PLA2G2E is encoded by the PLA2G2E gene located on human chromosome 1 . Key features include:
PLA2G2E requires Ca²⁺ for activity and operates via a single-water or assisted-water mechanism involving His-48 and Asp-99 residues . Unlike some phospholipases, it lacks activity against typical substrates like phosphatidylglycerol .
PLA2G2E hydrolyzes phospholipids in lipoproteins to generate:
Lyso-PC: A pro-inflammatory mediator implicated in atherosclerosis .
Free Fatty Acids: Including arachidonic acid, a precursor for eicosanoids (e.g., prostaglandins, leukotrienes) .
In mice, PLA2G2E regulates HDL particle size and cholesterol levels during inflammatory responses . Its deficiency alters lipid mediator profiles in lungs and T cells, affecting immune function .
Recent studies highlight PLA2G2E’s critical role in influenza virus defense :
Observation | Wild-Type Mice | Pla2g2e⁻/⁻ Mice | hPLA2G2E Transgenic Mice |
---|---|---|---|
Survival Rate Post-Infection | 60% | 20% | 80% |
Viral Load in Lungs (Day 7 p.i.) | Moderate | 3x Higher | 50% Lower |
Influenza-Specific CD8⁺ T Cells | Robust | Reduced Cytotoxicity | Enhanced Polyfunctionality |
Granzyme B Production | High | Low | Elevated |
PLA2G2E deficiency impairs T cell-mediated cytotoxicity and delays T cell recruitment, while overexpression enhances antiviral immunity . Mechanistically, PLA2G2E modulates lipid mediators like 13-HODE and dhk-PGE₂, which are critical for immune cell function .
PLA2G2E’s dual role in lipid signaling and immune regulation positions it as a potential target for:
Inflammatory Diseases: Atherosclerosis, asthma, and autoimmune disorders .
Antiviral Therapies: Enhancing T cell responses against influenza and other viruses .
Notably, recombinant human PLA2G2E (ENZ-327) is available for research, enabling further mechanistic studies .
Functional Redundancy: Overlap with other sPLA2 isoforms (e.g., PLA2G2D) complicates disease-specific targeting .
Lipid Mediator Networks: The enzyme’s impact on eicosanoid balance requires deeper profiling .
Clinical Translation: Human trials are needed to validate preclinical findings in murine models .
MRGSHHHHHH GMASHMNLVQ FGVMIEKMTG KSALQYNDYG CYCGIGGSHW PVDQTDWCCH AHDCCYGRLE KLGCEPKLEK YLFSVSERGI FCAGRTTCQR LTCECDKRAA LCFRRNLGTY NRKYAHYPNK LCTGPTPPC
PLA2G2E (phospholipase A2 group IIE) is a member of the secreted phospholipase A2 (sPLA2) family that catalyzes the hydrolysis of glycerophospholipids, releasing fatty acids and lysophospholipids. This enzyme plays a significant role in lipid metabolism and immune regulation, particularly in the context of respiratory viral infections .
The human PLA2G2E gene is located on Chromosome 1, specifically on the reference sequence NC_000001.11 in the GRCh38.p14 Primary Assembly . The gene is part of the group II subfamily within the broader sPLA2 family, characterized by specific structural motifs involved in calcium binding and catalytic activity .
Under normal physiological conditions, PLA2G2E exhibits relatively low expression levels in most tissues. It is primarily localized to glutamate-rich tissues and maintains minimal expression under homeostatic conditions .
Viral infection: H1N1 influenza virus infection induces marked upregulation of PLA2G2E expression in human lung cancer cell lines (A549 and NCI-H1299)
Cell-specific expression: During influenza infection, PLA2G2E expression increases predominantly in lung epithelial cells rather than alveolar macrophages or endothelial cells
Inflammatory stimuli: Alveolar macrophages show upregulated PLA2G2E expression in response to lipopolysaccharide (LPS) stimulation, highlighting its role in immune activation
This differential expression pattern suggests PLA2G2E plays a context-dependent role in inflammatory and immune responses, with specific regulation depending on cellular environment and stimulation conditions.
While the search results don't provide comprehensive comparative data on all PLA2 family members, we can extract some distinguishing characteristics of PLA2G2E:
Characteristic | PLA2G2E | Other sPLA2 Family Members |
---|---|---|
Expression Profile | Low basal expression with significant upregulation during viral infection | Variable depending on family member |
Primary Function in Immunity | Regulation of T cell responses during viral infection | Diverse roles across family members |
Tissue Specificity | Primarily upregulated in lung epithelial cells during infection | Different tissue tropism across family members |
Impact on Viral Defense | Critical for T cell-mediated antiviral immunity | Variable across family members |
PLA2G2E appears to have a specialized role in modulating T cell responses during viral infections, particularly influenza, distinguishing it from other phospholipases that may have broader or different immune regulatory functions .
