PRNP Human

Prion Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

PRNP Gene Overview

Location: Chromosome 20 (20p13)
Protein product: Prion protein (PrP^C), a 253-amino acid glycoprotein anchored to cell membranes via glycosylphosphatidylinositol (GPI) .

Key Features

FeatureDescription
Isoforms- PrP^C: Normal, protease-sensitive form.
  • PrP^Sc: Pathogenic, protease-resistant form linked to prion diseases . |
    Expression | Highest in CNS neurons; moderate in heart, skeletal muscle, and immune cells . |
    Polymorphisms | Codon 129 (Met/Val) strongly influences prion disease susceptibility and phenotype . |

Normal Physiological Functions

PrP^C participates in:

  • Copper binding: Binds Cu²⁺ ions via octapeptide repeats, aiding cellular copper uptake .

  • Synaptic plasticity: Regulates NMDA receptor activity via S-nitrosylation, impacting memory consolidation .

  • Neuroprotection: Mitigates oxidative stress and apoptosis in neurons .

  • Myelin maintenance: Interacts with adhesion G-protein coupled receptor ADGRG6 to support peripheral nerve myelination .

Pathogenic Mutations

MutationAssociated DiseaseClinical Features
D178NFatal familial insomnia (FFI)Insomnia, autonomic dysfunction, thalamic degeneration .
P102LGerstmann-Sträussler-Scheinker (GSS)Ataxia, dementia, PrP amyloid plaques .
E200KGenetic Creutzfeldt-Jakob disease (gCJD)Rapid dementia, myoclonus, cerebellar signs .

Codon 129 Polymorphism (M129V)

  • Homozygosity (MM/VV): Increases risk of sporadic CJD (sCJD) and accelerates disease progression .

  • Heterozygosity (MV): Protective against sCJD in Caucasians (OR: 0.20 vs. MM) .

Transgenic Models

  • 117VV Tg30 mice: Express human PrP^117V and spontaneously develop transmissible PrP aggregates mimicking GSS pathology .

  • PrP-knockout mice: Exhibit circadian rhythm disruptions and synaptic deficits, highlighting PrP^C’s role in neuronal homeostasis .

Biomarkers and Diagnostics

  • Nanopore sequencing: Enables full-length PRNP analysis, detecting non-coding variants and repeat expansions .

  • Cerebrospinal fluid (CSF): RT-QuIC assays detect PrP^Sc with >90% sensitivity in sCJD .

Targeting PrP^Sc

  • Monoclonal antibodies (e.g., PRN100): Bind PrP^C to block conversion to PrP^Sc. Early trials show delayed progression in CJD patients .

  • Gene silencing: Antisense oligonucleotides reduce PRNP expression in preclinical models .

Challenges

  • Blood-brain barrier penetration: Limits antibody efficacy .

  • Somatic mosaicism: Non-uniform PRNP mutations complicate targeting .

Tissue-Specific Expression

Data from The Human Protein Atlas :

TissueExpression Level
Brain (cerebral cortex)High
HeartModerate
LiverLow
Immune cells (lymph nodes)Moderate

