PPARG Human

Peroxisome Proliferator Activated Receptor Gamma Human Recombinant
Shipped with Ice Packs
In Stock

Description

Peroxisome Proliferator Activated Receptor Gamma Human Recombinant is expressed in E.coli having a molecular weight of 59.2 kDa and fused to an amino terminal hexahistidine tag.

Product Specs

Introduction
The peroxisome proliferator-activated receptors (PPARs) are a group of ligand-activated transcription factors belonging to the nuclear receptor superfamily. They play crucial roles in adipogenesis, glucose homeostasis, and inflammation. There are three distinct isotypes: alpha, beta (also known as delta), and gamma. PPAR gamma is predominantly expressed in adipose tissue, with lower levels found in the colon, immune system, and retina. While PPAR alpha is involved in fatty acid catabolism in the liver, PPAR gamma regulates fatty acid storage in adipose tissue and contributes to adipocyte differentiation. PPAR gamma forms a heterodimer with retinoid X receptor (RXR) to bind to DNA. This binding, upon ligand activation, initiates the expression of target genes.
Description
Recombinant Human Peroxisome Proliferator Activated Receptor Gamma is produced in E. coli. It has a molecular weight of 59.2 kDa and includes an amino-terminal hexahistidine tag.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The protein is supplied in a solution containing 20mM Tris-HCl buffer and 50% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Repeated freezing and thawing should be avoided.
Purity
The purity of the protein is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
Peroxisome proliferator-activated receptor gamma, PPAR-gamma, PPARG, NR1C3, PPARG1, PPARG2.
Source
Escherichia Coli.

Q&A

What is PPARG and what are its primary functions in human physiology?

PPARG is a member of the peroxisome proliferator-activated receptor subfamily of nuclear receptors located on chromosome 3p25. It functions as a transcription factor by forming heterodimers with retinoid X receptors (RXRs) to regulate transcription of various genes . PPARG serves as a key regulator of:

  • Adipocyte differentiation and adipose tissue metabolism

  • Insulin sensitivity across multiple tissues

  • Inflammatory responses

  • Lipid metabolism and homeostasis

  • Placental development

  • Negative regulation of acute inflammatory responses

PPARG has been implicated in numerous pathological processes including obesity, diabetes, atherosclerosis, and various cancers . The receptor demonstrates both pro-differentiation effects in adipose tissue and anti-proliferative effects in certain cancer contexts, highlighting its context-dependent roles .

How is the PPARG gene structured and what transcript variants exist?

The human PPARG gene spans approximately 146,506 base pairs (12,287,850 bp to 12,434,356 bp) on the plus strand of chromosome 3 . Multiple transcript variants arise through:

  • Alternative promoter usage

  • Alternative splicing mechanisms

  • Different transcription start sites

Notable transcript variants include:

  • PPARG1 (PPARG1)

  • PPARG2 (PPARG2)

  • Dominant negative isoforms (including ORF4 variants)

  • A newly identified transcript: γ1ORF4

These variants exhibit tissue-specific expression patterns and contribute differentially to PPARG function. The canonical transcripts encode the full functional receptor, while dominant negative isoforms can inhibit PPARG activity, suggesting a complex regulatory network .

What computational tools and databases are most effective for analyzing PPARG SNPs?

Research has employed a multi-stage computational workflow for analyzing PPARG SNPs with the following key tools and databases :

  • SNP Retrieval and Filtering:

    • NCBI SNP Database: Primary source for retrieving all known PPARG SNPs

    • GeneMania: For gene network analysis and interaction mapping

  • Functional Impact Prediction:

    • SIFT (Sorting Intolerant From Tolerant): Identifies deleterious SNPs based on sequence homology

    • PolyPhen: Determines pathogenicity degree based on structural and functional parameters

    • I-Mutant: Assesses the effect of mutations on protein stability

    • PHD-SNP: Predicts disease-association of non-synonymous SNPs

  • Structural Analysis:

    • Project HOPE: Analyzes structural effects of mutations, providing 3D visualization

    • Chimera: Generates mutated 3D models of PPARG proteins for comparative analysis

This computational pipeline has successfully identified critical SNPs (e.g., rs72551364 and rs121909244) that affect PPARG function and contribute to human diseases including diabetes mellitus .

