No.

Target Background

Function
PRKAA2 is the catalytic subunit of AMP-activated protein kinase (AMPK). AMPK is a crucial energy sensor protein kinase that plays a central role in regulating cellular energy metabolism. When intracellular ATP levels decline, AMPK activates energy-producing pathways while simultaneously inhibiting energy-consuming processes. These processes include the inhibition of protein, carbohydrate, and lipid biosynthesis, as well as cell growth and proliferation. AMPK exerts its effects through direct phosphorylation of metabolic enzymes and by longer-term effects via phosphorylation of transcription regulators.

AMPK also serves as a regulator of cellular polarity by remodeling the actin cytoskeleton, potentially by indirectly activating myosin. It regulates lipid synthesis by phosphorylating and inactivating lipid metabolic enzymes, including ACACA, ACACB, GYS1, HMGCR, and LIPE. This regulation impacts fatty acid and cholesterol synthesis through phosphorylation of acetyl-CoA carboxylase (ACACA and ACACB) and hormone-sensitive lipase (LIPE) enzymes, respectively.

AMPK further regulates insulin signaling and glycolysis by phosphorylating IRS1, PFKFB2, and PFKFB3. It is involved 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 mediating phosphorylation of TBC1D4/AS160.

AMPK exerts significant influence on transcription and chromatin structure by phosphorylating transcription regulators involved in energy metabolism. These include CRTC2/TORC2, FOXO3, histone H2B, HDAC5, MEF2C, MLXIPL/ChREBP, EP300, HNF4A, p53/TP53, SREBF1, SREBF2, and PPARGC1A. AMPK acts as a key regulator of glucose homeostasis in the liver by phosphorylating CRTC2/TORC2, leading to its sequestration in the cytoplasm. In response to stress, AMPK phosphorylates 'Ser-36' of histone H2B (H2BS36ph), promoting transcription.

AMPK is a key regulator of cell growth and proliferation. It accomplishes this by phosphorylating TSC2, RPTOR, and ATG1/ULK1. In response to nutrient limitation, AMPK negatively regulates the mTORC1 complex by phosphorylating RPTOR, a component of the mTORC1 complex, and by phosphorylating and activating TSC2. Under nutrient-limited conditions, AMPK promotes autophagy by phosphorylating and activating ATG1/ULK1, while also activating WDR45. AMPK also regulates circadian rhythm by mediating the phosphorylation of CRY1, leading to its destabilization. It may also regulate the Wnt signaling pathway by phosphorylating CTNNB1, stabilizing it. AMPK also phosphorylates CFTR, EEF2K, KLC1, NOS3, and SLC12A1.