Unresolved Questions

  1. PrP^C’s exact role in copper metabolism: Mechanistic links to neurodegeneration remain unclear .

  2. Non-prion disease associations: GWAS studies show no significant links to traits beyond prion disorders .

  3. Species-specific effects: Protective PRNP variants (e.g., G127V in PNG populations) require further study .

Product Specs

Introduction
Prion protein (PRNP) is a membrane glycoprotein found throughout the body. Its misfolded form is believed to cause a group of central nervous system disorders known as Transmissible Spongiform Encephalopathies (TSE). PRNP has a region of five repeating octapeptide sequences. Mutations in this repeat region and other parts of the PRNP gene are linked to diseases like Creutzfeldt-Jakob disease, fatal familial insomnia, Gerstmann-Straussler disease, Huntington disease-like 1, and kuru.
Description
This product is a recombinant human PRNP protein produced in E. coli bacteria. It is a single chain of 229 amino acids (positions 23-230) without glycosylation, resulting in a molecular weight of 25kDa. It includes a 21 amino acid His-tag attached to the N-terminus for purification purposes, which is achieved through proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The PRNP protein is supplied in a solution at a concentration of 0.25mg/ml. The solution is buffered with 20mM Tris-HCl at pH 8.0 and contains 1M Urea and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the product frozen at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Avoid repeatedly freezing and thawing the product.
Purity
The purity of the PRNP protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
ASCR, CD230, CJD, GSS, MGC26679, prion, PRIP, PrP, PrP27-30, PrP33-35C, PrPc, Major prion protein, PRNP.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MKKRPKPGGW NTGGSRYPGQ GSPGGNRYPP QGGGGWGQPH GGGWGQPHGG GWGQPHGGGW GQPHGGGWGQ GGGTHSQWNK PSKPKTNMKH MAGAAAAGAV VGGLGGYVLG SAMSRPIIHF GSDYEDRYYR ENMHRYPNQV YYRPMDEYSN QNNFVHDCVN ITIKQHTVTT TTKGENFTET DVKMMERVVE QMCITQYERE SQAYYQRGS.

Q&A

Basic Research Questions

  • What is the normal function of the PRNP gene and its encoded protein?

    While the precise function of prion protein (PrP) remains incompletely understood, research points to several critical biological roles. Current evidence suggests PrP is involved in copper transport into cells, neuroprotection of brain cells against injury, and formation of synapses between neurons .

    Methodological approach: To investigate normal PrP function, researchers should employ conditional knockout models rather than complete deletion, as compensatory mechanisms may mask phenotypes. Cell-specific promoters driving Cre recombinase expression in PRNP-floxed mice enable temporal and spatial control of gene deletion. Complementary approaches include proteomic identification of binding partners through co-immunoprecipitation followed by mass spectrometry, and super-resolution microscopy to visualize subcellular localization at synaptic junctions.

  • What is the structural basis for prion protein conversion and pathogenicity?

    The human prion protein structure consists of a globular domain with three α-helices and a two-strand antiparallel β-sheet, an NH2-terminal tail, and a short COOH-terminal tail . A glycophosphatidylinositol (GPI) membrane anchor at the COOH-terminal tethers PrP to cell membranes, which proves integral to the transmission of conformational change; secreted PrP lacking this anchor component remains unaffected by the infectious process .

    Methodological approach: Structural biology techniques including X-ray crystallography, NMR spectroscopy, and cryo-electron microscopy should be employed to compare normal (PrPᶜ) and disease-associated (PrPˢᶜ) conformations. In vitro conversion assays using recombinant PrP can identify regions critical for misfolding. Site-directed mutagenesis of conserved residues followed by biophysical characterization helps determine structural elements essential for maintaining proper folding versus those promoting pathological conversion.

  • How are PRNP knockout models generated and what phenotypes do they display?

    Historically, generating PRNP knockout models was challenging due to its location in a densely packed heterochromatic region of chromosome 20 . Modern CRISPR-Cas9 gene editing has overcome these difficulties, enabling successful knockout of PRNP in embryonic stem cells in mice .

    Methodological approach: For CRISPR-Cas9 PRNP knockout, design multiple sgRNAs targeting conserved exonic regions, preferably early in the coding sequence. Validate knockout through genomic PCR, RT-PCR, and Western blotting for complete protein absence. PRNP knockout mice display no drastic changes in behavior, physiology, or immune function, though minor alterations in circadian rhythms have been observed . Notably, double knockout of both PRNP and SRNP (coding for the Shadoo protein) proves lethal, suggesting essential combined functions in embryogenesis .

  • How does PRNP expression vary across tissues and what regulates this expression?

    Expression of the prion protein is most predominant in the nervous system but occurs in many other tissues throughout the body . This tissue-specific expression pattern likely reflects complex transcriptional regulation mechanisms.

    Methodological approach: Single-cell RNA sequencing across multiple tissue types can map cell-specific expression patterns. ChIP-seq targeting transcription factors with putative binding sites in the PRNP promoter region helps identify key regulatory elements. DNA methylation analysis through bisulfite sequencing can reveal epigenetic regulation patterns. Reporter constructs with PRNP promoter fragments driving luciferase expression allow functional validation of regulatory elements in different cell types.