Based on methodologies described in the research literature, the following approaches are recommended for measuring PLA2G2E expression:
Transcriptomic Analysis:
Cell-Specific Expression Analysis:
Protein Detection:
Western blotting using specific antibodies against PLA2G2E
Immunohistochemistry for tissue localization studies
When designing experiments to measure PLA2G2E expression, researchers should consider:
Including appropriate positive controls (e.g., influenza-infected lung epithelial cells)
Examining multiple timepoints post-stimulation to capture expression dynamics
Analyzing both mRNA and protein levels to account for post-transcriptional regulation
Research using PLA2G2E-deficient mouse models (Pla2g2e-/-) demonstrates significant impacts on viral susceptibility and immune responses :
Parameter | Wild-Type Mice | Pla2g2e-/- Mice | Significance |
---|---|---|---|
Survival Rate | Higher | Significantly lower | p < 0.05 |
Viral Load in Lungs | Lower | Higher | p < 0.05 |
Weight Loss During Infection | Less severe | More severe | p < 0.05 |
Viral Clearance | Efficient | Less efficient | p < 0.05 |
Interestingly, Pla2g2e-/- mice displayed comparable pulmonary pathological changes to wild-type mice, with similar wall thickening and inflammatory cell infiltration on days 7 and 14 post-infection. Both groups showed occasional hemorrhage and pulmonary edema .
The expression levels of chemokines (CCL2, CCL3, CCL5, CXCL2, CXCL5, and CXCL10) and proinflammatory cytokines (IL-1β, IL-6, and IL-8) were upregulated in lung tissues of both wild-type and Pla2g2e-/- mice at comparable levels, suggesting that PLA2G2E's protective effect against viral infection operates through mechanisms distinct from general inflammatory pathways .
PLA2G2E plays a critical role in regulating T cell-mediated immunity during influenza virus infection through several mechanisms :
T Cell Activation and Function:
Pla2g2e-/- mice show impaired influenza-specific cellular immunity
T cell-mediated cytotoxicity is reduced in PLA2G2E-deficient animals
The frequency of virus-specific CD8+ T cells (D^bNP 366-374+CD8+) in broncho-alveolar lavage fluid (BALF) is decreased in Pla2g2e-/- mice
Interestingly, I-A^bNP 311-325+CD4+ T cells are increased in Pla2g2e-/- mice, suggesting differential regulation of CD4+ and CD8+ T cell responses
Cytokine Production:
Humoral vs. Cellular Immunity:
These findings demonstrate that PLA2G2E contributes specifically to the regulation of T cell responses during influenza virus infection, with minimal impact on other aspects of antiviral immunity.
Transgenic mice expressing the human PLA2G2E gene (hPLA2G2E mice) at the Rosa26 genomic safe harbor site demonstrate enhanced antiviral responses compared to wild-type controls :
Parameter | Wild-Type Mice | hPLA2G2E Transgenic Mice | Significance |
---|---|---|---|
Weight Loss Rate | Rapid weight loss exceeding 30% by day 10 | Significantly slower weight loss with recovery starting at day 10 | p < 0.05 |
Survival Rate | 0% (all mice succumbed to infection) | >60% survival | p < 0.05 |
D^bNP366-374+CD8+ T Cells | Lower frequency | Significantly increased frequency | p < 0.05 |
The significant improvement in survival and reduced weight loss in hPLA2G2E transgenic mice strongly suggests that human PLA2G2E provides protective effects during influenza infection . This enhanced protection correlates with increased virus-specific CD8+ T cells, further supporting PLA2G2E's role in modulating T cell-mediated immunity.
These findings have important implications for developing potential therapeutic strategies targeting PLA2G2E enhancement to combat influenza and potentially other viral infections.
Pla2g2e deficiency results in perturbation of lipid mediators in both lung tissue and T cells, which likely contributes to the observed impacts on immune responses . As a phospholipase, PLA2G2E catalyzes the release of fatty acids from membrane phospholipids, potentially affecting the production of various bioactive lipid mediators.
While the search results don't provide detailed lipidomic analysis data, we can infer that PLA2G2E deficiency likely affects:
Eicosanoid Production: Altered release of arachidonic acid would impact the synthesis of prostaglandins, leukotrienes, and other eicosanoids involved in inflammation and immune regulation
Specialized Pro-resolving Mediators (SPMs): Changes in omega-3 fatty acid release could affect the production of resolvins, protectins, and maresins that regulate inflammation resolution
Lysophospholipids: As products of phospholipase A2 activity, altered levels of lysophospholipids may affect membrane properties and signaling pathways in immune cells
The perturbation of these lipid mediators in T cells specifically suggests a mechanism by which PLA2G2E deficiency leads to impaired T cell activation, proliferation, and effector functions during viral infection .