AMPK plays a crucial role in the differential regulation of pro-autophagy (composed of PIK3C3, BECN1, PIK3R4, and UVRAG or ATG14) and non-autophagy (composed of PIK3C3, BECN1, and PIK3R4) complexes in response to glucose starvation. It can inhibit the non-autophagy complex by phosphorylating PIK3C3 and can activate the pro-autophagy complex by phosphorylating BECN1.
Gene References Into Functions
  1. Polymorphisms in PRKAA2 at rs10789038 and rs2796498 have been linked to susceptibility to type 2 diabetes mellitus and diabetic nephropathy. PMID: 28322508
  2. AMPK phosphorylates DNMT1, RBBP7, and HAT1, enhancing their interactions. PMID: 28143904
  3. PGC-1alpha protein levels were higher after HIHVT compared to SIT (p < 0.05). Additionally, the AMPKpTHR172/AMPK ratio increased post-SIT (p < 0.05), while this effect was delayed after HIHVT, increasing after 3 hours. PMID: 28973039
  4. TNF-alpha treatment of colonic rho(0) cells significantly augmented IL-8 expression by 9-fold (P < 0.01) via NF-kappaB compared to TNF-alpha-treated controls. Reduced mitochondrial function facilitated TNF-alpha-mediated NF-kappaB luciferase promoter activity by lowering inhibitory IkappaBalpha (nuclear factor of kappa light polypeptide gene enhancer in B-cell inhibitor, alpha), leading to elevated NF-kappaB. ... PMID: 28183804
  5. Findings indicate that AMPKalpha2 plays a role in controlling bladder cancer growth by regulating proliferation through mTOR suppression and induction of p27 protein levels. This suggests how AMPKalpha2 loss may contribute to tumorigenesis. PMID: 27638620
  6. AMPK phosphorylation of cortactin followed by SIRT1 deacetylation modulates the interaction of cortactin and cortical-actin in response to shear stress. This AMPK/SIRT1 coregulated cortactin-F-actin dynamics are required for endothelial nitric oxide synthase subcellular translocation/activation and contribute to atheroprotection. PMID: 27758765
  7. Inactivation of AMPKalpha2, but not AMPKalpha1, abrogates the tumor attenuation caused by UBE2O loss. PMID: 28162974
  8. Research demonstrates that the AMPKalpha2 catalytic subunit in Kiss1 cells is dispensable for body weight and reproductive function in mice but is necessary for the reproductive adaptations to conditions of acute metabolic distress. PMID: 27732087
  9. Binding of miR-27a to the 3'-UTR of the AMPKalpha2 gene was required to increase the sensitivity of breast cancer cells to metformin. PMID: 27779715
  10. p-AMPKalpha was associated with cervical carcinogenesis, and high expression of AMPKalpha2 was correlated with better disease-free survival in patients with early-stage cervical cancer. PMID: 26337566
  11. AMPK-alpha-2 binds to both ER-alpha and ER-beta. PMID: 26374855
  12. Data shows that activated AMP-activated protein kinase (AMPK) limits retinal pigment epithelial cells (RPE) phagocytic activity by abolishing retinal photoreceptor cell outer segment (POS)-induced activation of c-mer proto-oncogene tyrosine kinase (MerTK). PMID: 26427488
  13. AMPKalpha signaling suppresses epithelial-mesenchymal transition (EMT) and secretion of chemokines in renal tubular epithelia through interaction with CK2beta to attenuate renal injury. PMID: 26108355
  14. A subunit composition of AMPK (alpha2beta2gamma1) is preferred for colorectal cancer cell survival, at least partially by stabilizing the tumor-specific expression of PGC1B. PMID: 26351140
  15. CaMKKbeta-AMPKalpha2 signaling contributes to mitotic Golgi fragmentation and the G2/M transition in mammalian cells. PMID: 25590814
  16. The basal level of ERK phosphorylation was suppressed in cells treated with siRNA against PRKAA2 (AMPK-knockdown cells). PMID: 25846811
  17. AMPK-alpha co-immunoprecipitates with the insulin receptor (IR). The compartmentalization of AMPK-alpha between plasma membrane and Golgi/endosomes fractions in rat's liver is insulin-dependent. Insulin receptor internalization was markedly decreased after AMPK-alpha2 knockdown, and treatment with the ATIC substrate AICAR, an allosteric activator of AMPK, increased IR endocytosis in human cultured cells and rat liver. PMID: 25687571
  18. AMPKalpha2 is an essential mediator of nicotine-induced whole-body insulin resistance (IR) despite reductions in adiposity. PMID: 25799226
  19. High PRKAA2 expression is associated with follicular lymphoma. PMID: 23396962
  20. AMPKalpha2 suppresses endothelial ACE expression via phosphorylation of p53 and upregulation of microRNA-143/145. PMID: 23476055
  21. WWP1 down-regulates AMPKalpha2 under high glucose culture conditions via the ubiquitin-proteasome pathway. PMID: 23293026
  22. Underexpression of AMPK is frequently observed in hepatocellular carcinoma (HCC), and its inactivation promotes hepatocarcinogenesis by destabilizing p53 in a SIRT1-dependent manner. PMID: 22728651
  23. Research demonstrated that intronic SNPs in the genes encoding the catalytic alpha2 (PRKAA2) subunit of AMPK are associated with antipsychotic-induced weight gain in schizophrenia or schizoaffective disorder patients. PMID: 22305490
  24. AMPK coordinates nutrient status with mitosis completion, which may be critical for the organism's response to low nutrients during development or in adult stem and cancer cells. PMID: 22137581
  25. AMP activated protein kinase-alpha2 regulates the expression of estrogen-related receptor-alpha, a metabolic transcription factor linked to heart failure development. PMID: 21825219
  26. IKK is a direct substrate of AMPKalpha2. PMID: 21673972
  27. The crystal structure of the phosphorylated-state mimic T172D mutant kinase domain from the human AMPK alpha2 subunit is reported in the apo form and in complex with a selective inhibitor, compound C. PMID: 21543851
  28. AMPKalpha2, by affecting Fyn phosphorylation and activity, plays a key role in platelet alphaIIbbeta3 integrin signaling, leading to clot retraction and thrombus stability. PMID: 20558612
  29. AMPK activates transcription by directly associating with chromatin and phosphorylating histone H2B at ser36. These findings place AMPK-dependent H2B Ser36 phosphorylation in a direct transcriptional and chromatin regulatory pathway leading to stress adaptation. PMID: 20647423
  30. Purification and characterization of truncated human AMPK alpha 2 beta 2 gamma 3 heterotrimer from baculovirus-infected insect cells. PMID: 19836452
  31. AMPKalpha2 functions as a physiological suppressor of NAD(P)H oxidase, ROS production, and 26S proteasome activity in endothelial cells. PMID: 20167927
  32. AMPK functions as a physiological suppressor of endoplasmic reticulum stress by maintaining SERCA activity and intracellular Ca2+ homeostasis. PMID: 20124121
  33. Hepatic amino acid-dependent signaling is under the control of AMP-dependent protein kinase. PMID: 12067722
  34. AMPK signaling is not a key regulatory system of muscle substrate combustion during prolonged exercise. Marked activation of AMPK via phosphorylation is not sufficient to maintain an elevated ACCbeta Ser(221) phosphorylation during prolonged exercise. PMID: 12413941
  35. Regulation of channel gating by AMP-activated protein kinase modulates cystic fibrosis transmembrane conductance regulator activity in lung submucosal cells. PMID: 12427743
  36. Overexpression of mutant AMPK-alpha1 enhanced forskolin-stimulated I short-circuit currents, consistent with dominant-negative reduction in inhibition of CFTR by endogenous AMPK. PMID: 12519745
  37. Results suggest that nuclear translocation of 5' AMP-activated protein kinase might mediate the effects of exercise on skeletal muscle gene and protein expression. PMID: 12663462
  38. Endothelial AMPK signaling may be a critical determinant of blood vessel recruitment to tissues subjected to ischemic stress. PMID: 12788940
  39. LKB1 kinase, associated with Peutz-Jeghers cancer-susceptibility syndrome, phosphorylates and activates AMPK in vitro. PMID: 12847291
  40. Acetyl-CoA carboxylase beta phosphorylation is particularly sensitive to exercise and tightly coupled to AMPK signaling. Notably, AMPK activation during exercise does not depend on AMPK kinase activation. PMID: 12941758
  41. This study demonstrates that protein content and basal AMPK activity in human skeletal muscle are highly susceptible to endurance exercise training. PMID: 14613924
  42. AMPK and eEF2 kinase may provide a key link between cellular energy status and the inhibition of protein synthesis, a major consumer of metabolic energy. PMID: 14709557
  43. AMPK is a major regulator of skeletal muscle HSL activity and can override beta-adrenergic stimulation. PMID: 15231718
  44. AMPK is a novel and critical component of HIF-1 regulation, suggesting its involvement in vanadate-induced prostate carcinogenesis. PMID: 15297373
  45. Results indicate that AMP-activated protein kinase (AMPK)alpha negatively regulates acetyl-CoA carboxylase activity and hepatic lipid content. PMID: 15371448
  46. AMPK plays a role in the phenobarbital induction of CYP2B gene expression. PMID: 15572372
  47. There is significant basal activity and phosphorylation of AMPK in LKB1-deficient cells that can be stimulated by Ca2+ ionophores. Studies using the CaMKK inhibitor STO-609 and isoform-specific siRNAs demonstrate that CaMKKbeta is required for this effect. PMID: 16054095
  48. Overexpression of CaMKKbeta in mammalian cells increases AMPK activity, whereas pharmacological inhibition of CaMKK or downregulation of CaMKKbeta using RNA interference nearly completely abolishes AMPK activation. PMID: 16054096
  49. Causes inappropriate AMP kinase activation, leading to glycogen accumulation and heart conduction system disease when transfected into mice. PMID: 16275868
  50. Data suggests that moderate endurance exercise promotes glucose transport, GLUT4 expression, and insulin sensitivity in skeletal muscle at least partially via activation of the alpha2 isoform of AMPK. PMID: 16483872

