PLCXD3 Human

Phosphatidylinositol Specific Phospholipase C X Domain Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to PLCXD3 Human

Phosphatidylinositol-specific phospholipase C X domain-containing protein 3 (PLCXD3) is a member of the phosphoinositide-specific phospholipase (PI-PLC) family, which hydrolyzes phosphatidylinositol-4,5-bisphosphate (PIP₂) to generate secondary messengers involved in cellular signaling . The recombinant human PLCXD3 protein (PRO-2651) is a 38.7 kDa polypeptide chain containing 344 amino acids, produced in Escherichia coli with a His-tag for purification . PLCXD3 has emerged as a critical regulator in metabolic and endocrine functions, particularly in pancreatic β-cells and insulin secretion .

Tissue Expression and Cellular Localization

PLCXD3 exhibits distinct tissue distribution:

  • High Expression: Pancreatic islets (top 94th percentile compared to other PI-PLCs) , brain , and testicular germ cells .

  • Low Expression: Diabetic islets show reduced PLCXD3 levels, correlating with impaired insulin secretion .

Single-Cell RNA Sequencing Data (Human Protein Atlas) :

Tissue/Cell TypeExpression Level
Pancreatic β-cellsHigh
NeuronsModerate
Immune CellsLow

4.1. Insulin Secretion and β-Cell Function

  • Silencing PLCXD3 in INS-1 (832/13) cells reduces glucose-stimulated insulin secretion (GSIS) by 40% and insulin content by 30% .

  • PLCXD3 interacts with insulin receptor signaling pathways and enhances glucose sensing .

  • Positive correlation with GLP1R expression suggests involvement in incretin-mediated insulin release .

4.2. Lipid Metabolism

  • PLCXD3 hydrolyzes PIP₂ to produce inositol trisphosphate (IP₃) and diacylglycerol (DAG), modulating calcium signaling and PKC activation .

5.1. Metabolic Disorders

  • Type 2 Diabetes (T2D): Reduced PLCXD3 expression in diabetic islets correlates with higher HbA1c and BMI .

  • Metabolic Syndrome (MetS): Genetic variants (rs319013-GG and rs9292806-CC) increase MetS risk (OR: 2.62–2.92) in the Emirati population .

5.2. Other Pathologies

  • ROHHAD Syndrome: Mutations linked to hypothalamic dysfunction and obesity .

  • Spermatogenic Failure: Absence of PLCXD3 in testicular germ cells is associated with infertility .

Genetic Variants and Clinical Implications

Key Variants and Phenotypic Effects :

VariantGenotypeAssociationClinical Impact
rs319013GG↑ Fasting glucose, ↓ HDL-CMetS risk (OR: 2.62)
rs9292806CC↑ HbA1c, ↑ BMIMetS risk (OR: 2.92)

These variants alter PLCXD3’s regulatory regions, potentially disrupting lipid signaling .

Research Applications

  • Drug Discovery: PLCXD3 is a target for therapies aiming to enhance insulin secretion in T2D .

  • Biomarker Development: Genetic screening for rs319013 and rs9292806 may predict MetS susceptibility .

Product Specs

Introduction
PLCXD3, also known as phosphatidylinositol Specific Phospholipase C X Domain Containing 3, possesses a PI-PLC X-box domain. This protein is implicated in Creutzfeldt-Jakob Disease and is associated with ectoderm differentiation, obsolete signal transducer activity, and phosphoric diester hydrolase activity.
Description
Recombinant human PLCXD3, expressed in E. coli, is a non-glycosylated polypeptide chain comprising 344 amino acids (residues 1-321) with a molecular weight of 38.7 kDa. The protein contains a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The PLCXD3 solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 20% glycerol, 0.15 M NaCl, and 1 mM DTT.
Stability
For short-term storage (up to 4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freezing and thawing cycles should be avoided.
Purity
The purity of PLCXD3 is greater than 85.0% as determined by SDS-PAGE analysis.
Synonyms

PI-PLC X domain-containing protein 3, phosphatidylinositol-specific phospholipase C X domain containing 3, phosphatidylinositol-specific phospholipase C, X domain containing 3

Source

Escherichia Coli.

Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMASSQGK NELKLADWMA TLPESMHSIP LTNLAIPGSH DSFSFYIDEA SPVGPEQPET VQNFVSVFGT VAKKLMRKWL ATQTMNFTGQ LGAGIRYFDL RISTKPRDPD NELYFAHGLF SAKVNEGLEE INAFLTDHHK EVVFLDFNHF YGMQKYHHEK LVQMLKDIYG NKMCPAIFAQ EVSLKYLWEK DYQVLVFYHS PVALEVPFLW PGQMMPAPWA NTTDPEKLIQ FLQASITERR KKGSFFISQV VLTPKASTVV KGVASGLRET ITERALPAMM QWVRTQKPGE SGINIVTADF VELGDFISTV IKLNYVFDEG EANT

Q&A

What is PLCXD3 and what structural domains characterize it?

PLCXD3 (phosphatidylinositol-specific phospholipase C X domain containing 3) is a member of the phosphoinositide-specific phospholipases (PI-PLC) family. It is a 321-amino acid protein containing a PI-PLC X-box domain, which is evolutionarily conserved from prokaryotes to mammals . This domain is essential for the catalytic activity of PLC proteins . When produced as a recombinant protein, PLCXD3 has a molecular mass of approximately 38.7 kDa . The X-box domain is critical for phosphoric diester hydrolase activity, suggesting its importance in signal transduction pathways .

In which tissues is PLCXD3 predominantly expressed?

PLCXD3 is among the highly expressed PI-PLCs in human pancreatic islets and rat INS-1 (832/13) cells, as demonstrated by microarray and RNA sequencing data . While the search results don't provide a comprehensive tissue expression profile, the data suggests significant expression in pancreatic β-cells where it plays important roles in insulin signaling and production . Expression analysis methods such as RNA-seq and microarray technologies have been instrumental in identifying the relative abundance of PLCXD3 across different tissue types.

What are the known physiological functions of PLCXD3?

PLCXD3 appears to play a critical role in pancreatic β-cell function, particularly in insulin signaling and biosynthesis . Research shows that PLCXD3 expression positively correlates with insulin and GLP1R expression in human islets . Its functional importance is demonstrated by the fact that expression silencing of PLCXD3 in INS-1 (832/13) cells reduces glucose-stimulated insulin secretion (GSIS) and insulin content . Additionally, PLCXD3 may be involved in signal transduction pathways through its phospholipase activity, potentially affecting cell growth, differentiation, and other cellular processes .

How does PLCXD3 expression correlate with metabolic parameters in human subjects?

Research on human pancreatic islets has revealed significant correlations between PLCXD3 expression and various metabolic parameters. PLCXD3 expression correlates inversely with donors' body mass index (BMI) and glycated hemoglobin (HbA1c) . This suggests that decreased PLCXD3 expression is associated with obesity and poorer glycemic control. Furthermore, PLCXD3 expression is reduced in islets from diabetic donors compared to non-diabetic controls . These clinical correlations highlight the potential significance of PLCXD3 in metabolic health and diabetes pathophysiology, suggesting that it may serve as a biomarker for β-cell function or as a therapeutic target.

What molecular mechanisms explain PLCXD3's role in insulin secretion and biosynthesis?

When PLCXD3 expression is silenced in β-cells, several key genes involved in insulin signaling and biosynthesis are downregulated, including Insulin, NEUROD1, GLUT2, GCK, INSR, IRS2, and AKT . This suggests that PLCXD3 may function as a regulator of these important pathways. The mechanism appears to involve both glucose sensing and insulin biosynthesis processes. Specifically, the downregulation of glucose transporters (GLUT2) and glucokinase (GCK) would impair glucose sensing, while reduced expression of insulin receptor signaling components (INSR, IRS2, AKT) would affect insulin signaling pathways . NEUROD1, a transcription factor crucial for insulin gene expression, is also affected, suggesting PLCXD3 may indirectly regulate insulin transcription .

What is the significance of the PLCXD3-ALK fusion in cancer research?

A novel fusion gene, PLCXD3-ALK (P1, A19), has been identified in a patient with advanced lung squamous cell carcinoma (LUSC) . This fusion represents a new type of ALK rearrangement in non-small cell lung cancer (NSCLC). Functional studies have demonstrated that PLCXD3-ALK, similar to other ALK fusions like EML4-ALK, can promote cell proliferation and non-anchorage-dependent growth in NIH-3T3 cells . The fusion protein activates ALK self-phosphorylation and downstream pathways including STAT3, AKT, and ERK signaling . Importantly, this fusion confers sensitivity to ALK tyrosine kinase inhibitors (TKIs) such as crizotinib and alectinib, as demonstrated both in vitro and in vivo in mouse xenograft models . This finding expands the spectrum of actionable ALK fusions in NSCLC and has direct clinical implications for targeted therapy.

What experimental approaches are recommended for studying PLCXD3 function in β-cells?

