p38a/SAPK2 Human

p38a/SAPK2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction

p38a/SAPK2, a serine/threonine kinase, is the most studied isoform of the p38 mitogen-activated protein kinase (MAPK) family. It plays a central role in cellular responses to environmental stressors, inflammation, and developmental signals. Originally identified as a 38 kDa protein phosphorylated in response to endotoxin and osmotic stress, p38α has been implicated in diverse physiological and pathological processes, including cytokine production, apoptosis, and cytoskeletal remodeling. Its dual role in promoting survival or cell death underscores its complexity in disease contexts .

Activation Pathways

p38α is activated via dual phosphorylation at Thr180 and Tyr182 by upstream kinases:

  1. MKK3/MKK6: Primary activators under stress, cytokine, or growth factor signals.

  2. Autophosphorylation: Induced by TAB1 binding under osmotic stress .

Key Substrates and Functions

SubstrateFunctionPathway
ATF2Transcriptional regulation of stress-response genesStress signaling
MEF2CCardiac and muscle differentiationDevelopmental regulation
CDC25BCell cycle regulation (G2/M checkpoint)Proliferation control
p53DNA damage response and apoptosis inductionGenotoxic stress response
HSP27Actin cytoskeleton remodeling (via MK2-mediated phosphorylation)Cytoskeletal dynamics

p38α also regulates post-transcriptional processes, including mRNA stability and translation, through targets like MK2 and MK3 .

Stress Response and Inflammation

p38α is a key mediator of proinflammatory cytokines (e.g., TNF-α, IL-6) and enzymes (e.g., COX-2, iNOS) in immune cells. In human models, p38 inhibition (e.g., BIRB 796 BS) reduces LPS-induced TNF-α release and neutrophil activation .

Apoptosis

p38α exhibits context-dependent roles:

  • Pro-apoptotic: Upregulates Bax, Fas, and FasL; inactivates survival kinases (e.g., ERK, Akt) .

  • Pro-survival: Enhances Bcl-2 and Bcl-xL expression in specific tissues (e.g., lung, keratinocytes) .

Cytoskeletal Dynamics

p38α/MK2 signaling phosphorylates HSP27, promoting actin polymerization and membrane blebbing. This mechanism is observed in oxidative stress-induced toxicity .

Pathological Implications

Diseasep38α RoleEvidence
CancerTumor progression (e.g., via p53 regulation) or suppression (e.g., Bax upregulation)Context-dependent; inhibits survival pathways in cardiomyocytes .
Autoimmune DisordersRegulates T-cell activation and cytokine release (e.g., IL-1β, TNF-α)Inhibitors reduce inflammation in rheumatoid arthritis models .
NeurodegenerationContributes to Alzheimer’s pathology via inflammatory cytokine productionLinked to amyloid-β-induced neuronal damage .

Inhibitors and Challenges

InhibitorMechanismClinical Status
SB203580Competitive ATP-bindingPreclinical (limited efficacy in mice)
BIRB 796 BSNon-ATP competitiveAttenuates human LPS-induced inflammation
MK2 InhibitorsBlocks downstream signalingReduces cytokine synthesis (e.g., TNF-α)

Dual Roles: p38α inhibition may paradoxically enhance survival in some contexts (e.g., ischemia-reperfusion injury), necessitating isoform-specific targeting .

Detection and Purification

  • Phosphorylation Assays: Western blot (Thr180/Tyr182 antibodies) or kinase activity assays .

  • Recombinant Protein: Purified to >95% via SDS-PAGE; molecular mass = 42.7 kDa (activated form) .

Functional Studies

  • Cytokine Induction: IL-1α or TNF-α treatment induces p38α phosphorylation, peaking at 30–60 minutes .

  • Apoptosis Models: Cisplatin or H₂O₂ triggers p38α-dependent Bax translocation and blebbing .

Key Studies

  1. Human LPS Model: BIRB 796 BS reduces TNF-α, IL-6, and neutrophil activation, validating p38α as a therapeutic target .