Show More

Hide All

Database Links

HGNC: 9377

OMIM: 600497

KEGG: hsa:5563

STRING: 9606.ENSP00000360290

UniGene: Hs.437039

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 PRKAA2 and what is its physiological significance?

PRKAA2 (Protein Kinase AMP-Activated Catalytic Subunit Alpha 2) is one of the catalytic subunits of AMP-activated protein kinase (AMPK), a crucial energy sensor protein kinase that regulates cellular energy metabolism. PRKAA2 plays a key role in responding to reductions in intracellular ATP levels by activating energy-producing pathways while inhibiting energy-consuming processes. It functions by directly phosphorylating metabolic enzymes and through longer-term effects via phosphorylation of transcription regulators .

PRKAA2 regulates lipid synthesis by phosphorylating and inactivating lipid metabolic enzymes including ACACA, ACACB, GYS1, HMGCR, and LIPE. It also regulates insulin signaling and glycolysis by phosphorylating IRS1, PFKFB2, and PFKFB3. Studies with mouse models suggest that this catalytic subunit controls whole-body insulin sensitivity and is essential for maintaining myocardial energy homeostasis during ischemia .

Notably, PRKAA2 has substrate specificity that differs from its paralog PRKAA1. Research indicates that PRKAA2 expression correlates with CRTC2, TNNI3, KCNA5, and TFEB expression, suggesting distinct regulatory roles compared to other AMPK subunits .

What is the significance of Thr172 phosphorylation in PRKAA2 function?

Phosphorylation of a conserved threonine residue within the activation loop (typically referred to as Thr172) can increase the kinase activity of PRKAA2 by more than 100-fold. This phosphorylation site represents a critical regulatory mechanism for AMPK activity .

The major upstream kinases responsible for phosphorylating Thr172 include the tumor suppressor kinase liver kinase B1 (LKB1) in complex with accessory subunits STRAD and MO25, as well as Ca²⁺/calmodulin-dependent protein kinase kinase 2 (CAMKK2). This phosphorylation is fundamental to AMPK's ability to respond to cellular energy status .

The regulatory mechanisms involving Thr172 phosphorylation are complex and tightly controlled. Binding of AMP to the γ subunit of the AMPK complex activates the kinase through three mechanisms: (a) allosteric activation, (b) promotion of Thr172 phosphorylation by LKB1, and (c) inhibition of Thr172 dephosphorylation by protein phosphatases. These mechanisms allow for precise control of AMPK activity in response to cellular energy status .

How do recombinant monoclonal antibodies against Phospho-PRKAA2 (Thr172) differ from conventional antibodies?

Recombinant monoclonal antibodies against Phospho-PRKAA2 (Thr172), such as clone 4F4, are generated by cloning PRKAA2 antibody genes into plasma vectors and transfecting vector clones, rather than through traditional hybridoma technology . This recombinant approach offers several advantages over conventional antibodies:

  • Increased reproducibility: By using cloned antibody genes, recombinant antibodies maintain consistent properties across different production batches, eliminating the batch-to-batch variation often seen with hybridoma-derived antibodies.

  • Higher specificity: These antibodies are designed to specifically recognize the phosphorylated form of PRKAA2 at Thr172, making them highly selective tools for studying AMPK activation states.

  • Defined sequence: The exact amino acid sequence of recombinant antibodies is known, allowing for better characterization and potential engineering to improve performance.

The immunogen for these antibodies is typically a synthesized peptide derived from human Phospho-PRKAA2 (Thr172), ensuring targeted recognition of this specific phosphorylation site .

What are the validated applications for Phospho-PRKAA2 (Thr172) recombinant monoclonal antibodies?

Phospho-PRKAA2 (Thr172) recombinant monoclonal antibodies have been validated for multiple research applications:

  • Western Blot (WB): These antibodies can detect phosphorylated PRKAA2 in protein lysates, with recommended dilutions typically ranging from 1:500 to 1:5000. This application allows researchers to quantify relative levels of AMPK activation in different experimental conditions .

  • Immunofluorescence (IF): For cellular localization studies, these antibodies can be used at dilutions of approximately 1:20 to 1:200, enabling visualization of phosphorylated PRKAA2 within cellular compartments .

  • Enzyme-Linked Immunosorbent Assay (ELISA): These antibodies can be used in ELISA applications for quantitative detection of phosphorylated PRKAA2 in complex biological samples .

These applications make Phospho-PRKAA2 (Thr172) antibodies versatile tools for studying AMPK activation in various research contexts, from basic signaling studies to disease models where AMPK dysregulation may play a role.