What genomic approaches are recommended for studying PPARG binding patterns and transcriptional activity?

A comprehensive approach to studying PPARG transcriptional activity requires integrated genomic methodologies :

  • ChIP-seq (Chromatin Immunoprecipitation Sequencing):

    • Maps genome-wide binding sites of PPARG

    • Identifies tissue-specific DNA occupancy patterns

  • RNA-seq:

    • Quantifies differential gene expression in response to PPARG activation/inhibition

    • Identifies splicing variants and their relative abundance

  • ATAC-seq (Assay for Transposase-Accessible Chromatin):

    • Identifies open chromatin regions accessible to PPARG binding

    • Maps regulatory elements involved in PPARG-mediated transcription

  • HiC and Chromosome Conformation Capture:

    • Analyzes long-range chromatin interactions mediated by PPARG

    • Identifies enhancer-promoter relationships

  • Integrated Multi-Omics Approach:

    • Combines sequence data with proteomics and metabolomics

    • Provides comprehensive understanding of PPARG signaling networks

As noted in the literature, "a combination of high-throughput sequencing applications and data integration is necessary to comprehensively understand transcriptional events in an unbiased, genome-wide manner during complex biological processes" .

How do PPARG mutations contribute to metabolic disorders?

PPARG mutations have significant impacts on human metabolic health through several mechanisms :

  • Diabetes and Insulin Resistance:

    • Loss-of-function mutations in PPARG are linked to severe insulin resistance

    • The P12A polymorphism affects progression to type 2 diabetes and response to thiazolidinedione drugs

    • Functional defects impair adipocyte insulin sensitivity and adipokine secretion

  • Lipodystrophy:

    • Certain PPARG mutations cause familial partial lipodystrophy

    • Results in abnormal fat distribution and metabolic dysfunction

    • Affected individuals show reduced adipose tissue in limbs with central fat accumulation

  • Hypertension:

    • PPARG regulates the renin-angiotensin system

    • Mutations can contribute to vascular tone dysregulation and hypertension

    • Often observed in conjunction with insulin resistance

  • Atherosclerosis:

    • Polymorphisms in PPARG have been associated with early atherosclerosis onset

    • Affects vascular inflammation and macrophage function in vessel walls

The clinical presentation of PPARG-related metabolic disorders typically includes a cluster of abnormalities (dyslipidemia, hypertension, glucose intolerance) that together increase cardiovascular risk substantially beyond individual components .

How does PPARG expression change during human adipocyte differentiation?

PPARG expression follows a distinct temporal pattern during human adipogenesis, with specific regulation of different transcript variants :

  • Temporal Expression Pattern:

    • Low baseline expression in undifferentiated mesenchymal stem cells

    • Significant upregulation during early differentiation (days 3-7)

    • Sustained high expression during terminal differentiation (days 14-21)

  • Differential Promoter Usage:

    • Early differentiation shows increased activity of the PPARG1 promoter

    • Later stages involve increased PPARG2 promoter activity

    • Promoter-specific regulation determines the ratio of isoforms throughout differentiation

  • Isoform-Specific Regulation:

    • Canonical transcripts (PPARG1 and PPARG2) increase substantially during differentiation

    • Dominant negative variants (including ORF4 variants) show a complex pattern

    • The γ1ORF4 variant has a unique temporal expression profile distinct from other variants

This differential regulation of PPARG transcript variants suggests a sophisticated mechanism for modulating adipogenesis, with dominant negative isoforms potentially serving as endogenous regulators of PPARG activity during specific phases of differentiation .

What is the role of PPARG in breast cancer and how does it interact with the immune microenvironment?

PPARG demonstrates complex roles in breast cancer biology with significant implications for both tumor progression and immune regulation :

  • Expression and Prognostic Value:

    • PPARG is generally downregulated in breast cancer compared to normal tissue

    • Expression levels correlate with pathological tumor stage (pT-stage) and TNM-stage

    • Higher expression in ER+ breast cancer correlates with better prognosis than in ER- tumors

  • Immune Microenvironment Interactions:

    • PPARG expression shows significant positive correlation with immune cell infiltration

    • Higher PPARG levels associate with better cumulative survival in breast cancer patients

    • Positive association with expression of immune-related genes and immune checkpoints

  • Pathway Involvement:

    • Strongly associated with angiogenesis, apoptosis, and fatty acid metabolism in ER+ breast cancer

    • Modulates key signaling pathways that affect cancer cell proliferation and survival

    • Influences inflammatory signaling within the tumor microenvironment

  • Therapeutic Implications:

    • Natural compounds that upregulate PPARG (particularly quercetin) show promise as anti-breast cancer agents

    • PPARG agonists may reduce breast cancer development by favorably modulating the immune microenvironment

    • ER+ patients with higher PPARG levels showed better responses to immune checkpoint blockade

These findings suggest PPARG may serve as both a prognostic marker and therapeutic target in breast cancer, with particular relevance to immunomodulatory approaches .

