MAP2K3 Human

Mitogen-Activated Protein Kinase Kinase 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to MAP2K3 Human

MAP2K3 (mitogen-activated protein kinase kinase 3) encodes a dual-specificity protein kinase critical in stress-response and signaling pathways. It activates p38 MAPK (MAPK14) by phosphorylating threonine and tyrosine residues, mediating cellular responses to cytokines, environmental stress, and oncogenic signals . This kinase is implicated in inflammation, apoptosis, and disease pathologies, including neurodegeneration and cancer .

Molecular Interactions and Pathways

MAP2K3 operates within the p38 MAPK cascade, interacting with upstream regulators and downstream effectors. Key partners include:

Interaction PartnerRoleEvidence
MAPK14 (p38)Direct substrate; phosphorylation activates stress- and cytokine-responses Co-expression, kinase assays
TAOK2Reciprocal phosphorylation; modulates DNA damage and stress responses Yeast two-hybrid, co-IP
MAP3K5 (ASK1)Upstream activator; integrates oxidative stress and apoptosis signals Genetic interaction studies
CCM2Scaffold protein; stabilizes MAP2K3-MAP3K3 signaling in vascular integrity Knockdown/rescue experiments

Alzheimer’s Disease and Memory

MAP2K3 variants correlate with superior memory in aging populations ("SuperAgers") and reduced AD risk .

  • Mechanism:

    • Microglial Regulation: MAP2K3 expression enriches in microglia, suggesting role in immune surveillance .

    • p38 MAPK Modulation: Lower MAP2K3 activity may reduce neuroinflammation and β-amyloid toxicity .

  • Therapeutic Potential: MAP2K3 inhibitors proposed to enhance cognition and AD resistance .

Glioma Malignancy and Prognosis

MAP2K3 overexpression is linked to glioma aggressiveness and poor survival .

Functional Impact:

  • Cell Proliferation: Silencing MAP2K3 reduces glioma cell viability and invasion .

  • Immune Checkpoint Links: Correlates with PD-1/PD-L1 expression, suggesting immunotherapy potential .

Tissue-Specific Expression Patterns

MAP2K3 exhibits widespread expression, with notable enrichment in brain and immune tissues:

TissueExpression LevelCellular Localization
Cerebral CortexModerateCytoplasmic (glioma cells)
HippocampusLowNotably expressed in microglia
Adrenal GlandHighN/A
LungModerateN/A
Immune OrgansHigh (e.g., lymph nodes)N/A

Data sourced from Human Protein Atlas and glioma-specific studies .

Research Implications and Future Directions

MAP2K3 emerges as a multifaceted therapeutic target:

  1. AD Therapeutics: Inhibitors may mitigate neuroinflammation and preserve cognitive function .

  2. Glioma Therapy: Targeting MAP2K3 could suppress tumor growth and enhance chemosensitivity .

  3. Immune Modulation: Potential to modulate microglial responses in neurodegenerative diseases and cancer .

Gaps and Challenges:

  • Mechanistic Studies: Limited data on isoform-specific functions and signaling crosstalk.

  • Biomarker Validation: Requires protein-level validation in patient cohorts .

Product Specs

Introduction
Dual specificity mitogen-activated protein kinase kinase 3 (MAP2K3) is a dual specificity protein kinase that belongs to the MAP kinase kinase family. It is activated by mitogenic and environmental stress and participates in the MAP kinase-mediated signaling cascade. MAP2K3 phosphorylates and activates MAPK14/p38-MAPK.
Description
Recombinant human MAP2K3, expressed in E. coli, is a single, non-glycosylated polypeptide chain containing 338 amino acids (residues 1-318) with a molecular weight of 38.3 kDa. A 20 amino acid His-tag is fused to the N-terminus. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
Solution containing 1 mg/ml MAP2K3 in 20 mM Tris-HCl buffer (pH 8.0) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, store frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity greater than 90.0% as determined by SDS-PAGE.
Synonyms
Dual specificity mitogen-activated protein kinase kinase 3, MAP kinase kinase 3, MAPKK 3, MAPK/ERK kinase 3, MEK 3, MAP2K3, MEK3, MKK3, PRKMK3.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSKPPAPNPT PPRNLDSRTF ITIGDRNFEV EADDLVTISE LGRGAYGVVE KVRHAQSGTI MAVKRIRATV NSQEQKRLLM DLDINMRTVD CFYTVTFYGA LFREGDVWIC MELMDTSLDK FYRKVLDKNM TIPEDILGEI AVSIVRALEH LHSKLSVIHR DVKPSNVLIN KEGHVKMCDF GISGYLVDSV AKTMDAGCKP YMAPERINPE LNQKGYNVKS DVWSLGITMI EMAILRFPYE SWGTPFQQLK QVVEEPSPQL PADRFSPEFV DFTAQCLRKN PAERMSYLEL MEHPFFTLHK TKKTDIAAFV KEILGEDS.