  2. MK2-Deficient Mice: Impaired cytokine synthesis and neutrophil migration highlight p38α’s role in inflammation .

  3. Cancer Context: p38α deletion enhances ERK/Akt survival pathways, suggesting tumor-suppressive potential .

Product Specs

Introduction
p38a/SAPK2, a member of the MAP kinase family, plays a crucial role in cellular responses to various stimuli. This kinase integrates multiple biochemical signals and participates in essential processes like cell proliferation, differentiation, and transcriptional regulation. Activated by environmental stresses and pro-inflammatory cytokines, p38a/SAPK2's activation is facilitated by phosphorylation via MAP kinase kinases (MKKs) or autophosphorylation triggered by interaction with the MAP3K7IP1/TAB1 protein. Its significant role in stress response and cell cycle regulation is evident through its substrates, which include transcription factors like ATF2, MEF2C, and MAX, cell cycle regulator CDC25B, and the tumor suppressor p53. Furthermore, four alternatively spliced transcript variants of this gene have been identified, encoding distinct isoforms.
Description
p38/SAPK2 is a non-glycosylated polypeptide with a molecular mass of 42.7 kDa, generated by phosphorylating purified p38 alpha using MKK6.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
p38/SAPK2 is supplied in a buffer containing 25mM Tris-HCl, 150mM NaCl, 1mM DTT, and 50% glycerol, at a pH of 8.5.
Unit Definition
The product exhibits no detectable protease activity. Its specific activity is determined to be 2,357,900 units per milligram, where one unit is defined as one picomole of phosphate transferred to myelin basic protein per minute at a temperature of 30 degrees Celsius.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4 degrees Celsius. For long-term storage, it is recommended to store the product frozen at -20 degrees Celsius. To maintain product integrity, avoid repeated freeze-thaw cycles.
Purity
Analysis by SDS-PAGE indicates a purity greater than 95%.
Synonyms
Mitogen-activated protein kinase 14, EC 2.7.11.24, Mitogen-activated protein kinase p38 alpha, MAP kinase p38 alpha, Cytokine suppressive anti-inflammatory drug-binding protein, CSAID-binding protein, CSBP, MAX-interacting protein 2, MAP kinase MXI2, SAPK2A, RK, p38, EXIP, Mxi2, CSBP1, CSBP2, CSPB1, PRKM14, PRKM15, p38ALPHA.
Source
Escherichia Coli.

Q&A

What is the structural organization of p38α/SAPK2 and how does it differ from other MAPK family members?

p38α (MAPK14/SAPK2a) is a proline-directed serine/threonine kinase of the MAPK family. Unlike ERK1/2 which primarily respond to mitogens, p38α is activated by environmental stresses and inflammatory signals. The protein consists of several domains that facilitate its function as a stress-responsive kinase. The ATP-binding pocket and substrate docking sites are particularly important for kinase activity .

Structurally, p38α shares more than 60% amino acid sequence identity with other p38 family members (p38β, p38γ, and p38δ). It has 75% identity with p38β specifically, while p38γ and p38δ are more closely related to each other (75% identity). These structural similarities and differences underlie their functional divergence, with p38α being the only family member essential for mouse embryonic development .

Methodologically, when investigating p38α structure-function relationships, researchers should consider using techniques such as X-ray crystallography, molecular dynamics simulations, or cryo-electron microscopy to elucidate structural determinants of activation and substrate specificity.

What are the key experimental models used to study p38α/SAPK2 signaling in human systems?

When designing experiments to study p38α signaling, researchers typically employ multiple complementary approaches:

  • Cell line models: Human cell lines with relevant physiological characteristics (e.g., macrophages for inflammation studies, cardiomyocytes for cardiac stress research)

  • Primary human cells: Providing more physiologically relevant contexts than immortalized cell lines

  • Genetic manipulation approaches: CRISPR/Cas9, siRNA, and shRNA for knockdown/knockout studies; overexpression systems for gain-of-function experiments

  • Chemical probe strategies: Using selective inhibitors like SB203580 for pharmacological inhibition while noting its selectivity profile (inhibits p38α and p38β but not p38γ or p38δ)

  • Patient-derived samples: For translational relevance across various pathological conditions

For rigorous experimental design, it's important to validate findings across multiple model systems and include appropriate controls for pathway specificity.