How can researchers optimize Western Blot protocols for detecting Phospho-PRKAA2 (Thr172)?

Optimizing Western Blot protocols for detecting Phospho-PRKAA2 (Thr172) requires careful attention to several critical factors:

  • Sample preparation: Phosphorylation states can be labile, so samples should be collected rapidly with phosphatase inhibitors included in lysis buffers. Flash freezing tissues immediately after collection is recommended to preserve phosphorylation status.

  • Antibody dilution optimization: While manufacturer recommendations suggest dilutions of 1:500 to 1:5000 for Western Blot applications , researchers should perform titration experiments with their specific samples to determine optimal concentration.

  • Blocking optimization: Using 5% BSA in TBS-T rather than milk-based blocking solutions is typically preferred for phospho-specific antibodies, as milk contains phosphoproteins that may interfere with detection.

  • Positive and negative controls: Include samples treated with AMPK activators (e.g., AICAR, metformin) as positive controls and samples from PRKAA2 knockdown cells or tissues as negative controls. Additionally, samples treated with lambda phosphatase can serve as dephosphorylation controls.

  • Stripping and reprobing: When comparing phosphorylated to total PRKAA2, consider running duplicate gels rather than stripping and reprobing, as stripping can lead to protein loss and signal reduction, particularly for phosphorylated epitopes.

  • Signal detection system selection: Enhanced chemiluminescence (ECL) systems with longer exposure times may be necessary for detecting low abundance phospho-proteins, but care must be taken to avoid signal saturation.

Researchers should also be aware that AMPK phosphorylation status can change rapidly during sample handling, making consistent protocols crucial for reproducible results.

What are the challenges in detecting phosphorylated PRKAA2 in different tissue or cell types?

Detecting phosphorylated PRKAA2 across different tissue or cell types presents several challenges that researchers must address:

  • Tissue-specific expression patterns: Different tissues express varying levels of PRKAA2 relative to PRKAA1. For example, PRKAA2 is more abundantly expressed in cardiac and skeletal muscle, while PRKAA1 is more ubiquitously expressed . These expression differences necessitate tissue-specific protocol optimization.

  • Subcellular localization variations: AMPK can localize to different subcellular compartments depending on cell type and activation status. Evidence suggests that N-myristoylation of the β subunits plays a role in AMPK localization to specific membranes, including mitochondrial membranes . These localization differences may affect extraction efficiency.

  • Basal phosphorylation differences: Different cell types exhibit varying basal levels of PRKAA2 phosphorylation, making it essential to establish appropriate baseline controls specific to each experimental system.

  • Substrate specificity across tissues: Research has shown that PRKAA2 interacts with different substrates in a tissue-specific manner. For instance, PRKAA2 expression correlates with CRTC2, TNNI3, KCNA5, and TFEB expression, which may vary across tissues . This substrate specificity might influence phosphorylation dynamics.

  • Phosphatase activity variations: Differences in phosphatase activity across tissue types can affect the stability of phosphorylated PRKAA2 during sample processing, requiring tissue-specific optimization of phosphatase inhibitor cocktails.

To address these challenges, researchers should consider tissue-specific protocol optimization, careful selection of controls, and potentially the use of phospho-specific antibodies in combination with total PRKAA2 antibodies to normalize for expression level differences.

How does PRKAA2 activation differ between normal and pathological conditions?

The activation patterns of PRKAA2 show significant differences between normal physiological conditions and various pathological states:

  • Normal physiological activation: Under normal conditions, PRKAA2 is activated in response to metabolic stress that increases the AMP:ATP ratio, such as during exercise, caloric restriction, or hypoxia. This activation helps maintain energy homeostasis by promoting ATP-generating pathways and inhibiting ATP-consuming processes .

  • Cancer pathology: Recent research indicates that PRKAA2 may contribute to liver hepatocellular carcinoma (LIHC) progression by promoting metabolic reprogramming and tumor immune escape. High expression of PRKAA2 in LIHC is associated with poor patient prognosis and immune cold phenotypes . This suggests that in certain cancer contexts, PRKAA2 activation may be detrimental rather than protective.

  • Substrate targeting in disease: In pathological states, PRKAA2 may phosphorylate different substrates compared to normal conditions. For example, in cancer cells, PRKAA2 has been shown to regulate the interferon-gamma response and MHC class I expression, potentially contributing to immune evasion .

  • Down-regulation mechanisms in cancer: Various cancers have developed mechanisms to down-regulate AMPK activity, including phosphorylation of AMPK-α1 at Ser487 by Akt (inhibiting subsequent Thr172 phosphorylation) and degradation of AMPK-α1 following polyubiquitylation by the E3 ligase TRIM28, which is targeted to AMPK by MAGE-A3/A6 .

  • Role in immune responses: PRKAA2 has been implicated in promoting CD8+ T-cell exhaustion and the formation of CD4+ Treg cells, potentially altering immune responses in pathological conditions .

Understanding these context-dependent differences in PRKAA2 activation is crucial for developing targeted therapeutic approaches for diseases involving AMPK dysregulation.

What methods can be employed to validate the specificity of Phospho-PRKAA2 (Thr172) antibodies?

Validating the specificity of Phospho-PRKAA2 (Thr172) antibodies is crucial for generating reliable research data. Several complementary approaches should be employed:

  • Genetic knockdown/knockout validation: The most definitive validation involves using PRKAA2 knockdown or knockout samples. Researchers can generate PRKAA2 knockout cells using CRISPR-Cas9 or knockdown cells using shRNA (e.g., with sequences like "GTGGCTTATCATCTTATCATT" or "GTCATCCTCATATTATCAAAC" as described in the literature) . The antibody should show substantially reduced or absent signal in these samples.