Q&A

What is MAP2K3 and what are its primary functions in cellular signaling?

MAP2K3 (also known as MKK3) is a member of the dual specificity protein kinase group belonging to the MAP kinase kinase family. It functions as a critical component in the mitogen-activated protein kinase (MAPK) signaling pathway, which mediates cellular responses to various stimuli .

MAP2K3 primarily serves as an intermediary in stress-response signaling cascades. Upon activation by mitogenic or stress-inducing stimuli and inflammatory cytokines, MAP2K3 phosphorylates and activates p38 MAPK, leading to downstream signaling events that influence gene expression, protein synthesis, and cellular adaptation to stress . This activation sequence contributes to numerous cellular programs including differentiation, movement, division, and apoptosis.

Research has demonstrated that MAP2K3 plays essential roles in:

  • Mediating cellular responses to environmental stressors

  • Facilitating inflammatory signaling cascades

  • Supporting cell survival and proliferation mechanisms, particularly in cancer contexts

  • Regulating immune system function, with enriched expression in microglia

  • Participating in signaling pathways associated with cognitive function and neurodegeneration

What techniques are most effective for measuring MAP2K3 expression and activity in human samples?

Researchers employ multiple complementary approaches to assess MAP2K3 expression, activity, and function in human samples:

mRNA Expression Analysis:

  • Quantitative RT-PCR for specific transcript level quantification

  • RNA sequencing for comprehensive transcriptomic profiling

  • Microarray analysis using data from public databases (TCGA, GEO, CGGA)

Protein Expression Analysis:

  • Western blotting with anti-MAP2K3 antibodies to quantify protein levels

  • Immunohistochemistry (IHC) using specific antibodies (e.g., anti-MAP2K3 primary antibody HPA043783) to visualize expression in tissue sections

  • Immunofluorescence staining to determine cellular localization patterns

Functional Activity Assessment:

  • Kinase activity assays using recombinant proteins

  • Phosphorylation state analysis of MAP2K3 downstream substrates (particularly p38 MAPK)

  • RNA interference to study loss-of-function effects on cellular phenotypes

Genetic Characterization:

  • Whole Exome Sequencing (WES) to identify genetic variants

  • Single nucleotide polymorphism (SNP) analysis (e.g., rs2363221, rs2230435, rs736103)

  • Linkage Disequilibrium analysis to understand genetic associations

Pathway Analysis:

  • Gene Set Variation Analysis (GSVA) to identify biological pathways influenced by MAP2K3

  • Single-sample Gene Set Enrichment Analysis (ssGSEA) for scoring oncogenic signaling pathways

When selecting methods, researchers should consider the specific research question, available tissue samples, and whether the focus is on expression levels, functional activity, or genetic variation.

How does MAP2K3 expression vary across different human tissues and disease states?

MAP2K3 demonstrates significant expression variations across tissues and disease conditions, providing important insights into its biological roles:

Tissue-Specific Expression Patterns:

  • Brain tissue: MAP2K3 shows enriched expression in microglia in mouse cortex

  • Differential expression between normal brain tissue and glioma tissues of varying grades

  • Expression levels can be visualized through immunohistochemistry, revealing distinct patterns in normal versus diseased tissues

Expression in Neurological Conditions:

  • Significantly upregulated in the middle temporal gyrus of Alzheimer's disease patients compared to controls

  • Expression patterns correlate with cognitive function in aging populations, with certain genetic variants associated with superior cognitive performance

Cancer-Associated Expression:

  • Elevated expression in various cancer types compared to corresponding normal tissues

  • Expression levels correlate with glioma malignancy and WHO classification, with higher expression in more aggressive tumors

  • Higher expression is associated with poorer prognosis in glioma patients, suggesting potential as a prognostic biomarker

Age-Related Expression Changes:

  • Research in "SuperAgers" (individuals 80+ years with superior memory) suggests specific MAP2K3 genetic variants may influence expression and function across the lifespan

  • These expression variations may contribute to differential cognitive aging trajectories

These tissue-specific and disease-associated expression patterns highlight MAP2K3's diverse roles in physiological and pathological processes, providing rationale for targeted therapeutic approaches in conditions with aberrant MAP2K3 activity.

What are the primary molecular pathways influenced by MAP2K3 activation?

MAP2K3 influences several key molecular pathways that regulate cellular responses to stress, inflammation, and growth signals:

Core Signaling Pathways:

  • p38 MAPK Pathway: MAP2K3 directly phosphorylates and activates p38 MAPK, leading to downstream effects on transcription factors and gene expression that control stress responses .

  • Cell Survival and Proliferation Pathways: MAP2K3 participates in signaling cascades that promote cell proliferation and survival, particularly in cancer contexts .