How do canonical and non-canonical activation pathways of p38α/SAPK2 differ in experimental settings?

p38α can be activated through three distinct mechanisms, each requiring specific experimental approaches for accurate investigation:

Canonical Activation Pathway:

  • Involves the classical MAP3K→MAP2K→MAPK cascade

  • MAP2Ks (primarily MKK3 and MKK6) phosphorylate p38α on both Thr180 and Tyr182 in the activation loop

  • Experimental detection: Dual phospho-specific antibodies (pT180/pY182) are standard tools

Non-canonical Pathway 1 (TAB1-mediated):

  • Involves binding to TAB1 (TGF-β-activated kinase 1-binding protein 1)

  • Induces p38α autophosphorylation independent of upstream MAP2Ks

  • Critical in specific contexts: myocardial ischemia, T cell senescence, skin inflammation

  • Experimental detection: Co-immunoprecipitation with TAB1 alongside phosphorylation assays

Non-canonical Pathway 2 (TCR-specific):

  • Operates specifically in T cells stimulated through TCR

  • Involves ZAP70-mediated phosphorylation on Tyr323

  • Results in preferential autophosphorylation on Thr180 (mono-phosphorylated p38α)

  • Experimental detection: Phospho-Tyr323 specific antibodies and analysis of mono-phosphorylated vs. dual-phosphorylated states

When designing experiments, researchers should carefully consider which activation mechanism is likely operational in their system of interest, as this affects both the choice of activation stimuli and detection methods.

What methodologies are most effective for measuring real-time p38α/SAPK2 activation dynamics in living cells?

For investigating the temporal dynamics of p38α activation, several complementary approaches are recommended:

  • FRET-based biosensors: Fluorescence resonance energy transfer sensors allow real-time visualization of p38α activity in living cells with subcellular resolution

  • Phospho-flow cytometry: Enables quantitative single-cell analysis of p38α phosphorylation states in heterogeneous cell populations

  • Time-resolved Western blotting: Traditional approach using phospho-specific antibodies at multiple time points following stimulation

  • Mass spectrometry-based phosphoproteomics: Provides comprehensive analysis of phosphorylation events within the pathway and downstream targets

  • Live-cell imaging with phospho-specific antibody fragments: For spatial and temporal resolution of activation

Each method has distinct advantages and limitations. For instance, while Western blotting provides population-averaged data, FRET biosensors can reveal cell-to-cell variability and oscillatory behavior in p38α signaling, which has been shown to be important for pro-inflammatory gene expression and stress-induced cell death responses .

What experimental approaches best elucidate the dynamics of p38α/SAPK2 signal termination?

Signal termination is critical for preventing deleterious effects of p38α hyperactivation. To investigate termination mechanisms, researchers should consider these methodological approaches:

  • Phosphatase activity assays: Multiple phosphatases can inactivate p38α, including:

    • Serine/threonine phosphatases

    • Tyrosine phosphatases

    • Dual-specificity phosphatases (DUSPs/MKPs)

  • Negative feedback loop analysis: Several feedback mechanisms control p38α activity:

    • DUSP1 induction by p38α signaling creates negative feedback

    • p38α-mediated limitation of MKK6 expression

    • Tracking these dynamics requires time-course experiments with simultaneous measurement of p38α activity and negative regulator expression

  • Post-translational modification profiling: Beyond phosphorylation, monitor:

    • Acetylation (e.g., Lys53 that enhances activity)

    • Proline isomerization (e.g., Pro224 that facilitates phosphorylation)

    • Arginine methylation (that promotes specific functions)

  • Protein-protein interaction studies: Identify regulatory binding partners using:

    • Co-immunoprecipitation

    • Proximity labeling

    • Fluorescence correlation spectroscopy

The table below summarizes key post-translational modifications regulating p38α activity:

ModificationSiteEffect on ActivityDetection Method
PhosphorylationThr180/Tyr182ActivationPhospho-specific antibodies
PhosphorylationTyr323Alternative activationPhospho-Y323 antibodies
AcetylationLys53Enhanced activity via ATP bindingAcetyl-lysine antibodies, mass spectrometry
IsomerizationPro224Facilitates MAP2K phosphorylationMass spectrometry, conformation-specific antibodies
MethylationArginine residuesPromotes specific functionsMethyl-arginine antibodies, mass spectrometry

How can researchers accurately distinguish between p38α/SAPK2 pathway-specific effects and parallel stress response pathways?