Advanced Research Questions

  • How can we design effective clinical trials for PRNP-targeted therapies given the rarity of prion diseases?

    The rarity, heterogeneity, and rapid progression of prion diseases create significant challenges for clinical trial design . These factors "profoundly frustrate the successful undertaking of well-powered therapeutic trials and patient identification in the asymptomatic or early stage before the development of significant brain damage" .

    Methodological approach: Innovative trial designs for rare prion diseases should incorporate: (1) adaptive trial designs allowing modification based on interim analyses, (2) basket trials grouping different prion disease subtypes to increase sample size, (3) biomarker-based endpoints including CSF/plasma neurofilament light chain and RT-QuIC seeding activity, (4) international research consortia to expand recruitment, and (5) prevention trials in asymptomatic PRNP mutation carriers where disease is predictable. Patient registries and natural history studies are essential prerequisites for trial planning.

  • How do PRNP and other prion-like protein genes interact in neurodegenerative processes?

    Research has identified interesting relationships between PRNP and other genes. PRND (prion-like protein doppel gene) is located 20 kilobase pairs downstream of PRNP, and strong linkage disequilibrium exists between PRNP and PRND SNPs in mammals susceptible to prion diseases . Additionally, SRNP (coding for the Shadoo protein) shows upregulation in prion-diseased mice, suggesting compensatory mechanisms .

    Methodological approach: To investigate these interactions, researchers should employ multi-omics approaches including: (1) transcriptome analysis comparing wild-type and PRNP knockout models during disease progression, (2) chromatin interaction studies using Hi-C to identify physical interactions between PRNP and PRND loci, (3) double and triple knockout/knockdown models to identify synthetic lethality or rescue phenotypes, and (4) protein interaction studies to map the interactome of PrP, Doppel, and Shadoo proteins in health and disease states.

  • What mechanisms underlie the selective neuronal vulnerability observed in PRNP-associated diseases?

    Despite widespread PRNP expression, prion diseases display region-specific patterns of neurodegeneration. Understanding this selective vulnerability is crucial for therapeutic development.

    Methodological approach: Research approaches should include: (1) single-nucleus RNA-seq of affected versus spared brain regions to identify cell type-specific vulnerability factors, (2) spatial transcriptomics to map regional variations in PRNP expression and processing, (3) comparative proteomics of affected versus resistant neuronal populations to identify protective or sensitizing factors, and (4) region-specific conditional PRNP knockout or restoration to establish causality in vulnerability patterns.

  • How can GWAS and other genomic approaches identify novel PRNP associations beyond prion disease?

    Current evidence suggests PRNP is predominantly associated with prion diseases, with limited evidence for other phenotypes . The GWAS Catalog includes four studies associating PRNP to sporadic and variant Creutzfeldt-Jakob disease , with one additional association to the KANNO blood group (a molecular measurement of PrP antigenicity in blood) .

    Methodological approach: Advanced genomic analyses to uncover novel PRNP associations should include: (1) gene-based association tests that aggregate signals across the PRNP locus, (2) rare variant association studies through exome or genome sequencing, (3) pleiotropic analyses to identify shared genetic effects across related traits, (4) integration of GWAS with epigenomic and transcriptomic data through approaches like Mendelian Randomization, and (5) systems biology approaches linking PRNP to broader gene networks and pathways using tools like MAGMA and DEPICT.

  • How does the prion protein interact with other misfolded proteins in neurodegenerative diseases?

    Evidence suggests PRNP may have broader roles in neurodegenerative pathology beyond prion diseases. PRNP displays antimicrobial activity, inhibiting viral replication, and directly interacts with Alzheimer's disease amyloid-β (Aβ) peptides .

    Methodological approach: To investigate these interactions, researchers should employ: (1) co-immunoprecipitation followed by mass spectrometry to identify disease-specific interaction partners, (2) proximity labeling techniques like BioID to capture transient interactions in living cells, (3) in vitro aggregation assays with purified proteins to assess cross-seeding potential, (4) double transgenic animal models co-expressing PRNP mutations and other neurodegenerative disease proteins, and (5) structural studies of co-aggregates using techniques like solid-state NMR.