  • Inflammatory Signaling: MAP2K3 is part of the signaling cascade leading to inflammation, playing a crucial role in immune system regulation, especially in microglia .

  • Apoptotic Pathways: Research shows MAP2K3 depletion can induce endoplasmic reticulum stress and autophagy, affecting both wild-type and mutant p53 proteins and influencing cell death mechanisms .

Cancer-Associated Pathways:
Gene Set Variation Analysis has revealed that higher MAP2K3 expression correlates with elevated activity in several critical pathways:

  • Hippo Pathway: Involved in organ size control and tumor suppression

  • NRF2 Pathway: Regulates antioxidant responses and cancer progression

  • PI3K Pathway: Controls cell growth, proliferation, and survival

  • TGF Pathway: Regulates cell growth, differentiation, and immune functions

These pathways are known to be involved in tumor immune evasion responses, suggesting MAP2K3 may influence cancer progression through multiple mechanisms .

Neurodegenerative Disease Pathways:

  • MAP2K3 participates in signaling cascades associated with beta-amyloid mediated apoptosis, suggesting relevance to Alzheimer's disease mechanisms

  • Its enriched expression in microglia indicates potential roles in neuroinflammatory pathways that contribute to neurodegenerative processes

Understanding these pathway interactions provides crucial insights for developing targeted interventions in conditions with aberrant MAP2K3 activity.

How is MAP2K3 activated in response to cellular stress and inflammatory signals?

MAP2K3 activation occurs through a precise sequence of events in response to specific cellular stimuli:

Activation Mechanism:

  • Exposure to stressful stimuli (oxidative stress, UV radiation) or inflammatory cytokines triggers upstream signaling cascades

  • Activation of upstream MAP kinase kinase kinases (MAP3Ks)

  • MAP3Ks phosphorylate MAP2K3 at specific serine and threonine residues

  • This phosphorylation induces a conformational change in MAP2K3 structure, leading to its activation

  • Activated MAP2K3 subsequently phosphorylates downstream targets, primarily p38 MAPK

Specific Activation Stimuli:

  • Environmental Stressors: Oxidative stress, UV radiation, osmotic shock

  • Inflammatory Cytokines: TNF-α, IL-1

  • Pathogen-Associated Molecular Patterns (PAMPs): Components recognized during infection

  • Damage-Associated Molecular Patterns (DAMPs): Released during cellular damage or stress

Cell-Type Specific Activation:

  • In microglia, MAP2K3 activation occurs in response to inflammatory signals and contributes to neuroinflammatory responses

  • In cancer cells, activation may occur through both stress-response mechanisms and oncogenic signaling pathways

Regulatory Mechanisms:

  • Phosphatases can dephosphorylate and inactivate MAP2K3

  • Scaffold proteins help organize MAP2K3 and its pathway components to ensure signaling specificity

  • Cross-talk with other signaling pathways can modulate MAP2K3 activation

This complex activation system allows for precise control of stress and inflammatory responses, with dysregulation contributing to various pathological conditions.

How does MAP2K3 contribute to cancer progression and metastasis?

MAP2K3 influences cancer progression and metastasis through multiple mechanisms affecting proliferation, survival, therapy resistance, and tumor microenvironment interactions:

Proliferation and Survival Mechanisms:

  • Functions as a transcriptional target of mutant p53, sustaining cell proliferation and survival in cancer cells

  • Depletion of MAP2K3 reduces cancer cell proliferation and viability while having minimal effects on normal cells, suggesting a cancer-specific dependency

  • MAP2K3 depletion induces endoplasmic reticulum stress and autophagy, contributing to cancer cell death

Therapy Resistance Pathways:

  • Contributes to chemotherapy resistance in both wild-type and mutant p53-carrying tumors

  • Targeting MAP2K3 enhances tumor response to chemotherapeutic agents, demonstrated by increased PARP cleavage and reduced clonogenic ability in vitro

  • MAP2K3 depletion reduces tumor growth and improves biological response to chemotherapeutics in vivo, indicating potential for combination therapy approaches

Invasion and Metastasis Promotion:

  • Influences cellular migration and invasion capabilities as demonstrated in wound healing and Transwell migration assays

  • Expression correlates with several chemokines (CXCL10, CCR5, CCR10, CCL5, CCL7, CCR2, and CCL22) that facilitate immune escape and can promote invasive phenotypes

Oncogenic Signaling Pathway Regulation:

  • Higher MAP2K3 expression correlates with elevated activity in Hippo, NRF2, PI3K, and TGF pathways

  • These pathways collectively contribute to tumor progression, therapy resistance, and immune evasion

Prognostic Significance:

  • Higher expression levels associate with poorer prognosis in glioma patients

  • Expression varies with tumor grade, correlating with more aggressive phenotypes

These multifaceted effects on cancer biology position MAP2K3 as a promising target for anticancer strategies, with potential applications in both wild-type and mutant p53-carrying tumors .

