PARP1 Human

Poly (ADP-Ribose) Polymerase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of PARP1

PARP1 comprises six functional domains that enable DNA damage recognition and catalytic activity:

DomainFunction
Zinc-finger (Zn1-3)Binds DNA strand breaks and mediates chromatin interactions
BRCTFacilitates protein-protein interactions and auto-PARylation
WGRCooperates with Zn domains to regulate catalytic activation upon DNA binding
Helical (HD)Auto-inhibits NAD+ binding in inactive state
ARTCatalyzes ADP-ribose transfer using NAD+ as substrate

This modular architecture allows PARP1 to detect DNA lesions, recruit repair proteins, and modulate chromatin structure .

DNA Repair Mechanisms

PARP1 orchestrates multiple DNA repair pathways:

PathwayRole of PARP1
Base excision repairRecruits XRCC1 and other repair factors to single-strand breaks
Homologous recombinationFacilitates MRE11-mediated end resection at double-strand breaks
Non-homologous end joiningStabilizes broken DNA ends for ligation
Replication fork stabilizationPrevents collapse during replication stress

PARP1 also promotes chromatin relaxation via PARylation of histones, enabling repair machinery access .

Non-Repair Roles

  • Transcriptional regulation: Modulates NF-κB and glucocorticoid receptor activity .

  • Metabolic regulation: Essential for pancreatic β-cell differentiation and insulin secretion .

  • Inflammation: Contributes to cytokine production via PARylation of transcription factors .

Cancer Biology and Therapy

PARP1 is overexpressed in aggressive cancers, particularly triple-negative breast cancer (TNBC) and ovarian carcinomas :

Cancer TypePARP1 mRNA Fold Increase vs. NormalClinical Implication
Breast IDC1.63×TNBC shows highest PARP1 levels (HER2−: 2.0×)
Ovarian serous2.21×Correlates with platinum resistance
Lung adenocarcinoma1.67×Predicts poor survival

PARP inhibitors (PARPi) exploit synthetic lethality in BRCA1/2-deficient tumors by blocking residual DNA repair . Clinical trials show PARPi efficacy in ovarian (72% response rate) and prostate cancers .

Immune Modulation

PARP1 alterations correlate with improved response to immune checkpoint inhibitors (ICIs):

  • 2.9% of 45,604 tumors harbor PARP1 mutations/splice variants .

  • PARP1-altered tumors exhibit higher CD8+ T-cell infiltration (e.g., colorectal adenocarcinoma, p = 0.0061) .

  • Upregulated immune checkpoints: PD-1 (3.1×), CTLA-4 (2.7×), LAG3 (2.5×) in PARP1-altered groups .

Role in Metabolic Disorders

  • Diabetes: PARP1 knockout improves β-cell survival and insulin secretion in human islet models .

  • Muscle atrophy: PARP1 inhibition enhances myogenic differentiation and counters glucocorticoid-induced muscle wasting .

Aging and Longevity

  • PARP1 activity correlates with lifespan across 13 mammalian species (r = 0.76) .

  • Centenarians retain youthful PARP1 levels, enabling efficient oxidative DNA damage repair .

  • Werner syndrome (premature aging) involves defective PARP1-WRN protein interactions .

Expression and Regulation

  • Transcript variants: 26 splice isoforms identified, though most are non-coding .

  • Epigenetic control: Hypomethylation of ETS1-binding sites upregulates PARP1 in cancers .

  • Post-translational modification: Auto-PARylation reduces enzymatic activity via charge repulsion .

Therapeutic Challenges

  • Resistance mechanisms:

    • Restoration of homologous recombination (BRCA reversion) .

    • Upregulation of drug efflux pumps (ABCB1) .

  • Combination strategies:

    • PARPi + ICIs in TNBC (NCT04690868) .

    • PARPi + ATM inhibitors to overcome replication stress tolerance .