Distinguishing p38α-specific effects from other stress-responsive pathways presents significant challenges due to extensive crosstalk. Methodologically sound approaches include:

  • Pharmacological inhibitor profiles: Use multiple structurally distinct inhibitors with well-characterized selectivity profiles:

    • SB203580 inhibits p38α and p38β but not p38γ or p38δ

    • BIRB796 has broader p38 isoform coverage

    • Control experiments should include JNK and ERK pathway inhibitors to rule out crosstalk effects

  • Genetic manipulation strategies:

    • CRISPR/Cas9 knockout of p38α specifically

    • Rescue experiments with wild-type vs. kinase-dead p38α

    • Isoform-specific knockdown of all p38 family members individually

    • Analysis of compound genetic models (e.g., p38α/JNK double knockouts)

  • Substrate phosphorylation patterns:

    • Monitor phosphorylation of established p38α-specific substrates (e.g., MK2, MK3)

    • Distinguish from JNK substrates (c-Jun) and ERK substrates (RSK)

    • Use phospho-motif specific antibodies targeting the p38α consensus sequence

  • Context-dependent activation:

    • Design experiments with stimuli showing differential activation of stress pathways

    • Time-course analyses to detect temporal differences in pathway activation

Careful experimental design incorporating these approaches can help attribute biological effects specifically to p38α rather than parallel stress pathways.

How does p38α/SAPK2 function differ across tissue types and what experimental designs best capture this tissue specificity?

p38α shows remarkable context specificity across tissues, requiring tailored experimental approaches:

  • Tissue-specific expression patterns:

    • p38α is ubiquitously expressed, but at varying levels

    • p38α functions differently depending on the tissue-specific expression of:

      • Upstream activators

      • Scaffold proteins

      • Substrates

      • Phosphatases

  • Tissue-specific experimental systems:

    • Primary cell isolation from different human tissues

    • Tissue-specific conditional knockout mouse models

    • Organoid/3D culture systems recapitulating tissue architecture

    • Tissue-specific expression of p38α biosensors in vivo

  • Specific physiological roles by tissue type:

    • Placenta: Essential for embryonic development and placental morphogenesis

    • Immune cells: Critical for inflammatory responses and cytokine production

    • Cardiomyocytes: TAB1-mediated activation during myocardial ischemia

    • Adipose tissue: Triiodothyronine-mediated browning of white adipose tissue

    • T cells: TCR-specific activation mechanism affecting T cell function

Researchers should design tissue-specific experimental models that accurately recapitulate the cellular environment, signaling networks, and physiological stressors relevant to their tissue of interest.

What methodological approaches best elucidate the dual roles of p38α/SAPK2 in both promoting and suppressing tumor development?

p38α exhibits context-dependent roles in cancer, functioning as both a tumor suppressor and tumor promoter. Methodologically sound investigations require:

  • Cancer type-specific approaches:

    • Analysis across multiple cancer types and stages

    • Correlation with patient outcome data

    • Integration with genomic and proteomic cancer datasets

  • Dual-function experimental designs:

    • Early vs. late stage cancer models

    • Acute vs. chronic p38α inhibition/activation

    • Cell-autonomous vs. non-cell-autonomous effects (e.g., immune cell interaction)

  • Substrate-specific analysis:

    • Identification of context-specific p38α substrates in different tumor types

    • Determination of substrate phosphorylation status in patient samples

    • Correlation of specific substrate activation with disease progression

  • Systems biology approaches:

    • Network analysis of p38α signaling in different tumors

    • Mathematical modeling of pathway dynamics

    • Integration of multi-omics data to identify context-specific patterns

The divergent roles of p38α in cancer highlight the importance of considering disease stage, tissue context, and the specific cellular processes being regulated when designing experiments and interpreting results.

What are the most sensitive approaches for detecting low-level p38α/SAPK2 activation in primary human samples?

Detecting subtle p38α activation in limited primary human samples presents technical challenges. Advanced methodologies include:

  • Digital ELISA technologies:

    • Single molecule array (Simoa) for ultrasensitive protein detection

    • Can detect femtomolar concentrations of phosphorylated p38α

    • Requires minimal sample input

  • Mass cytometry (CyTOF):

    • Metal-tagged antibodies for single-cell profiling

    • Simultaneous detection of multiple phosphorylation sites

    • No spectral overlap issues compared to flow cytometry

    • Preserves rare cell populations

  • Proximity ligation assay (PLA):

    • In situ detection of protein interactions and modifications

    • Amplification step increases sensitivity

    • Allows visualization of p38α activation in fixed tissue sections

  • Nano-immunoassay platforms:

    • Capillary-based immunoassays with enhanced sensitivity

    • Can distinguish between mono- and dual-phosphorylated p38α forms

    • Requires minimal sample input

  • Phosphoproteomics with enrichment strategies:

    • Titanium dioxide or immunoaffinity enrichment

    • Targeted mass spectrometry (MRM/PRM)

    • Data-independent acquisition methods

These approaches should be selected based on sample availability, preservation method, required sensitivity, and whether spatial information is needed.