What is the relationship between MAP2K3 genetic variants and superior cognitive function in aging?

Research investigating "SuperAgers" (individuals 80+ years old with episodic memory performance at least average for 50-60 year olds) has revealed fascinating associations between MAP2K3 genetic variants and preserved cognitive function:

Genetic Association Findings:

  • Whole Exome Sequencing (WES) identified significant associations between MAP2K3 gene variants and the SuperAging phenotype

  • Three specific SNPs in MAP2K3 contributed to this association:

    • rs2363221 (located in intron 1)

    • rs2230435 (located in exon 5)

    • rs736103 (located in intron 7)

  • SNPs rs2230435 and rs736103 showed high Linkage Disequilibrium (r² = 0.790), suggesting they may be inherited together

Functional Implications of Variants:

  • The identified variants are not predicted to fully impair MAP2K3 activity but may slightly decrease it from birth

  • This subtle alteration is likely critical, as complete loss of MAP2K3 function would be detrimental to normal cellular physiology

  • Researchers hypothesize that decreased MAP2K3 activity results in lowered p38-MAPK activity in neuronal cells and reduced inflammation mediated by microglia

Neurobiological Mechanisms:

  • MAP2K3 resides in a biological pathway linked to memory and participates in signaling cascades associated with beta-amyloid mediated apoptosis

  • Its enriched expression in microglia, the resident immune cells of the brain, suggests a role in brain immune system regulation that may influence cognitive aging

  • The gene is significantly upregulated in the middle temporal gyrus of Alzheimer's disease patients compared to controls, suggesting its involvement in pathological processes

Potential Therapeutic Implications:

  • These findings suggest MAP2K3 inhibition may represent a novel therapeutic strategy for enhancing cognition and potentially providing resistance to Alzheimer's disease

  • The natural genetic variations in SuperAgers provide a model for developing interventions that might mimic these protective effects

This research highlights the potential significance of MAP2K3 modulation in cognitive aging and positions it as a promising target for addressing age-related cognitive decline.

How does MAP2K3 expression influence immune cell infiltration in the tumor microenvironment?

MAP2K3 expression significantly impacts immune cell infiltration and the immune microenvironment in tumors, with particular relevance to gliomas:

Immune Cell Infiltration Patterns:

  • Differential MAP2K3 expression levels correlate with variations in immune cell infiltration as revealed by analyses using CIBERSORT and ssGSEA algorithms

  • These algorithms demonstrated clear links between MAP2K3 expression and the infiltration of various immune cell types into the tumor microenvironment

Chemokine Regulation:

  • Groups with higher MAP2K3 expression showed elevated expression of several key chemokines, including CXCL10, CCR5, CCR10, CCL5, CCL7, CCR2, and CCL22

  • These chemokines exert immunosuppressive effects by attracting regulatory T cells (Tregs), macrophages, myeloid-derived suppressor cells (MDSCs), and monocytes

  • This attraction of immunosuppressive cells contributes to immune escape mechanisms in tumors with high MAP2K3 expression

Cytokine Signaling Modulation:

  • Analysis revealed increased expression of interferon receptors, interleukins, interleukin receptors, and other cytokines in the high MAP2K3 expression group

  • These changes create an altered cytokine milieu that can further modulate immune cell function and tumor-immune interactions

Implications for Immunotherapy Response:

  • Tumor Immune Dysfunction and Exclusion (TIDE) scores were calculated to assess sensitivity to immune checkpoint inhibitor therapy

  • Analysis of interferon gamma (IFNG) score, T cell receptor abundance (TCR), TCR Shannon score, microsatellite instability (MSI), and single nucleotide variant (SNV) neoantigens provided insights into how MAP2K3 expression might influence immunotherapy response

Pathway Associations:

  • Higher MAP2K3 expression correlates with elevated activity in several signaling pathways (Hippo, NRF2, PI3K, and TGF) that contribute to tumor immune evasion responses

  • These pathways collectively shape the immune microenvironment and influence anti-tumor immune responses

These findings suggest MAP2K3 expression levels could serve as a biomarker for predicting immunotherapy response and highlight the potential of MAP2K3 inhibition as a strategy to enhance anti-tumor immunity.

What experimental models and assays are most appropriate for studying MAP2K3 inhibition effects?