What methodological approaches can be used to investigate PPARG ligands for cancer therapy?

Investigating PPARG ligands for cancer therapy requires a multi-disciplinary approach combining computational, in vitro, and in vivo methodologies :

  • Computational Screening:

    • Molecular docking simulations to identify potential PPARG-binding compounds

    • Pharmacophore modeling to determine key structural features for PPARG activation

    • Database mining (e.g., BenCaoZuJian database for natural compounds)

  • In Vitro Validation:

    • Luciferase reporter assays to measure PPARG transcriptional activation

    • Competitive binding assays to determine binding affinity

    • Cancer cell line panels to assess anti-proliferative effects

    • Immune cell co-culture systems to evaluate immunomodulatory effects

  • Mechanistic Studies:

    • ChIP-seq to identify genomic binding sites affected by ligand treatment

    • RNA-seq to characterize transcriptional changes

    • Protein-protein interaction studies to assess co-activator/co-repressor recruitment

    • Metabolic profiling to evaluate effects on cancer cell metabolism

  • In Vivo Models:

    • Xenograft models to assess tumor growth inhibition

    • Syngeneic models to evaluate immune microenvironment effects

    • Patient-derived xenografts for translational relevance

    • Genetic models with PPARG modifications to determine specificity

The identification of quercetin as a promising PPARG-targeting natural compound for breast cancer treatment exemplifies the success of such integrated approaches .

How do dominant negative PPARG isoforms regulate canonical PPARG activity?

Dominant negative PPARG isoforms represent a sophisticated layer of endogenous regulation of PPARG activity :

  • Structural Basis of Dominant Negative Activity:

    • Dominant negative isoforms (including ORF4 variants) retain DNA-binding domains

    • They lack functional ligand-binding domains or contain truncated activation domains

    • This allows competitive binding to PPARG response elements without transcriptional activation

  • Temporal Regulation During Differentiation:

    • The γ1ORF4 transcript shows a distinct expression pattern during adipogenesis

    • Different ORF4 variants contribute differentially to the adipogenic process

    • Temporal coordination between canonical and dominant negative isoforms regulates differentiation

  • Mechanism of Action:

    • Competitive interference with DNA binding of canonical PPARG

    • Disruption of coactivator recruitment

    • Formation of non-functional heterodimers with RXR partners

    • Alteration of chromatin remodeling at PPARG target genes

  • Physiological Significance:

    • Fine-tuning of PPARG activity in different tissues and contexts

    • Potential role in preventing excessive adipogenesis

    • Contribution to metabolic flexibility in response to environmental changes

Understanding these dominant negative mechanisms provides crucial insights into PPARG regulation in both physiological and pathological conditions, suggesting potential therapeutic approaches targeting isoform ratios rather than total PPARG activity .

What is the relationship between PPARG and aging-related metabolic processes?

PPARG demonstrates significant connections to aging-related processes with implications for longevity and metabolic health :

These connections position PPARG as a potential therapeutic target for age-related metabolic dysfunction and suggest that modulating PPARG activity may have implications for healthy aging beyond individual disease states .

What are the most promising areas for future PPARG research?