  • Phosphatase treatment controls: Treating cell lysates with lambda phosphatase before Western blot analysis should eliminate the phospho-specific signal while preserving total PRKAA2 detection with a total PRKAA2 antibody.

  • AMPK activation/inhibition: Treatment with known AMPK activators (e.g., AICAR, metformin, glucose deprivation) should increase phospho-PRKAA2 signal, while inhibitors should decrease it. These pharmacological validations provide functional confirmation of antibody specificity.

  • Peptide competition assays: Pre-incubating the antibody with excess phosphorylated peptide immunogen (derived from human Phospho-PRKAA2 Thr172) should block specific binding and eliminate signal in subsequent applications.

  • Isoform cross-reactivity testing: Test the antibody against both PRKAA1 and PRKAA2 to ensure it does not cross-react with phosphorylated PRKAA1, which has high sequence homology around the Thr172 site.

  • Multiple detection methods: Confirm specificity using different techniques such as Western blot, immunofluorescence, and ELISA to ensure consistent specificity across applications .

By employing these validation strategies, researchers can confidently interpret their results and avoid potential artifacts or misinterpretations due to nonspecific antibody reactivity.

What are the recent discoveries about PRKAA2's role in tumor immunology and cancer progression?

Recent research has revealed significant insights into PRKAA2's previously unrecognized roles in tumor immunology and cancer progression:

  • Immune cold phenotype: Tumors with high PRKAA2 expression display an "immune cold" phenotype, characterized by significantly lower proportions of immune cells compared to tumors with low PRKAA2 expression (43.8% vs. 75.84%) . This suggests PRKAA2 may contribute to immune exclusion in the tumor microenvironment.

  • MHC-I expression regulation: High PRKAA2 expression has been associated with inhibition of major histocompatibility complex class I (MHC-I) expression in malignant cells through regulation of interferon-gamma activity . This decreased antigen presentation capacity can help tumor cells evade immune surveillance.

  • T-cell exhaustion promotion: PRKAA2 has been implicated in promoting CD8+ T-cell exhaustion and the formation of CD4+ regulatory T cells (Tregs), both of which can suppress anti-tumor immune responses . This represents a novel mechanism by which AMPK signaling might influence immune escape.

  • Metabolic reprogramming: Beyond its immune effects, PRKAA2 may contribute to cancer progression by inducing metabolic reprogramming of malignant cells. Single-sample Gene Set Variation Analysis (GSVA) revealed that IFN-γ response pathways were enriched in malignant cells with low PRKAA2 expression, suggesting PRKAA2 may suppress this anti-tumor pathway .

  • Prognostic significance: High PRKAA2 expression is associated with poor prognosis in liver hepatocellular carcinoma (LIHC), indicating its potential value as a prognostic biomarker .

These findings highlight the complex and context-dependent roles of PRKAA2 in cancer biology, suggesting that in some contexts, PRKAA2 may function as a tumor promoter rather than a tumor suppressor, particularly through its effects on tumor immunity.

How does the AMPK heterotrimeric complex structure influence antibody selection and experimental design?

The complex structural organization of AMPK heterotrimers has important implications for antibody selection and experimental design:

  • Subunit composition considerations: AMPK exists as heterotrimeric complexes containing catalytic α subunits (PRKAA1 or PRKAA2) and regulatory β and γ subunits, each with multiple isoforms . This diversity creates 12 possible heterotrimeric combinations with potentially different functions, subcellular localizations, and regulatory properties. When designing experiments to study specific AMPK complexes, researchers must consider which α subunit (PRKAA1 or PRKAA2) is relevant for their biological context.

  • Domain-specific epitope targeting: The α subunits contain N-terminal kinase domains (α-KD) with the phosphorylatable Thr172 in the activation loop . Antibodies specifically targeting phospho-Thr172 must access this site, which can be influenced by the complex's conformational state.

  • Regulatory interactions and epitope accessibility: The binding of AMP to the γ subunit induces conformational changes that affect the α-linker region and the accessibility of Thr172 . These conformational states may influence epitope availability for antibody binding, potentially affecting detection efficiency in different activation states.

  • Post-translational modification interplay: Beyond Thr172 phosphorylation, other post-translational modifications like phosphorylation of AMPK-α1 at Ser487 by Akt can inhibit subsequent phosphorylation at Thr172 . Researchers should consider how these modifications might affect epitope recognition by phospho-specific antibodies.

  • Subcellular localization effects: N-myristoylation of the β subunits affects AMPK localization to specific membranes, including lysosomal and mitochondrial membranes . This localization may affect extraction efficiency and detection, requiring specific sample preparation protocols depending on the cellular compartment of interest.

Understanding these structural considerations is essential for choosing appropriate antibodies and designing experiments that accurately detect and quantify phosphorylated PRKAA2 in its native context.

What are the recommended protocols for measuring PRKAA2 activity in cell and tissue samples?

Measuring PRKAA2 activity in biological samples requires careful consideration of multiple methodological approaches:

  • Western blot analysis of Thr172 phosphorylation: This is the most common approach for assessing AMPK activation, using phospho-specific antibodies targeting Thr172. Recommended dilutions for Western blot applications typically range from 1:500 to 1:5000 . Normalization to total PRKAA2 levels is essential for accurate interpretation.

  • AMPK substrate phosphorylation: Measuring the phosphorylation of direct AMPK substrates provides functional validation of PRKAA2 activity. Key substrates include acetyl-CoA carboxylase (ACC) at Ser79, which can be detected using phospho-specific antibodies. The substrate specificity of PRKAA2 (correlating with CRTC2, TNNI3, KCNA5, and TFEB) should be considered when selecting appropriate substrates for monitoring.

  • In vitro kinase assays: Immunoprecipitated PRKAA2 can be used in kinase assays with recombinant substrates and radiolabeled ATP to directly measure enzymatic activity. This approach separates PRKAA2 from PRKAA1 activity.