Research into MAP2K3 inhibition requires a comprehensive experimental approach using complementary models and assays:

Genetic Inhibition Approaches:

  • RNA Interference:

    • siRNA or shRNA targeting MAP2K3 has been successfully employed to deplete MAP2K3 in both wild-type and mutant p53-carrying cells

    • This approach allows for specific targeting of MAP2K3 with minimal off-target effects when properly controlled

  • CRISPR-Cas9 Gene Editing:

    • Provides another genetic method for studying MAP2K3 loss-of-function through precise genome editing

    • Can be used to create stable cell lines or animal models with MAP2K3 knockout or specific mutations

In Vitro Functional Assays:

  • Proliferation and Viability Assessment:

    • Cell counting and MTT/MTS assays to quantify effects on cell proliferation

    • Colony formation assays to evaluate long-term clonogenic ability following MAP2K3 inhibition

    • These assays have demonstrated that MAP2K3 depletion reduces cancer cell proliferation while sparing normal cells

  • Cell Death and Stress Response Analysis:

    • Endoplasmic reticulum stress marker evaluation

    • Autophagy detection methods (LC3 conversion, autophagic flux)

    • PARP cleavage assays to assess apoptosis induction, which increases in response to MAP2K3 inhibition combined with chemotherapy

  • Migration and Invasion Evaluation:

    • Wound healing assays: Cells grown to confluence with a scratch made using a 200 μL pipette tip; migration measured by gap closure over time

    • Transwell migration assays: Cells placed in upper chamber with serum-free medium and chemoattractant in lower chamber; migrated cells stained and counted

    • These assays have revealed MAP2K3's influence on cancer cell motility and invasion potential

In Vivo Models:

  • Tumor xenograft models to assess growth and response to therapeutics following MAP2K3 inhibition

  • Combination studies with established chemotherapeutic agents to evaluate enhanced efficacy

  • Genetic models with conditional MAP2K3 deletion to study tissue-specific effects

Molecular and Pathway Analysis:

  • Signaling Pathway Assessment:

    • Western blotting for phosphorylated downstream targets (e.g., p38 MAPK)

    • Gene Set Variation Analysis (GSVA) to understand global pathway alterations

    • ssGSEA enrichment scoring of oncogenic signaling pathways to identify mechanisms of action

  • Immune Response Evaluation:

    • Flow cytometry to assess immune cell infiltration in tumor models

    • Cytokine/chemokine profiling to understand inflammatory changes

    • Evaluation of immune checkpoint expression in different MAP2K3 expression contexts

These complementary approaches provide a comprehensive understanding of MAP2K3 inhibition effects from molecular mechanisms to functional outcomes in cancer biology and other disease contexts.

How does MAP2K3 interact with the p53 pathway in tumor cells?

MAP2K3 demonstrates a fascinating dual relationship with the p53 pathway that varies depending on p53 mutational status, creating important implications for targeted therapy approaches:

Interaction with Wild-type p53 (wtp53):

  • MAP2K3 depletion induces endoplasmic reticulum stress that contributes to the stabilization of wild-type p53 protein

  • This stabilization enhances p53's tumor suppressor functions and promotes cancer cell death

  • The stress response triggered by MAP2K3 inhibition appears to activate p53-dependent cell death pathways in wtp53-carrying cells

Interaction with Mutant p53 (mutp53):

  • MAP2K3 has been identified as a transcriptional target of mutant p53, creating a feed-forward oncogenic loop

  • Mutant p53 upregulates MAP2K3 to sustain cell proliferation and survival in cancer cells

  • MAP2K3 depletion in mutp53-carrying cells induces autophagy that contributes to the degradation of mutant p53 protein

  • This degradation may partially restore normal cellular functions by reducing the oncogenic effects of mutant p53

Therapeutic Implications:

  • The dual effect of MAP2K3 depletion (stabilizing wtp53 and degrading mutp53) creates a unique therapeutic opportunity

  • MAP2K3 inhibition enhances tumor cell response to chemotherapeutic agents in both wtp53 and mutp53 cancer cells

  • Enhanced therapeutic response is demonstrated by increased poly (ADP-ribose) polymerase (PARP) cleavage and reduced clonogenic ability in vitro

  • In vivo studies confirm that MAP2K3 depletion reduces tumor growth and improves biological response to chemotherapeutics

Molecular Mechanisms:

  • In wtp53 cells, MAP2K3 inhibition likely activates stress-responsive elements that stabilize p53 protein and enhance its activity

  • In mutp53 cells, MAP2K3 inhibition appears to trigger autophagy that selectively targets the mutant p53 protein for degradation

  • These differential effects provide a mechanistic basis for the therapeutic potential of MAP2K3 targeting in diverse cancer contexts

This complex interplay between MAP2K3 and p53 highlights the importance of understanding p53 status when developing MAP2K3-targeted therapeutic strategies, as the mechanisms of action may differ significantly between wild-type and mutant p53-carrying tumors.

What experimental approaches can be used to investigate MAP2K3's role in microglial function?