To investigate PLCXD3 function in β-cells, researchers should consider a multi-faceted approach:

  • Gene Expression Manipulation: RNA interference techniques (siRNA, shRNA) or CRISPR-Cas9 gene editing to silence or modify PLCXD3 expression in β-cell lines (e.g., INS-1 cells) or primary islets .

  • Functional Assays:

    • Glucose-stimulated insulin secretion (GSIS) assays to measure the impact on insulin release

    • Insulin content measurements using ELISA

    • Cell viability and apoptosis assays to assess cellular health

  • Molecular Profiling:

    • qRT-PCR or RNA-seq to analyze changes in expression of insulin signaling and biosynthesis genes

    • Western blotting to assess protein levels and phosphorylation status of insulin signaling pathway components

  • Correlation Analysis: In human islet samples, correlating PLCXD3 expression with clinical parameters (BMI, HbA1c) and expression of other genes (Insulin, GLP1R) .

These methods have successfully demonstrated the role of PLCXD3 in insulin production and secretion, making them validated approaches for further research in this area.

How can researchers effectively characterize novel PLCXD3 fusion proteins?

Based on the methodologies used to characterize the PLCXD3-ALK fusion, researchers should employ the following approaches to study novel PLCXD3 fusion proteins:

  • Identification Methods:

    • Next-generation sequencing (NGS) for initial detection

    • Validation by RT-PCR and Sanger sequencing

    • Immunohistochemistry (IHC) to confirm protein expression

  • Functional Characterization:

    • Generation of stable cell lines expressing the fusion protein using lentiviral transduction

    • Soft agar colony formation assays to assess anchorage-independent growth

    • Cell proliferation and viability assays

    • Transwell assays for migration and invasion capabilities

  • Signaling Pathway Analysis:

    • Western blotting to evaluate activation of the fusion protein (phosphorylation) and downstream signaling pathways

    • Inhibitor studies to determine dependency on specific pathways

  • In Vivo Validation:

    • Xenograft models in nude mice to assess tumorigenicity

    • Drug response studies in xenograft models to evaluate potential therapeutic targets

These methodologies provide a comprehensive framework for characterizing novel fusion proteins involving PLCXD3 and determining their functional significance and therapeutic implications.

What are the optimal conditions for producing and purifying recombinant PLCXD3 protein?

Based on established protocols for PLCXD3 recombinant protein production:

  • Expression System: E. coli is a suitable host for PLCXD3 expression, yielding a non-glycosylated polypeptide chain .

  • Protein Design:

    • Include the full 321 amino acid sequence

    • Add a His-tag (typically 23 amino acids) at the N-terminus to facilitate purification

    • Total molecular mass: approximately 38.7 kDa

  • Purification Method:

    • Affinity chromatography leveraging the His-tag

    • Additional proprietary chromatographic techniques to achieve >85% purity as verified by SDS-PAGE

  • Buffer Formulation:

    • 20mM Tris-HCl buffer (pH 8.0)

    • 20% glycerol

    • 0.15M NaCl

    • 1mM DTT

    • Recommended protein concentration: 0.25mg/ml

  • Storage Conditions:

    • Store at 4°C if using within 2-4 weeks

    • For longer storage, keep frozen at -20°C

    • Add carrier protein (0.1% HSA or BSA) for long-term storage

    • Avoid multiple freeze-thaw cycles

These conditions have been validated to produce functional PLCXD3 protein with preserved structural integrity suitable for downstream applications.

Table 1: Correlation of PLCXD3 Expression with Clinical Parameters and Gene Expression in Human Islets

ParameterCorrelation with PLCXD3SignificanceReference
Insulin expressionPositive correlationp < 0.05
GLP1R expressionPositive correlationp < 0.05
Body Mass Index (BMI)Negative correlationp < 0.05
HbA1cNegative correlationp < 0.05
Diabetic statusReduced in diabetic isletsp < 0.05

Table 2: Effects of PLCXD3 Silencing in INS-1 (832/13) Cells

ParameterEffect of PLCXD3 SilencingMagnitudeReference
Glucose-stimulated insulin secretionDecreasedSignificant reduction
Insulin contentDecreasedSignificant reduction
Insulin gene expressionDownregulatedSignificant reduction
NEUROD1 expressionDownregulatedSignificant reduction
GLUT2 expressionDownregulatedSignificant reduction
GCK expressionDownregulatedSignificant reduction
INSR expressionDownregulatedSignificant reduction
IRS2 expressionDownregulatedSignificant reduction
AKT expressionDownregulatedSignificant reduction
Cell viabilityUnaffectedNo significant change
Apoptosis rateUnaffectedNo significant change