Product Specs

Introduction
Poly (ADP-ribose) polymerase 1 (PARP1) is an enzyme involved in DNA repair, particularly the base excision repair pathway. It catalyzes the addition of poly (ADP-ribose) chains to itself and other target proteins, including those involved in chromatin structure and DNA metabolism. PARP1 plays a role in various cellular processes, including DNA repair, transcription regulation, cell differentiation, proliferation, and response to cellular stress. Its dysregulation has been implicated in various diseases, including cancer and inflammatory disorders.
Description
Recombinant human PARP1, expressed in E. coli, is a non-glycosylated polypeptide chain comprising amino acids 662-1014 of the full PARP1 protein sequence. This truncated form of PARP1 retains its catalytic activity and is commonly used for in vitro studies. The protein has a molecular weight of approximately 39.6 kDa and is purified to high purity using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The PARP1 protein is supplied in a buffer consisting of 20mM Tris at pH 8.0, 1mM DTT (dithiothreitol), and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the PARP1 solution can be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. To ensure stability during long-term storage, adding a carrier protein such as 0.1% HSA (human serum albumin) or BSA (bovine serum albumin) is advised. Repeated freezing and thawing of the protein should be avoided to prevent degradation.
Purity
The purity of the PARP1 protein is determined to be greater than 95% using SDS-PAGE analysis, indicating a high level of protein purity with minimal contaminants.
Synonyms
ADPRT, ADPRT1, pADPRT, pADPRT-1, PARP, PARP-1, PPOL, Poly [ADP-ribose] polymerase 1, NAD(+) ADP-ribosyltransferase 1, Poly[ADP-ribose] synthase 1, PARP1.
Source
Escherichia Coli.
Amino Acid Sequence
MKSKLPKPVQ DLIKMIFDVE SMKKAMVEYE IDLQKMPLGK LSKRQIQAAY SILSEVQQAV SQGSSDSQIL DLSNRFYTLI PHDFGMKKPP LLNNADSVQA KAEMLDNLLD IEVAYSLLRG GSDDSSKDPI DVNYEKLKTD IKVVDRDSEE AEIIRKYVKN THATTHNAYD LEVIDIFKIE REGECQRYKP FKQLHNRRLL WHGSRTTNFA GILSQGLRIA PPEAPVTGYM FGKGIYFADM VSKSANYCHT SQGDPIGLIL LGEVALGNMY ELKHASHISK LPKGKHSVKG LGKTTPDPSA NISLDGVDVP LGTGISSGVN DTSLLYNEYI VYDIAQVNLK YLLKLKFNFK TSLW.

Q&A

What is PARP1 and where is it primarily located in human cells?

PARP1 (Poly[ADP-ribose] polymerase 1), also known as NAD+ ADP-ribosyltransferase 1 or poly[ADP-ribose] synthase 1, is an enzyme encoded by the PARP1 gene in humans. It is the most abundant member of the PARP family, accounting for approximately 90% of the NAD+ utilized by this enzyme family . PARP1 is predominantly localized in the cell nucleus, although cytosolic fractions have also been reported .

The protein functions by synthesizing poly ADP-ribose (PAR) using NAD+ as a substrate and transferring these PAR moieties to target proteins through a process called ADP-ribosylation . This post-translational modification is crucial for various cellular processes, particularly the DNA damage response.

How does PARP1 function in DNA damage repair pathways?

PARP1 acts as a first responder in DNA damage detection and facilitates the choice of appropriate repair pathways. When PARP1 detects DNA damage, it:

  • Binds to the damaged DNA site with high affinity

  • Becomes catalytically activated

  • Synthesizes PAR chains using NAD+ as a substrate

  • Modifies itself (auto-modification) and other target proteins

  • Recruits repair factors to the damage site

PARP1 contributes to repair efficiency through multiple mechanisms:

  • ADP-ribosylation of histones, which leads to chromatin decompaction and improves access to DNA damage sites

  • Interaction with and modification of various DNA repair factors

  • Involvement in multiple repair pathways including nucleotide excision repair, non-homologous end joining, microhomology-mediated end joining, homologous recombination, and DNA mismatch repair

For single-stranded DNA breaks, research has demonstrated that knocking down PARP1 expression with siRNA or inhibiting PARP1 activity with small molecules significantly reduces repair efficiency. When these breaks remain unrepaired and are encountered during DNA replication, replication forks stall and double-strand breaks accumulate .

What experimental evidence links PARP1 to human longevity?

Several lines of evidence connect PARP1 activity to longevity:

  • Comparative studies across 13 mammalian species (including rat, guinea pig, rabbit, marmoset, sheep, pig, cattle, chimpanzee, horse, donkey, gorilla, elephant, and human) have shown that PARP activity in permeabilized mononuclear leukocyte blood cells correlates positively with maximum species lifespan .

  • Lymphoblastoid cell lines established from human centenarians (individuals 100 years or older) demonstrate significantly higher PARP activity compared to cell lines from younger individuals (20-70 years old) .

  • PARP1 and Wrn proteins (the latter being deficient in Werner syndrome, a human premature aging disorder) form a complex involved in DNA break processing .

  • PARP1 can counteract reactive oxygen species production, potentially contributing to longevity by inhibiting oxidative damage to DNA and proteins .

  • Constitutive expression of PARP1 decreases by approximately 50% in cells from older adults (69-75 years) compared to young adults (19-26 years), yet remarkably, centenarians (100-107 years) maintain PARP1 expression levels similar to those of young individuals .