Given MAP2K3's enriched expression in microglia and its potential role in brain immune system regulation , several sophisticated experimental approaches can elucidate its function in these cells:

Cell Culture Models:

  • Primary Microglia Isolation and Culture:

    • Isolation of primary microglia from rodent or human brain tissue

    • Treatment with MAP2K3-targeting compounds or genetic manipulation (siRNA, CRISPR) to assess functional effects

    • Co-culture systems with neurons to evaluate microglial-neuronal interactions

  • Microglia Cell Lines:

    • Established microglia cell lines (BV-2, HMC3) for high-throughput mechanistic studies

    • Generation of stable MAP2K3 knockdown or overexpression lines

    • CRISPR-based approaches for precise genetic modifications

Functional Assays:

  • Inflammatory Response Assessment:

    • Measurement of pro-inflammatory cytokine production (IL-1β, TNF-α, IL-6) following stimulation with LPS or other PAMPs with and without MAP2K3 inhibition

    • Quantification of anti-inflammatory mediator production

    • RNA-seq analysis to identify global changes in inflammatory gene expression

  • Phagocytosis Assays:

    • Fluorescent bead uptake assays to quantify general phagocytic capacity

    • Amyloid-β peptide clearance assays to assess disease-relevant phagocytosis

    • Live-cell imaging to monitor phagocytosis dynamics following MAP2K3 modulation

  • Migration and Chemotaxis Assays:

    • Transwell migration assays with relevant chemoattractants

    • Scratch assays to assess motility in confluent cultures

    • Live imaging of microglial movement in response to ATP or other damage signals

Signaling Pathway Analysis:

  • p38 MAPK Signaling:

    • Western blotting for phosphorylated p38 MAPK to assess MAP2K3 activity

    • Examination of downstream transcription factor activation (ATF2, MEF2)

    • Pharmacological inhibition studies to dissect pathway components

  • Neuroinflammatory Pathway Mapping:

    • Phospho-proteomics to identify global phosphorylation changes

    • RNA-seq to define transcriptional networks regulated by MAP2K3

    • ChIP-seq to identify transcription factor binding affected by MAP2K3 activity

In Vivo Approaches:

  • Conditional Knockout Models:

    • Generation of microglia-specific MAP2K3 knockout mice using Cx3cr1-Cre or similar systems

    • Assessment of microglial morphology, density, and function in these models

    • Behavioral testing to evaluate cognitive outcomes

  • Neuroinflammation Models:

    • LPS-induced neuroinflammation with assessment of microglial activation

    • MPTP model of Parkinson's disease to study microglial responses

    • Amyloid models of Alzheimer's disease with MAP2K3 modulation

  • In Vivo Imaging:

    • Two-photon microscopy to visualize microglial dynamics in living brain tissue

    • PET imaging with microglial tracers to assess activation states

    • Correlative studies linking imaging findings with molecular analyses

These comprehensive approaches would provide detailed insights into MAP2K3's specific roles in microglial function and its potential as a therapeutic target for neuroinflammatory and neurodegenerative conditions.

How do MAP2K3 expression levels correlate with patient survival in glioma?

Research has revealed significant correlations between MAP2K3 expression and patient outcomes in glioma, providing important prognostic insights:

Survival Analysis Findings:

  • Kaplan-Meier curve analysis demonstrated a strong association between MAP2K3 expression levels and patient survival

  • Higher MAP2K3 expression levels consistently associated with poorer prognosis in glioma patients across multiple datasets

  • This correlation remained significant after controlling for various clinical factors, suggesting MAP2K3 as an independent prognostic indicator

Grade-Specific Expression Patterns:

  • MAP2K3 expression varies significantly across gliomas of different WHO classifications

  • Analysis of expression patterns in low-grade glioma (LGG), glioblastoma multiforme (GBM), and normal tissues revealed grade-dependent increases

  • The Kruskal-Wallis test confirmed statistically significant differences in MAP2K3 expression across different WHO grades using TCGA data

Molecular Mechanisms Underlying Prognostic Impact:

  • Poor prognosis associated with high MAP2K3 expression likely stems from multiple biological effects:

    • Enhanced cell migration and invasion capabilities demonstrated in functional assays

    • Increased activity in oncogenic signaling pathways (Hippo, NRF2, PI3K, and TGF)

    • Development of an immunosuppressive microenvironment that facilitates tumor immune evasion

    • Upregulation of chemokines that recruit immunosuppressive cell populations

Potential as a Biomarker:

  • The consistent association with survival outcomes positions MAP2K3 as a valuable prognostic biomarker in glioma

  • Expression analysis could potentially inform treatment decisions, particularly regarding the potential benefit of immunotherapeutic approaches

  • Combined with other molecular markers, MAP2K3 expression could contribute to more precise patient stratification systems

These findings collectively establish MAP2K3 as an important prognostic indicator in glioma and highlight its potential utility in clinical decision-making and therapeutic targeting.