How can researchers effectively distinguish between the functions of different p38 family members in experimental systems?

Distinguishing between p38 isoforms requires careful experimental design:

  • Isoform-selective chemical probes:

    • SB203580 inhibits p38α and p38β but not p38γ or p38δ

    • Skepinone-L offers improved selectivity for p38α

    • Always validate selectivity in your specific system

  • Genetic manipulation strategies:

    • CRISPR/Cas9 knockout of individual isoforms

    • Isoform-specific siRNA/shRNA approaches

    • Rescue experiments with individual isoforms

    • Analysis of compound genetic models (e.g., p38γ/p38δ double knockouts)

  • Isoform-specific substrates:

    • Identify and monitor phosphorylation of selective substrates

    • Design peptide arrays with isoform-selective sequences

    • Develop biosensors with isoform-specific substrate sequences

  • Expression analysis:

    • p38α and p38β are ubiquitously expressed (p38α usually at higher levels)

    • p38γ and p38δ have more tissue-specific expression patterns

    • Quantify relative isoform levels in your specific system

  • Functional compensation analysis:

    • Monitor potential upregulation of other isoforms upon p38α downregulation

    • Evidence suggests p38α may negatively regulate other p38 kinases

    • Design experiments to detect functional redundancy vs. distinct functions

These approaches help delineate isoform-specific functions while accounting for potential compensatory mechanisms between family members.

What strategies can address the recurring failure of p38α/SAPK2 inhibitors in clinical trials despite promising preclinical data?

The disconnect between preclinical success and clinical trial outcomes for p38α inhibitors presents a significant research challenge. Methodological strategies to address this include:

  • Improved preclinical models:

    • Humanized mouse models

    • Patient-derived xenografts

    • Organ-on-chip technologies

    • Ex vivo human tissue culture systems

  • Pathway compensation analysis:

    • Systematic profiling of compensatory pathway activation

    • Analysis of temporal dynamics following p38α inhibition

    • Multi-targeting approaches (e.g., combined p38α and JNK inhibition)

  • Selective targeting approaches:

    • Substrate-selective inhibitors that block specific downstream functions

    • Context-selective inhibitors that operate only under specific cellular conditions

    • Targeted protein degradation approaches (PROTACs) for p38α

  • Biomarker-driven patient stratification:

    • Identify patient subsets with hyperactive p38α signaling

    • Develop companion diagnostics to predict treatment response

    • Monitor pathway activity during treatment to detect resistance mechanisms

  • Consideration of p38α's dual roles:

    • Design trials accounting for potential pro-tumorigenic and anti-tumorigenic roles

    • Timing-dependent p38α inhibition strategies based on disease stage

    • Cell type-specific targeting approaches

These approaches may help reconcile the discrepancy between promising preclinical data and clinical trial outcomes.

What experimental approaches can best elucidate the mechanisms underlying oscillatory p38α/SAPK2 signaling patterns and their functional significance?

Oscillatory p38α signaling has been observed in contexts such as pro-inflammatory gene expression and stress-induced cell death . Investigating these dynamic patterns requires specialized approaches:

  • High-temporal resolution techniques:

    • FRET-based biosensors with automated imaging

    • Microfluidic systems for precise control of stimulation timing

    • Optogenetic tools for temporal control of pathway activation

  • Single-cell analysis platforms:

    • Live-cell imaging of pathway activity in individual cells

    • Single-cell RNA-seq at multiple time points following stimulation

    • Flow cytometry with phospho-specific antibodies

    • Mass cytometry (CyTOF) for comprehensive signaling analysis

  • Mathematical modeling approaches:

    • Ordinary differential equation models of the signaling network

    • Stochastic modeling to capture cell-to-cell variability

    • Parameter estimation from experimental data

    • Sensitivity analysis to identify key regulatory nodes

  • Perturbation strategies:

    • Targeted disruption of feedback loops (e.g., DUSP1 knockout)

    • Controlled modulation of input signal frequency

    • Pharmacological or genetic manipulation of oscillation frequency

  • Functional output correlation:

    • Single-cell correlation between oscillation patterns and cellular outcomes

    • Modulation of oscillation frequency and amplitude to determine functional impact

    • Comparison across different cell types with distinct oscillatory behaviors

Understanding oscillatory signaling may reveal how p38α achieves context-specific responses despite its involvement in numerous cellular processes.