These findings collectively support the DNA damage theory of aging and suggest that PARP1-mediated DNA repair capability is a significant factor in mammalian longevity.

What is the precise chemical mechanism of poly(ADP-ribosyl)ation catalyzed by PARP1?

The poly(ADP-ribosyl)ation (PARylation) reaction catalyzed by PARP1 involves the synthesis of linear or branched chains of ADP-ribose units derived from NAD+ . The reaction mechanism occurs in distinct phases:

  • Initiation phase: PARP1 transfers the first ADP-ribose unit from NAD+ to an acceptor amino acid residue (typically glutamate, aspartate, serine, or lysine) on the target protein.

  • Elongation phase: Additional ADP-ribose units are added to form a linear chain through glycosidic ribose-ribose bonds between the ADP-ribose units.

  • Branching phase: Branch points can be created in the PAR chain by forming additional glycosidic bonds.

The direction of chain growth during PAR elongation has been debated, with two models proposed:

  • Distal addition model (tail-out mechanism): The established model where new ADP-ribose units are added to the 2'-OH terminus of the growing chain distal to the PARylation target .

  • Proximal addition model (head-out mechanism): An alternative model where the PAR chain grows by adding new ADP-ribose units at the 1'' terminus adjacent to the protein being modified .

Current evidence supports the distal addition model, especially considering that PARP1 catalyzes initiation in a distributive manner and elongation in a processive manner, with the elongation rate exceeding that of initiation by 232 times .

How does PARP1 efficiently locate DNA damage sites in the nucleus?

PARP1 uses a sophisticated mechanism called the "monkey bar" mechanism (also known as intersegment transfer) to efficiently navigate the nuclear environment and locate DNA damage sites. This process involves:

  • PARP1 initially binds to DNA in a diffusion-limited manner, with an association rate constant (k₁) of approximately 3.1 nM⁻¹s⁻¹ .

  • Once bound to DNA, PARP1 can release from this site by binding to a second DNA molecule, facilitating transfer between DNA segments (like moving between monkey bars) .

  • The WGR domain of PARP1 is essential for this intersegment transfer mechanism. Research has shown that a point mutation (W589A) in this domain significantly alters the kinetics of PARP1-DNA interactions .

  • This mechanism allows PARP1 to rapidly scan large amounts of DNA without getting "stuck" on undamaged portions, explaining how it can effectively find damage sites among the overwhelming amount of intact DNA in the nucleus .

The physiological importance of this monkey bar mechanism has been demonstrated experimentally: the W589A mutant accumulates at sites of DNA damage more slowly following laser micro-irradiation than wild-type PARP1 .

What factors influence PARP1 target specificity during PARylation?

PARP1 target specificity during PARylation is influenced by several factors:

  • Interaction with regulatory proteins: The most notable example is Histone PARylation Factor 1 (HPF1), which switches PARP1 target specificity to serine residues . Two highly conserved amino acid residues in the C-terminal region of HPF1, Tyr238 and Arg239, play crucial roles in HPF1-PARP1 interaction .

  • Target protein structure: The accessibility of potential acceptor amino acids affects whether they can be modified by PARP1.

  • Acceptor amino acid type: PARP1 can modify various amino acid residues including glutamate, aspartate, serine, and lysine, though with different efficiencies.

  • Conformational changes in PARP1: HPF1 may alter the conformation of the D-loop in PARP1's catalytic domain, explaining how it regulates target specificity .

Interestingly, when HPF1 directs PARP1 to modify serine residues, the modification appears to be primarily mono(ADP-ribosyl)ation rather than poly(ADP-ribosyl)ation . This demonstrates how regulatory proteins can influence not only target specificity but also the extent of modification.

What are the key kinetic parameters of PARP1-DNA interactions?

PARP1-DNA interaction kinetics are crucial for understanding PARP1's function in DNA damage recognition and repair. Key parameters from experimental studies include:

  • Association rate constant (k₁): 3.1 nM⁻¹s⁻¹ for PARP1 binding to double-strand breaks, which is significantly faster than previous estimates using surface plasmon resonance .

  • Dissociation rate constant (k₋₁): An upper bound of 10 s⁻¹ has been derived from global fitting of experimental data .

  • Equilibrium dissociation constant (KD): <3 nM for double-strand breaks under experimental conditions, which is lower than previously reported values of 31 nM, 14 nM, and 97 nM .

These parameters indicate that PARP1 association with DNA is diffusion-limited, allowing for rapid scanning of the genome to locate damage sites. The relatively slow dissociation rate suggests stable binding once PARP1 locates damaged DNA.