What techniques can be used to analyze MAP2K3-related signaling pathways in human tissue samples?

Several sophisticated techniques can be employed to comprehensively analyze MAP2K3-related signaling pathways in human tissue samples:

Computational and Bioinformatic Approaches:

  • Gene Set Variation Analysis (GSVA):

    • Assesses the distribution of genes in predefined sets to determine their role in phenotype definition

    • Identifies functional and pathway significance differences between high and low MAP2K3 expression groups

    • Utilizes reference gene sets like "h.all.v7.2.symbols" and "c2.cp.kegg.v7.2.symbols" from the MsigDB database

  • Differential Pathway Analysis:

    • The "Limma" program analyzes differences in GSVA pathways between patient groups

    • Heat maps display Hallmarks and KEGG differential pathways associated with MAP2K3 expression

    • Adjusted criteria (p < 0.05 and abs(log2FC) > 0.3) can identify statistically significant pathway alterations

  • Single Sample Gene Set Enrichment Analysis (ssGSEA):

    • Enables enrichment scoring of classical oncogenic signaling pathways (Wnt, TP53, TGF, RAS, PI3K, NRF2, NOTCH, MYC, cell cycle, Hippo)

    • Has identified pathways with elevated activity in high MAP2K3 expression contexts

Tissue-Based Molecular Methods:

  • Immunohistochemistry (IHC) and Multiplex Immunofluorescence:

    • Evaluation of MAP2K3 protein expression and phosphorylation state in tissue sections

    • Use of specific antibodies (e.g., anti-MAP2K3 primary antibody HPA043783)

    • Multiplexed staining to simultaneously assess multiple pathway components and immune cell markers

  • Phospho-Protein Analysis:

    • Phospho-specific IHC or Western blotting for activation states of pathway components

    • Reverse Phase Protein Array (RPPA) for high-throughput assessment of multiple phosphorylated proteins

    • Proximity ligation assays to detect protein-protein interactions in tissue sections

Genetic and Transcriptomic Methods:

  • RNA Sequencing with Pathway Analysis:

    • Comprehensive transcriptome profiling from tissue samples

    • KEGG and Gene Ontology enrichment analysis of differentially expressed genes

    • Construction of gene signatures related to MAP2K3 pathway activation

  • Mutation and Copy Number Analysis:

    • Analysis of somatic mutations and copy number alterations (CNAs) in MAP2K3 pathway components

    • VarScan2 software for analyzing whole-genome sequence data of somatic mutations

    • The CoMEt algorithm to identify co-occurring and mutually exclusive mutations in pathway genes

  • Spatial Transcriptomics:

    • Emerging techniques to map gene expression patterns with spatial resolution in tissue sections

    • Allows for analysis of regional heterogeneity in MAP2K3 pathway activation

These complementary approaches provide researchers with a comprehensive toolkit to dissect MAP2K3-related signaling pathway activities in human tissue samples, enabling deeper understanding of MAP2K3's role in disease processes and identification of potential therapeutic vulnerabilities.

How does modulating MAP2K3 affect cell migration and invasion in experimental cancer models?

MAP2K3 modulation significantly impacts cancer cell migration and invasion capabilities, with important implications for understanding metastatic potential and developing targeted therapies:

Methodological Approaches to Study Migration and Invasion:

  • Wound Healing Assay (Scratch Assay):

    • Cells grown to confluence with a scratch made using a sterile 200 μL pipette tip

    • Debris washed away with phosphate-buffered saline and cells incubated in serum-free medium

    • Images captured at baseline (0h) and later timepoints (e.g., 24h) using an inverted microscope

    • Migration rate quantified by measuring gap closure with ImageJ or similar software

  • Transwell Migration Assay:

    • Cells suspended in serum-free medium (1 × 10^5 cells/mL) and placed in the upper chamber of a Transwell insert

    • Lower chamber containing medium with 10% fetal bovine serum as a chemoattractant

    • Following 24-hour incubation, cells on upper membrane removed while those on lower surface fixed, stained, and counted

    • Provides quantitative measure of directed cell migration

Effects of MAP2K3 Inhibition:

  • Reduced Migration Rate: Cells with MAP2K3 inhibition show significantly decreased gap closure in wound healing assays

  • Decreased Invasive Potential: Fewer cells traverse the membrane in Transwell migration assays when MAP2K3 is inhibited

  • Morphological Changes: Alterations in cell morphology and cytoskeletal arrangement often accompany these functional changes

Molecular Mechanisms Underlying Migration Effects:

  • Pathway Modulation: MAP2K3 inhibition affects signaling pathways that regulate cell migration, including PI3K and TGF pathways

  • Chemokine Alterations: Changes in expression of motility-related chemokines, including CXCL10, CCR5, CCR10, CCL5, CCL7, CCR2, and CCL22

  • Cytoskeletal Regulation: Likely effects on actin cytoskeleton organization and focal adhesion dynamics through downstream signaling

Therapeutic Implications:

  • The significant effects on migration and invasion suggest MAP2K3 inhibitors could potentially reduce metastatic potential in cancer

  • Combined with effects on cell proliferation, survival, and immune modulation, this creates a comprehensive anti-cancer profile

  • The relatively selective effect on cancer cells versus normal cells suggests a potential therapeutic window

These findings highlight MAP2K3's critical role in promoting cancer cell motility and invasion, supporting its potential as a therapeutic target to prevent or reduce metastatic spread.