  • Genetic manipulation approaches: Targeted knockdown or knockout of PRKAA2 using shRNA (with validated sequences like "GTGGCTTATCATCTTATCATT" or "GTCATCCTCATATTATCAAAC") or CRISPR-Cas9 can help distinguish PRKAA2-specific effects from PRKAA1 or compensatory pathways.

  • Real-time PCR quantification: For measuring PRKAA2 expression levels rather than activity, qPCR can be performed using validated primer pairs such as forward 5′-CGGGTGAAGATCGGACACTA-3′ and reverse 5′-TCCAACAACATCTAAACTGCGA-3′, with GAPDH as a reference gene .

When implementing these protocols, researchers should be mindful of rapid changes in phosphorylation status during sample handling and include appropriate positive controls (AMPK activators) and negative controls (phosphatase treatment or PRKAA2 knockout/knockdown).

How can researchers differentiate between PRKAA1 and PRKAA2 activity in experimental systems?

Differentiating between the activities of the two catalytic α subunits of AMPK (PRKAA1 and PRKAA2) is crucial for understanding their specific roles:

  • Isoform-specific antibodies: Use highly specific antibodies that can distinguish between phosphorylated PRKAA1 and PRKAA2. Validation using knockout or knockdown models for each isoform is essential to confirm specificity.

  • Selective genetic manipulation: Employ isoform-specific knockdown or knockout strategies. For PRKAA2 knockdown, validated shRNA sequences (such as "GTGGCTTATCATCTTATCATT" or "GTCATCCTCATATTATCAAAC") can be used to selectively reduce PRKAA2 without affecting PRKAA1.

  • Isoform-specific substrate analysis: Leverage the distinct substrate preferences of the two isoforms. Research has shown that PRKAA1 expression correlates with BRAF, C18orf25, EEF2K, and EP300 expression, while PRKAA2 expression correlates with CRTC2, TNNI3, KCNA5, and TFEB expression . Monitoring phosphorylation of these specific substrates can provide indirect measures of isoform-specific activity.

  • Tissue-specific analysis: Consider the relative expression levels of each isoform in different tissues. PRKAA2 is more abundantly expressed in cardiac and skeletal muscle, while PRKAA1 is more ubiquitously expressed . This tissue distribution can be leveraged to study predominantly one isoform in certain contexts.

  • Subcellular fractionation: The two isoforms may have different subcellular localizations, which can be exploited through careful subcellular fractionation followed by isoform-specific detection.

  • Pathway enrichment analysis: Perform KEGG pathway analysis to identify differentially enriched pathways between the two isoforms. Research has shown that diverse AMPK isoforms participate in different regulatory programs , which can provide functional readouts of isoform-specific activity.

By combining these approaches, researchers can more confidently attribute observed phenotypes to specific AMPK catalytic subunits.

What experimental conditions are optimal for studying PRKAA2 phosphorylation dynamics?

Optimizing experimental conditions for studying PRKAA2 phosphorylation dynamics requires careful consideration of several key factors:

  • Physiological stimuli selection: Choose stimuli relevant to the research question. Common AMPK activators include:

    • Energy stress inducers: glucose deprivation, 2-deoxyglucose

    • Pharmacological activators: AICAR, metformin, A-769662

    • Physiological conditions: exercise mimetics, calcium ionophores

  • Time course considerations: AMPK phosphorylation is dynamic and often transient. Design experiments with multiple time points (ranging from minutes to hours) to capture both acute activation and potential adaptation/feedback inhibition.

  • Cell culture conditions: Standard culture media containing high glucose can suppress basal AMPK activity. Consider using physiologically relevant glucose concentrations (5.5 mM vs. 25 mM) when studying AMPK dynamics.

  • Sample harvesting and preservation: Rapid sample collection and processing are essential to preserve phosphorylation status. Use phosphatase inhibitor cocktails in lysis buffers and consider flash-freezing samples in liquid nitrogen immediately after collection.

  • Controls for antibody validation: Include appropriate controls for validating phospho-specific antibody performance:

    • Positive controls: samples treated with known AMPK activators

    • Negative controls: phosphatase-treated samples or PRKAA2 knockdown/knockout samples

    • Dose-response experiments: titration of activating stimuli to demonstrate proportional phosphorylation changes

  • Serum conditions: Serum contains growth factors that can activate pathways (like Akt) that inhibit AMPK. Serum starvation prior to experiments may be necessary to reduce background signaling and improve signal-to-noise ratio.

  • Normalization strategy: Normalize phospho-PRKAA2 signals to total PRKAA2 protein levels to account for expression differences between samples. This requires careful antibody selection for total PRKAA2 detection that doesn't cross-react with PRKAA1.

  • Consideration of substrate specificity: Monitor phosphorylation of known PRKAA2-specific substrates (CRTC2, TNNI3, KCNA5, and TFEB) to confirm functional activation beyond Thr172 phosphorylation.

By optimizing these conditions, researchers can more reliably study the dynamic regulation of PRKAA2 phosphorylation in their experimental systems.

How can phospho-proteomics approaches complement antibody-based detection of PRKAA2 activity?

Phospho-proteomics approaches offer powerful complementary strategies to antibody-based methods for studying PRKAA2 activity:

  • Unbiased phosphorylation site identification: Mass spectrometry-based phospho-proteomics can identify not only the canonical Thr172 phosphorylation site but also other, potentially novel phosphorylation sites on PRKAA2 that may regulate its function. This approach can reveal complex regulatory mechanisms beyond what antibody-based methods can detect.

  • Global substrate identification: Phospho-proteomics can identify the full spectrum of proteins phosphorylated in response to AMPK activation, including previously unknown PRKAA2 substrates. This is particularly valuable given the substrate specificity of PRKAA2 compared to PRKAA1 .