Based on current research findings, several methodological approaches would be valuable for investigating PLA2G2E as a therapeutic target :
Animal Models:
Utilize both knockout (Pla2g2e-/-) and transgenic (hPLA2G2E) mouse models to assess loss- and gain-of-function effects
Consider humanized mouse models expressing human immune system components to better predict translational outcomes
Employ various viral challenge models beyond influenza to determine breadth of protective effects
Cell-Based Assays:
Primary T cell isolation and functional assays (proliferation, cytotoxicity, cytokine production)
Ex vivo stimulation of patient-derived T cells with recombinant PLA2G2E
Co-culture systems to examine interactions between PLA2G2E-producing epithelial cells and T cells
Therapeutic Delivery Strategies:
Gene therapy approaches targeting PLA2G2E expression
Recombinant PLA2G2E protein administration via appropriate delivery systems
Small molecule modulators of PLA2G2E expression or activity
Biomarker Development:
Lipidomic profiling to identify specific lipid mediator signatures associated with PLA2G2E activity
T cell functional assays as pharmacodynamic markers of PLA2G2E modulation
Viral load quantification as an efficacy endpoint
Safety Assessment:
Comprehensive evaluation of potential off-target effects, particularly on inflammatory pathways
Dose-ranging studies to determine therapeutic window
Long-term consequences of PLA2G2E modulation on immune homeostasis
This multi-faceted approach would provide valuable insights into PLA2G2E's potential as a therapeutic target for viral infections and possibly other conditions involving T cell dysfunction.
Several significant challenges must be addressed when translating PLA2G2E research findings from animal models to human applications:
Species-Specific Differences:
Target Cell Identification and Accessibility:
Determining which human cell populations should be targeted to enhance PLA2G2E expression
Developing delivery methods that can effectively reach these target cells
Understanding tissue-specific regulation of PLA2G2E in humans
Therapeutic Timing:
Identifying the optimal intervention window during infection or disease progression
Determining duration of treatment needed for efficacy
Assessing potential for prophylactic versus therapeutic applications
Individual Variability:
Genetic polymorphisms in human PLA2G2E may affect function or therapeutic response
Existing immune status and comorbidities could influence treatment outcomes
Potential interactions with concurrent medications or treatments
To accurately assess PLA2G2E's influence on T cell responses, researchers should consider these methodological approaches :
Antigen-Specific T Cell Quantification:
Functional T Cell Assays:
T Cell Activation and Differentiation:
Analysis of activation markers (CD25, CD69, etc.)
Assessment of memory T cell development (central memory vs. effector memory)
Evaluation of exhaustion markers in chronic stimulation settings
In vivo Approaches:
Adoptive transfer of labeled, antigen-specific T cells to track proliferation and migration
In vivo killing assays to assess cytotoxic function
Two-photon microscopy to visualize T cell interactions in tissues
When designing experiments, researchers should consider using multiple complementary assays and incorporate appropriate controls, including both positive controls (wild-type mice) and negative controls (Pla2g2e-/- mice) .
To effectively study PLA2G2E's influence on lipid mediator profiles, researchers should consider these methodological approaches:
Comprehensive Lipidomic Analysis:
Liquid chromatography-mass spectrometry (LC-MS) for detailed lipid profiling
Targeted analysis of eicosanoids, specialized pro-resolving mediators, and lysophospholipids
Spatial lipidomics to determine tissue-specific lipid alterations
Enzymatic Activity Assays:
Fluorogenic substrate assays to measure PLA2G2E enzymatic activity
Competition assays with other phospholipases to determine substrate specificity
In vitro reconstitution with defined phospholipid substrates
Cellular Models:
Comparison of lipid profiles between wild-type, Pla2g2e-/-, and hPLA2G2E transgenic models
Cell-specific analysis (epithelial cells, T cells, macrophages) to determine cell-type-dependent effects
Stimulation conditions that mimic viral infection or inflammatory environments
Time-Course Analysis:
Kinetic studies to track changes in lipid mediators over time following stimulation
Correlation with T cell activation states and functional outcomes
Assessment of lipid mediator persistence and metabolism
Research indicates that PLA2G2E deficiency results in perturbation of lipid mediators in both lung tissue and T cells, potentially contributing to impaired T cell responses during viral infection . Detailed characterization of these lipid alterations would provide valuable insights into the mechanisms by which PLA2G2E influences immune function.