Based on current literature, several promising research directions emerge for advancing PPARG research :

  • Isoform-Specific Biology:

    • Further characterization of dominant negative isoforms and their tissue-specific functions

    • Development of isoform-selective modulators to target specific PPARG functions

    • Investigation of isoform ratio changes in disease states

  • Genomic Integration Approaches:

    • Comprehensive mapping of the PPARG cistrome across tissues and conditions

    • Integration of chromatin conformation, transcriptome and epigenome data

    • Identification of tissue-specific enhancers and regulatory elements

  • Targeted Therapeutics:

    • Development of selective PPARG modulators with reduced side effects

    • Natural compound screening and optimization (e.g., quercetin derivatives)

    • Tissue-specific drug delivery approaches

  • Immune/Metabolic Interface:

    • Further characterization of PPARG's role at the intersection of immunity and metabolism

    • Investigation of PPARG in immune cell function during metabolic stress

    • Exploration of immunometabolic targeting in cancer and inflammatory diseases

  • Aging and Longevity:

    • Deeper investigation of PPARG-klotho axis in aging

    • Examination of PPARG modulation as a caloric restriction mimetic

    • Studies on PPARG's role in age-related tissue dysfunction

These research directions hold promise for translating our understanding of PPARG biology into therapeutic approaches for metabolic disease, cancer, and age-related conditions.

What methodological challenges remain in PPARG research?

Despite significant advances, several methodological challenges persist in PPARG research that require innovative approaches :

  • Structural Biology Limitations:

    • Obtaining complete crystal structures of full-length PPARG with co-regulators

    • Visualizing dynamic conformational changes during transcriptional activation

    • Structural characterization of dominant negative isoforms

  • Tissue and Context Specificity:

    • Developing models that recapitulate tissue-specific PPARG actions

    • Understanding contextual determinants of PPARG function (beneficial vs. detrimental)

    • Accounting for species differences in PPARG biology when translating findings

  • Systems Biology Integration:

    • Integrating multi-omics data to construct comprehensive PPARG signaling networks

    • Computational modeling of dynamic PPARG-mediated transcriptional responses

    • Capturing complex feedback mechanisms in PPARG regulation

  • Translational Barriers:

    • Developing tissue-selective PPARG modulators that maintain benefits while minimizing adverse effects

    • Identifying reliable biomarkers of PPARG activity in clinical settings

    • Designing appropriate clinical trials for PPARG-targeting therapies

  • Technical Limitations:

    • Achieving isoform-specific detection and manipulation in complex tissues

    • Developing methods for single-cell analysis of PPARG activity

    • Creating tools for temporal control of PPARG function in vivo

Addressing these methodological challenges will require interdisciplinary approaches combining structural biology, genomics, computational biology, and innovative in vivo models to fully unlock the therapeutic potential of PPARG modulation.

Product Science Overview

Introduction

Peroxisome Proliferator-Activated Receptor Gamma (PPAR-γ) is a ligand-activated nuclear receptor that plays a crucial role in the regulation of lipid and glucose homeostasis, inflammation, and cellular differentiation . It is a member of the nuclear receptor family and is involved in various physiological processes, including adipogenesis, insulin sensitivity, and vascular homeostasis .

Discovery and Structure

PPAR-γ was first identified in the early 1990s and has since been extensively studied for its role in adipocyte differentiation, maintenance, and function . The receptor has four functional domains: the N-terminal A/B domain, the DNA binding domain (DBD), the hinge domain, and the C-terminal ligand binding domain (LBD) . The LBD is responsible for ligand specificity and is crucial for homo- or hetero-dimerization, allowing interaction with co-repressors, coactivators, and other cofactors .

Mechanism of Action

PPAR-γ acts through the formation of heterodimers with retinoid X receptors (RXRs) to modify target gene expression at the transcriptional level in response to endogenous or synthetic ligands . When activated by its ligand, PPAR-γ can interact with specific DNA response elements to control gene transcription and expression . This regulation is essential for lipid metabolism, improving insulin sensitivity, modulating antitumor mechanisms, reducing oxidative stress, and inhibiting inflammation .

Therapeutic Potential

PPAR-γ has been shown to have significant therapeutic potential in various medical conditions. It has been studied for its neuroprotective effects in cerebral ischemic injury, where it helps prevent post-ischemic inflammation and neuronal damage . Additionally, PPAR-γ agonists have shown therapeutic effects in kidney conditions and various cancers . However, the associated side effects of synthetic agonists restrict their widespread use .

Recombinant Human PPAR-γ

Recombinant human PPAR-γ is produced using molecular cloning and transient expression techniques. One such method involves the use of an inducible Tet-On 3G system in human embryonic kidney 293T (HEK293T) cells . This system allows for high expression levels of PPAR-γ, making it a valuable model for studying the receptor’s function and potential therapeutic applications .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.