  • Quantitative dynamics: Modern phospho-proteomics techniques (such as SILAC, TMT, or label-free quantification) provide quantitative data on hundreds to thousands of phosphorylation sites simultaneously, allowing researchers to place PRKAA2 activity within the broader signaling network.

  • Modification crosstalk detection: Beyond phosphorylation, mass spectrometry can detect other post-translational modifications on PRKAA2 or its substrates, revealing potential crosstalk between phosphorylation and other modifications like acetylation, ubiquitination, or SUMOylation.

  • Targeted approaches: Parallel reaction monitoring (PRM) or multiple reaction monitoring (MRM) mass spectrometry can provide highly sensitive and specific quantification of PRKAA2 Thr172 phosphorylation without antibody limitations like cross-reactivity.

  • Validation strategy: Phospho-proteomics findings can guide the selection of phospho-specific antibodies for routine use, while antibody-based methods can validate key phospho-proteomics discoveries in larger sample sets.

  • Motif analysis: Phospho-proteomics data can reveal the consensus phosphorylation motifs of AMPK in different contexts, helping to distinguish PRKAA1 versus PRKAA2 substrate preferences.

When implementing phospho-proteomics approaches, researchers should consider experimental design (biological replicates, appropriate controls), sample preparation optimization (phosphopeptide enrichment methods), and bioinformatics analysis strategies tailored to their specific research questions.

What are the emerging roles of PRKAA2 in immune system regulation and cancer immunotherapy?

Recent research has uncovered unexpected and significant roles for PRKAA2 in immune regulation with important implications for cancer immunotherapy:

  • Immune cell proportion modulation: Studies have demonstrated that tumors with high PRKAA2 expression contain significantly lower proportions of immune cells compared to tumors with low PRKAA2 expression (43.8% vs. 75.84%) . This suggests PRKAA2 may contribute to creating an "immune cold" tumor microenvironment resistant to immunotherapy.

  • MHC-I expression regulation: PRKAA2 has been implicated in the inhibition of major histocompatibility complex class I (MHC-I) expression through modulation of interferon-gamma activity in malignant cells . This represents a novel mechanism by which cancer cells might evade immune surveillance, as reduced MHC-I expression limits antigen presentation to cytotoxic T cells.

  • T-cell exhaustion promotion: High PRKAA2 expression has been associated with promotion of CD8+ T-cell exhaustion and formation of CD4+ regulatory T cells (Tregs) . T-cell exhaustion is characterized by decreased effector function and expression of inhibitory receptors, while Tregs actively suppress anti-tumor immune responses.

  • Interferon signaling suppression: Gene set enrichment analyses have revealed that IFN-γ response pathways are significantly downregulated in malignant cells with high PRKAA2 expression . As interferon signaling is crucial for anti-tumor immunity, this suggests another mechanism by which PRKAA2 might promote immune evasion.

  • Potential therapeutic implications: These findings suggest that targeting PRKAA2 might have dual benefits in cancer treatment: direct effects on cancer cell metabolism and indirect effects on enhancing anti-tumor immunity. Combining PRKAA2 inhibitors with immune checkpoint inhibitors could potentially overcome resistance to immunotherapy in "immune cold" tumors.

These emerging roles position PRKAA2 at the intersection of metabolism and immunity, highlighting the complex interplay between these fundamental biological processes in cancer progression.

How does the application of Phospho-PRKAA2 (Thr172) antibodies advance our understanding of AMPK-mediated autophagy?

Phospho-PRKAA2 (Thr172) antibodies have become instrumental in elucidating the complex relationship between AMPK activation and autophagy regulation:

  • Subcellular localization studies: Immunofluorescence applications using Phospho-PRKAA2 (Thr172) antibodies (at dilutions of 1:20 to 1:200) enable researchers to visualize the dynamic localization of activated AMPK during autophagy induction. Recent research has highlighted a potential role for N-myristoylation of the β subunits in localizing AMPK to mitochondrial membranes during mitophagy .

  • Temporal dynamics of activation: Western blot analysis with Phospho-PRKAA2 (Thr172) antibodies allows researchers to track the timing of AMPK activation relative to autophagy induction, helping establish cause-effect relationships and distinguish between early regulatory events and feedback mechanisms.

  • Isoform-specific contributions: By specifically tracking PRKAA2 phosphorylation rather than total AMPK activity, researchers can determine the distinct contributions of this catalytic subunit to autophagy regulation, especially in tissues where both PRKAA1 and PRKAA2 are expressed. Recent research suggests PRKAA2 may have connections to selective autophagy processes .

  • Target protein interactions: Combined with co-immunoprecipitation and proximity ligation assays, phospho-specific antibodies help identify direct interactions between activated PRKAA2 and autophagy-related proteins, revealing mechanistic details of how AMPK signaling is translated to autophagy machinery activation.

  • Quantitative assessment in disease models: These antibodies enable quantification of PRKAA2 activation status in various disease models where autophagy dysregulation is implicated, helping establish connections between metabolic stress, AMPK activation, and autophagy responses in pathological contexts.

  • BCL2L13 phosphorylation: Recent studies have begun to explore the relationship between PRKAA2 and BCL2L13 phosphorylation in the context of autophagy activation, suggesting new mechanistic connections in selective autophagy pathways .

By providing tools to specifically track the active form of PRKAA2, these antibodies continue to advance our understanding of the nuanced regulatory mechanisms connecting energy sensing to autophagy induction.

What are the technical challenges in developing next-generation phospho-specific antibodies for PRKAA2?

The development of improved phospho-specific antibodies for PRKAA2 faces several technical challenges that researchers and manufacturers must address:

  • Isoform specificity optimization: The high sequence homology between PRKAA1 and PRKAA2 around the Thr172 phosphorylation site makes developing truly isoform-specific antibodies technically challenging. Next-generation approaches may include longer epitope recognition sequences or targeting regions that contain subtle isoform differences adjacent to the phosphorylation site.