What control experiments are essential when investigating p38α/SAPK2 signaling to ensure reliable and reproducible results?

Rigorous experimental design for p38α research requires comprehensive controls:

  • Phosphorylation state controls:

    • Both positive controls (strong activators like anisomycin or sorbitol)

    • Negative controls (unstimulated or inhibitor-treated)

    • Time-course analysis to capture transient activation

    • Controls for phosphatase inhibition during sample preparation

  • Inhibitor specificity controls:

    • Use of structurally distinct inhibitors

    • Dose-response experiments

    • Inactive analogs as negative controls

    • Rescue experiments with inhibitor-resistant p38α mutants

    • Monitor off-target effects on parallel pathways

  • Genetic manipulation controls:

    • Include non-targeting siRNA/shRNA controls

    • Rescue experiments with wild-type p38α

    • Control for compensatory upregulation of other p38 isoforms

    • Multiple independent siRNA/shRNA sequences

  • Substrate specificity controls:

    • In vitro kinase assays with purified components

    • Mutation of predicted phosphorylation sites

    • Analysis of phosphorylation in p38α-knockout cells

  • Biological relevance controls:

    • Multiple cell types/tissues

    • Different activation stimuli

    • Physiologically relevant concentrations and time frames

    • Correlation with functional outcomes

Implementing these controls systematically enhances reproducibility and reliability of p38α signaling research.

How can researchers resolve contradictory findings about p38α/SAPK2 functions across different experimental systems and disease models?

Contradictory findings in p38α research often stem from context-dependent functions. Methodological approaches to resolve these include:

  • Systematic meta-analysis:

    • Comprehensive literature review with formal meta-analysis

    • Identification of patterns in contradictory findings

    • Analysis of experimental variables correlating with divergent outcomes

  • Standardized experimental comparisons:

    • Direct side-by-side comparison using identical readouts

    • Controlled variation of single experimental parameters

    • Development of standard operating procedures for p38α research

  • Context-specific analysis:

    • Detailed characterization of baseline pathway activity

    • Analysis of compensatory mechanisms in different systems

    • Mapping of the complete signaling network in each system

    • Multi-omics profiling to identify system-specific features

  • Integrative experimental approaches:

    • Parallel in vitro, ex vivo, and in vivo experiments

    • Consistent methodologies across experimental systems

    • Translation between animal models and human samples

    • Cross-validation in multiple independent laboratories

  • Advanced statistical and computational methods:

    • Bayesian analysis frameworks

    • Machine learning approaches to identify patterns

    • Sensitivity analysis to identify critical variables

    • Systems biology modeling of context-dependent effects

This multilayered approach can help resolve seemingly contradictory findings by identifying the specific contexts in which different p38α functions predominate.

Product Science Overview

Introduction

p38a, also known as Stress-Activated Protein Kinase 2 (SAPK2), is a member of the p38 MAPK family. This family of kinases plays a crucial role in cellular responses to stress and inflammation. The p38 MAPK pathway is highly conserved across species and is involved in converting extracellular stimuli into a wide range of cellular responses .

Structure and Function

p38a/SAPK2 is a non-glycosylated polypeptide with a molecular mass of approximately 42.7 kDa . It is produced by phosphorylation of the purified p38 alpha with MKK6. The protein is relatively inactive in its non-phosphorylated form and becomes rapidly activated by dual phosphorylation of a Thr-Gly-Tyr motif .

Biological Role

The p38 MAPK pathway, including p38a/SAPK2, is known for its role in transducing stress signals from the environment. It regulates various cellular activities, including inflammation, cell differentiation, and apoptosis . When cells are exposed to stressors such as tumor necrosis factor, interleukin-1, or heat shock, the activation of MAPK kinase-3/6 occurs by phosphorylation. This, in turn, phosphorylates each residue of Thr180 and Tyr182 in p38 MAPK, leading to its activation .

Applications in Research and Medicine

Human recombinant p38a/SAPK2 proteins are widely used in kinase assays and other research applications. These proteins are typically expressed in E. coli and are available in various product grades . The p38 MAPK pathway has been extensively studied for its role in cancer, with p38a/SAPK2 functioning both as a tumor suppressor and a tumor promoter . This dual nature has led to the development of specific inhibitors that are being explored as potential cancer therapies .

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.