What are the implications of MAP2K3 activity in Alzheimer's disease pathogenesis?

Research has revealed several important implications of MAP2K3 in Alzheimer's disease (AD) pathogenesis, suggesting potential therapeutic opportunities:

MAP2K3 Expression Alterations in AD:

  • The MAP2K3 gene is significantly upregulated in the middle temporal gyrus of AD patients compared to healthy controls

  • This upregulation appears to be a response to AD-related pathological processes

  • Expression changes may contribute to disease progression through multiple mechanisms

Relationship to Beta-Amyloid Pathology:

  • MAP2K3 participates in signaling cascades associated with beta-amyloid mediated apoptosis, a key pathological process in AD

  • This connection suggests MAP2K3 activation may contribute to neuronal loss downstream of amyloid accumulation

  • The pathway linking MAP2K3 to amyloid toxicity could represent a therapeutic intervention point

Microglial Function in AD Context:

  • MAP2K3 shows enriched expression in microglia, the brain's resident immune cells

  • Microglia play critical roles in AD progression through inflammatory responses and amyloid clearance

  • MAP2K3's role in microglia suggests it may influence neuroinflammatory processes that contribute to AD pathology

  • Research in other contexts has linked MAP2K3 to inflammatory signaling cascades that could exacerbate AD progression

Genetic Insights from SuperAgers:

  • Studies in SuperAgers (elderly individuals with superior memory) identified specific MAP2K3 genetic variants associated with preserved cognitive function

  • These findings suggest that reduced MAP2K3 activity from birth may provide neuroprotection against age-related cognitive decline

  • The protective effect may stem from lowered p38-MAPK activity in neuronal cells and reduced microglial-mediated inflammation

Therapeutic Development Implications:

  • Research postulates that MAP2K3 inhibitors may represent a novel therapeutic strategy for enhanced cognition and resistance to AD

  • Such inhibitors would likely need to partially rather than completely block MAP2K3 activity, as the naturally protective variants do not fully impair function

  • The dual effects on neuronal survival and microglial function suggest MAP2K3 inhibition could address multiple aspects of AD pathophysiology

These findings position MAP2K3 as a promising therapeutic target for Alzheimer's disease, with potential benefits for both preventing neurodegeneration and enhancing cognitive function.

Product Science Overview

Structure and Function

MAP2K3 is a dual-specificity threonine/tyrosine protein kinase. It is activated by mitogenic and environmental stress and participates in the MAP kinase-mediated signaling cascade. Upon activation, MAP2K3 phosphorylates and activates MAPK14/p38-MAPK . This activation is crucial for the regulation of several biological processes, including inflammation and cellular stress responses .

Activation and Regulation

MAP2K3 can be activated by various stimuli, including insulin and environmental stressors . It plays a significant role in the expression of glucose transporters, which are essential for cellular glucose uptake . Additionally, MAP2K3 is involved in the regulation of the stress-activated protein kinase (SAPK) and extracellular signal-regulated protein kinase (ERK) pathways . These pathways are vital for cellular responses to stress and inflammatory stimuli.

Biological Significance

The activation of MAP2K3 leads to the phosphorylation of downstream targets, which in turn regulate various cellular processes. For instance, the activation of MAPK14/p38-MAPK by MAP2K3 is essential for the cellular response to stress and inflammation . This pathway also plays a role in the regulation of apoptosis, cell cycle progression, and differentiation.

Clinical Relevance

Mutations or dysregulation of MAP2K3 have been associated with various diseases, including cancer and inflammatory disorders . Understanding the role of MAP2K3 in these diseases can provide insights into potential therapeutic targets. For example, targeting MAP2K3 with specific inhibitors could be a strategy for treating diseases characterized by excessive inflammation or uncontrolled cell proliferation.

Recombinant MAP2K3

Recombinant MAP2K3 is a form of the protein that is produced through recombinant DNA technology. This technology allows for the production of large quantities of the protein, which can be used for research and therapeutic purposes. Recombinant MAP2K3 is used in various studies to understand its function and regulation, as well as to develop potential therapeutic interventions.

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.