While current research has focused on PLA2G2E's role in influenza virus infection, several experimental models could expand our understanding of its broader biological functions:
Additional Viral Infection Models:
Respiratory viruses beyond influenza (RSV, SARS-CoV-2, parainfluenza viruses)
Systemic viral infections to assess non-respiratory roles
Persistent viral infections to examine chronic immune regulation
Autoimmune Disease Models:
Experimental autoimmune encephalomyelitis (EAE) for multiple sclerosis
Collagen-induced arthritis
Type 1 diabetes models
These models would help determine whether PLA2G2E plays a protective or pathogenic role in aberrant T cell responses
Cancer Immunotherapy Models:
Tumor challenge models to assess impact on anti-tumor immunity
Checkpoint inhibitor combination studies
Adoptive T cell therapy models with PLA2G2E-modified T cells
Tissue-Specific Conditional Models:
Cell-specific PLA2G2E deletion or overexpression using Cre-lox systems
Inducible expression systems to control timing of PLA2G2E modulation
Tissue-specific expression to determine local versus systemic effects
These diverse experimental approaches would provide a more comprehensive understanding of PLA2G2E's role in immune regulation beyond respiratory viral infections.
Based on its role in T cell regulation, PLA2G2E could be integrated into therapeutic strategies in several innovative ways:
Combination with Existing Antivirals:
PLA2G2E enhancement could complement direct-acting antivirals by strengthening T cell-mediated viral clearance
Potential for reduced viral resistance through multi-mechanism approach
Enhanced resolution of infection through improved T cell function
Vaccine Adjuvant Development:
PLA2G2E modulators could potentially enhance T cell responses to vaccination
Particularly valuable for vaccines requiring strong cellular immunity
Could improve protection in populations with suboptimal vaccine responses
Cell-Based Immunotherapies:
Engineering T cells to express or respond to PLA2G2E for adoptive cell therapies
Modulation of lipid mediator environment to enhance CAR-T cell function
Ex vivo expansion protocols incorporating PLA2G2E stimulation
Precision Medicine Applications:
Genetic screening for PLA2G2E variants that impact susceptibility to viral infections
Tailored therapeutic approaches based on individual PLA2G2E expression or activity
Biomarker development to identify patients most likely to benefit from PLA2G2E-targeted therapies
Research with transgenic mice expressing human PLA2G2E demonstrates significant protection against influenza virus infection, suggesting that enhancing PLA2G2E expression or activity could be a viable therapeutic strategy for managing viral infections .
Current research provides compelling evidence for PLA2G2E's significant role in T cell-mediated immunity, particularly during viral infections. The findings from both knockout and transgenic models consistently demonstrate that PLA2G2E:
Contributes to protection against influenza virus infection through regulation of T cell responses
Specifically modulates T cell-mediated immunity without significantly affecting innate or humoral immune responses
Influences lipid mediator profiles in lung tissue and T cells, likely affecting immune cell function
When overexpressed (human PLA2G2E in transgenic mice), provides enhanced protection against viral challenge
These consistent findings across multiple experimental approaches establish PLA2G2E as an important regulator of T cell-mediated immunity with potential therapeutic implications. The selective effect on T cell responses, rather than broad immunomodulation, suggests PLA2G2E could be targeted with potentially fewer off-target effects than general immunomodulatory approaches.
The research also highlights the interconnection between lipid metabolism and immune function, with PLA2G2E serving as a critical link between these physiological systems. This emerging understanding opens new avenues for immunomodulatory strategies that target specific aspects of lipid metabolism to enhance desired immune responses.
Secreted Phospholipase A2-IIE (sPLA2-IIE) is a member of the secreted phospholipase A2 (sPLA2) family, which are enzymes involved in the hydrolysis of phospholipids at the sn-2 position, releasing free fatty acids and lysophospholipids. These enzymes play crucial roles in various physiological processes, including inflammation, host defense, and lipid metabolism .
sPLA2-IIE is characterized by its high content of disulfide bonds, typically 6-8, which contribute to its stability and activity. The enzyme specifically targets the sn-2 acyl bond of phospholipids, leading to the production of arachidonic acid, a precursor for pro-inflammatory eicosanoids . The human recombinant form of sPLA2-IIE is often produced with an N-terminal His-Tag to facilitate purification and study .
The recombinant sPLA2-IIE is commonly expressed in the yeast Pichia pastoris, which allows for high-yield production. The protein is purified using a combination of cation exchange and size-exclusion chromatography. This method ensures the production of high-purity sPLA2-IIE, which is essential for functional and structural studies .
sPLA2-IIE is involved in various biological processes:
The study of recombinant sPLA2-IIE has provided insights into its role in disease mechanisms, particularly in inflammatory and cardiovascular diseases. The high-purity recombinant protein is used in various assays to understand its enzymatic activity, substrate specificity, and potential as a therapeutic target .