  • What are the current approaches for detecting pathological prion protein in research and clinical settings?

    Detecting pathological prion conformations presents unique challenges due to their proteinaceous nature and the absence of nucleic acids.

    Methodological approach: A comprehensive detection strategy includes: (1) proteinase K digestion followed by Western blotting to distinguish protease-resistant PrPᵛᶜ, (2) conformation-dependent immunoassays using antibodies recognizing epitopes differentially exposed in normal versus misfolded states, (3) real-time quaking-induced conversion (RT-QuIC) assay for sensitive detection of seeding activity in CSF or other biofluids, (4) protein misfolding cyclic amplification (PMCA) for research applications requiring high sensitivity, and (5) novel approaches like conformation-specific aptamers or synthetic nanobodies for improved specificity.

  • How do epigenetic mechanisms regulate PRNP expression and influence disease susceptibility?

    Epigenetic regulation of PRNP may contribute to variable disease penetrance and age of onset in individuals carrying identical mutations.

    Methodological approach: Epigenetic studies should include: (1) genome-wide DNA methylation profiling comparing affected and unaffected mutation carriers, (2) ChIP-seq for histone modifications at the PRNP locus across brain regions and developmental stages, (3) identification of non-coding RNAs regulating PRNP expression through RNA-seq and functional validation, (4) epigenome editing using CRISPR-dCas9 fused to epigenetic modifiers to establish causality, and (5) longitudinal epigenetic profiling in accessible tissues (blood, CSF) to identify potential biomarkers of disease progression.

  • What is the feasibility of germline PRNP modification as a preventative strategy for familial prion diseases?

    Research has proposed investigations into heritable PRNP knockouts following the observation that PRNP knockout mice remain healthy and immune to prion disease .

    Methodological approach: Ethical and scientific assessment requires: (1) comprehensive phenotyping of PRNP knockout animals across their lifespan, including subtle cognitive, behavioral, and physiological measurements, (2) targeted PRNP modification rather than complete knockout, introducing protective polymorphisms, (3) mosaic approaches with partial editing to minimize developmental risks, (4) stem cell models comparing development with and without PRNP, and (5) careful ethical framework development balancing disease severity against germline modification concerns. Any clinical translation would require extensive preclinical safety data and robust ethical oversight.

Product Science Overview

Introduction

Prion proteins are a unique class of proteins that have garnered significant attention due to their role in neurodegenerative diseases. The term “prion” was first coined by Stanley Prusiner in 1982 to describe “proteinaceous infectious particles” responsible for diseases such as Creutzfeldt-Jakob disease (CJD), scrapie, and kuru . Prion proteins exist in two forms: the normal cellular prion protein (PrP^C) and the disease-causing scrapie prion protein (PrP^Sc).

Structure and Function

The prion protein (PrP) is encoded by the PRNP gene and is primarily expressed in the central and peripheral nervous systems . The normal cellular form, PrP^C, is a cell surface protein that plays a role in various cellular processes, including cell signaling and protection against oxidative stress . However, the exact physiological function of PrP^C remains not fully understood.

Pathogenesis

Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are caused by the conformational conversion of PrP^C into PrP^Sc . This misfolded form is resistant to protease digestion and tends to aggregate, leading to neuronal damage and the characteristic spongiform changes in the brain . The accumulation of PrP^Sc is associated with cell death, inflammation, and neurodegeneration .

Recombinant Prion Protein

Recombinant prion protein (human) is produced using recombinant DNA technology, which involves inserting the human PRNP gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant form is used extensively in research to study the structure, function, and pathogenic mechanisms of prion proteins .

Research and Therapeutic Implications

Research on prion proteins has provided valuable insights into the mechanisms of protein misfolding and aggregation, which are relevant not only to prion diseases but also to other neurodegenerative disorders like Alzheimer’s and Parkinson’s diseases . Understanding the structural features and toxicity of prion proteins can aid in the development of therapeutic strategies to combat these devastating diseases .

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.