Table 3: Functional Characteristics of PLCXD3-ALK Fusion

ParameterObservationComparison to EML4-ALKReference
ALK phosphorylationPositiveSimilar
STAT3 pathway activationIncreased phosphorylationSimilar
AKT pathway activationIncreased phosphorylationSimilar
ERK pathway activationIncreased phosphorylationSimilar
Colony formationEnhancedSimilar
Cell proliferationEnhancedSimilar
Tumorigenicity in vivoPositive (15/15 inoculations)Similar
Response to crizotinibSensitiveSimilar
Response to alectinibSensitiveSimilar
Clinical response in patientPartial response (PR)Typical for ALK fusions

What are the unresolved questions about PLCXD3 structure and function?

While current research has established important roles for PLCXD3, several critical questions remain unanswered:

  • Structural biology: The three-dimensional structure of PLCXD3 remains to be elucidated, which would provide insights into its mechanism of action and potential for drug targeting.

  • Enzymatic activity: While PLCXD3 contains a PI-PLC X-box domain, its specific substrates and the products of its enzymatic activity in different cellular contexts require further characterization .

  • Tissue-specific functions: Beyond pancreatic β-cells, the roles of PLCXD3 in other tissues remain largely unexplored, despite indications of its potential involvement in neurological disorders like Creutzfeldt-Jakob Disease .

  • Regulation mechanisms: The factors governing PLCXD3 expression, localization, and activity under normal and pathological conditions require further investigation.

Addressing these knowledge gaps would significantly advance our understanding of PLCXD3 biology and its therapeutic potential in metabolic and potentially other diseases.

How might PLCXD3 serve as a therapeutic target in metabolic disorders?

The research findings suggest several potential avenues for therapeutic targeting of PLCXD3 in metabolic disorders:

  • Gene therapy approaches: Increasing PLCXD3 expression in diabetic β-cells might restore insulin production and secretion capacity .

  • Small molecule modulators: Compounds that enhance PLCXD3 activity could potentially improve β-cell function in type 2 diabetes.

  • Pathway targeting: Understanding the downstream effectors of PLCXD3 signaling could reveal alternative therapeutic targets within the same pathway.

  • Biomarker utility: PLCXD3 expression levels might serve as a biomarker for β-cell function or predictor of diabetes risk or progression.

Future research should focus on validating these approaches through preclinical models and eventually clinical studies to determine the viability of PLCXD3-targeted therapies for diabetes and related metabolic disorders.

Product Science Overview

Introduction

Phosphatidylinositol-specific phospholipase C (PI-PLC) is an enzyme that plays a crucial role in cellular signaling by hydrolyzing phosphatidylinositol into diacylglycerol (DAG) and inositol trisphosphate (IP3). The X domain of PI-PLC is essential for its catalytic activity and interaction with other cellular components.

Classification

PI-PLC enzymes are classified into several isoforms based on their structure and function. The X domain is a conserved region found in various PI-PLC isoforms, including the human recombinant form. This domain is responsible for the enzyme’s ability to bind and hydrolyze phosphatidylinositol.

Biological Properties

The X domain of PI-PLC is characterized by its ability to interact with phospholipid membranes and catalyze the cleavage of phosphatidylinositol. This activity is crucial for the generation of second messengers, such as DAG and IP3, which are involved in various cellular processes, including cell growth, differentiation, and apoptosis.

Functions

The primary function of the PI-PLC X domain is to hydrolyze phosphatidylinositol into DAG and IP3. DAG acts as a secondary messenger that activates protein kinase C (PKC), while IP3 triggers the release of calcium ions from intracellular stores. These signaling molecules play a vital role in regulating various cellular functions, including metabolism, gene expression, and cell proliferation.

Modes of Action

The PI-PLC X domain operates by binding to the phosphatidylinositol substrate and catalyzing its hydrolysis through an intramolecular nucleophilic attack. This reaction results in the formation of DAG and IP3, which then participate in downstream signaling pathways. The X domain’s structure allows it to interact with other proteins and lipids, facilitating its role in cellular signaling.

Regulatory Mechanisms

The activity of the PI-PLC X domain is regulated by various factors, including phosphorylation, interaction with other proteins, and changes in cellular calcium levels. These regulatory mechanisms ensure that the enzyme’s activity is tightly controlled, preventing aberrant signaling that could lead to pathological conditions.

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.