  • Conformational state recognition: AMPK undergoes significant conformational changes upon activation, with the α-linker region playing a crucial role in exposing or protecting Thr172 . Developing antibodies that can reliably detect phospho-Thr172 regardless of these conformational states, or alternatively, creating conformation-specific antibodies, presents significant challenges.

  • Context-dependent phosphorylation: PRKAA2 can be phosphorylated at Thr172 by different upstream kinases (LKB1, CAMKK2) in different contexts . These kinases may induce subtle differences in the local structure around phospho-Thr172 or in accompanying phosphorylation events, potentially affecting epitope recognition.

  • Quantitative accuracy improvements: Current antibodies may have limited linear dynamic range for quantification. Developing calibrated antibody systems with known response characteristics would improve quantitative accuracy across laboratories.

  • Multiplex capability development: Creating antibody formats compatible with multiplex detection systems would allow simultaneous measurement of phospho-PRKAA2 alongside other relevant phospho-proteins in signaling networks.

  • Improved compatibility with fixed tissues: Developing phospho-specific antibodies that maintain specificity in formalin-fixed, paraffin-embedded tissues would expand their utility in clinical research and diagnostics.

  • Recombinant technology standardization: While recombinant monoclonal technology offers advantages in consistency , standardizing production and validation protocols across manufacturers remains challenging.

Addressing these challenges will require innovative approaches combining advances in antibody engineering, epitope design, and validation methodologies to create next-generation reagents with improved specificity, sensitivity, and reliability.

How can computational modeling enhance our interpretation of PRKAA2 phosphorylation data?

Computational modeling approaches can significantly enhance the interpretation of experimental data on PRKAA2 phosphorylation through several innovative strategies:

  • Structural modeling of phosphorylation effects: Molecular dynamics simulations can predict how Thr172 phosphorylation affects the three-dimensional structure and dynamics of PRKAA2 within the heterotrimeric complex. These models can help interpret how binding of AMP to the γ subunit induces conformational changes that affect the α-linker region and the accessibility of Thr172 .

  • Network-based interpretation: Pathway and network analysis tools can place PRKAA2 phosphorylation in the context of broader signaling networks. This is particularly valuable for understanding how PRKAA2 connects to distinct downstream pathways in different tissues or disease states, such as the correlation between PRKAA2 and CRTC2, TNNI3, KCNA5, and TFEB expression .

  • Machine learning for biomarker development: Machine learning algorithms can identify patterns in multi-dimensional data that correlate PRKAA2 phosphorylation status with disease outcomes or treatment responses. This could help identify patient subgroups most likely to benefit from AMPK-targeting therapies.

  • Dynamic modeling of phosphorylation cycles: Ordinary differential equation (ODE) models can capture the temporal dynamics of PRKAA2 phosphorylation/dephosphorylation cycles, including the three mechanisms by which AMP binding activates AMPK: allosteric activation, promotion of Thr172 phosphorylation, and inhibition of Thr172 dephosphorylation .

  • Multi-scale modeling approaches: Combining molecular, cellular, and tissue-level models can help translate molecular findings (e.g., PRKAA2 phosphorylation) to physiological or pathological outcomes (e.g., metabolic responses, tumor immune evasion).

  • In silico prediction of substrate specificity: Computational tools can predict potential PRKAA2-specific substrates based on consensus phosphorylation motifs and protein-protein interaction data, generating hypotheses for experimental validation.

  • Integration of multi-omics data: Computational frameworks can integrate phospho-proteomics data with transcriptomics, metabolomics, and other datasets to provide a systems-level view of how PRKAA2 phosphorylation influences cellular phenotypes.

By leveraging these computational approaches, researchers can extract deeper biological insights from experimental data on PRKAA2 phosphorylation and generate testable hypotheses for further investigation.

What are the key considerations for researchers selecting Phospho-PRKAA2 (Thr172) antibodies for their studies?

When selecting Phospho-PRKAA2 (Thr172) antibodies for research applications, investigators should carefully consider several critical factors to ensure reliable and interpretable results:

  • Validation documentation: Prioritize antibodies with comprehensive validation data demonstrating specificity for phosphorylated PRKAA2 at Thr172. Ideal validation would include evidence from multiple techniques and controls such as PRKAA2 knockout/knockdown samples and phosphatase-treated controls .

  • Application compatibility: Select antibodies validated for your specific application, whether Western blot (typically at dilutions of 1:500-1:5000), immunofluorescence (1:20-1:200), or ELISA . Performance can vary significantly between applications, so application-specific validation is crucial.

  • Isoform specificity: Consider whether the antibody distinguishes between phosphorylated PRKAA1 and PRKAA2, which have high sequence homology around the Thr172 site. This is particularly important in tissues or cells expressing both isoforms.

  • Clonality considerations: Recombinant monoclonal antibodies like clone 4F4 offer advantages in batch-to-batch consistency compared to polyclonal alternatives, which can be important for longitudinal studies.

  • Species reactivity: Confirm that the antibody recognizes your species of interest. While many antibodies are raised against human sequences , cross-reactivity with other model organisms should be experimentally verified.

  • Storage and handling requirements: Follow manufacturer recommendations for storage (-20°C or -80°C) and aliquoting to avoid repeated freeze-thaw cycles that can compromise antibody performance .

  • Blocking considerations: For phospho-specific antibodies, BSA-based blocking solutions are generally preferred over milk-based alternatives, which contain phosphoproteins that can interfere with detection.

  • Context-dependent optimization: Recognize that optimal antibody conditions may vary with experimental context. Factors like cellular stress, fixation methods, and protein extraction protocols can all affect epitope accessibility and antibody performance.

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.