How can researchers measure PARP1 enzymatic activity in experimental settings?

Several experimental approaches can be employed to measure PARP1 enzymatic activity:

  • Radioactive NAD⁺ incorporation assays: Using [³²P]-labeled NAD⁺ as a substrate to track PAR synthesis and incorporation into target proteins.

  • Pulse-and-chase experiments: Alternating between radioactive and non-radioactive NAD⁺ to determine the mechanism of PAR chain elongation .

  • Click chemistry with fluorescent dyes: The use of 2'-deoxy NAD⁺ analogues as chain terminators, followed by click chemistry with fluorescent dyes to detect incorporation into PAR chains .

  • PARG treatment experiments: Poly(ADP-ribose)-glycohydrolase (PARG) degrades PAR chains primarily from the terminus distal to the protein. Treatment with PARG followed by analysis of remaining PAR can provide insights into PAR chain structure and elongation mechanisms .

  • Competition assays: Pre-forming complexes between PARP1 and fluorescently labeled DNA, then using excess unlabeled DNA to compete for PARP1 binding. This approach can measure dissociation rates and study the intersegment transfer mechanism .

  • Laser micro-irradiation: Used to induce DNA damage in living cells and measure the rate of PARP1 accumulation at damage sites, particularly useful for comparing wild-type and mutant PARP1 variants .

What experimental approaches are used to study PARP1's role in chromatin regulation?

Researchers employ various methodologies to investigate PARP1's function in chromatin regulation:

  • Chromatin immunoprecipitation (ChIP): To identify genomic regions where PARP1 binds and to study its co-localization with other chromatin factors.

  • Histone modification analysis: To examine how PARP1-mediated PARylation affects histone modifications and chromatin structure.

  • Micrococcal nuclease (MNase) sensitivity assays: To assess chromatin accessibility changes induced by PARP1 activity.

  • PARP1 domain mutant studies: Creating specific mutations or domain deletions (such as the WGR domain mutation W589A) to examine how different structural elements contribute to PARP1's chromatin functions .

  • In vitro reconstitution systems: Reconstructing chromatin with purified components to study direct effects of PARP1 and PARylation on nucleosome structure and dynamics.

  • Live-cell imaging: Using fluorescently tagged PARP1 to visualize its dynamics and interactions with chromatin in living cells, particularly in response to DNA damage.

What is the significance of PARP1 in cancer biology and treatment strategies?

PARP1 plays a critical role in cancer biology through several mechanisms:

  • Synthetic lethality: Cells with defective homologous recombination repair (such as those with BRCA1/2 mutations) become highly dependent on PARP1-mediated DNA repair. When PARP1 is inhibited in these cells, DNA damage accumulates to lethal levels, providing the basis for synthetic lethality-based cancer treatments .

  • DNA repair in cancer cells: Cancer cells often have higher levels of DNA damage and may rely more heavily on DNA repair mechanisms, including those involving PARP1.

  • Inflammation and tumor microenvironment: PARP1 is involved in inflammatory processes that can contribute to cancer progression .

  • Cellular proliferation: PARP1 participates in pathways regulating cellular proliferation and can be activated by factors such as Helicobacter pylori in gastric cancer development .

Experimental approaches to study PARP1 in cancer include:

  • Cancer cell line studies comparing PARP1 inhibitor sensitivity between BRCA-deficient and BRCA-proficient cells

  • Patient-derived xenograft models

  • Analysis of PARP1 expression and activity in different cancer types

  • Combination therapy studies to identify synergistic drug interactions with PARP inhibitors

How do researchers differentiate between PARP1's enzymatic versus scaffolding functions in experimental settings?

Distinguishing between PARP1's catalytic and structural roles requires sophisticated experimental approaches:

  • Catalytically dead mutants: Creating PARP1 variants with mutations in the catalytic domain that eliminate enzymatic activity while preserving protein structure and DNA binding capability.

  • Chemical genetic approaches: Using analog-sensitive PARP1 mutants that can be selectively inhibited by specific compounds, allowing temporal control of PARP1 inhibition.

  • Domain-specific mutations: Introducing mutations in specific domains (such as the WGR domain W589A mutation) to selectively alter certain functions while preserving others .

  • Separation-of-function mutants: Developing PARP1 variants that retain catalytic activity but are deficient in specific protein-protein interactions.

  • Rapid protein depletion systems: Using technologies like auxin-inducible degron systems to rapidly deplete PARP1 protein and distinguish between immediate effects (likely dependent on enzymatic activity) versus delayed effects (potentially related to scaffolding functions).

  • Sequential inhibition and knockout studies: Comparing the effects of catalytic inhibition versus complete protein depletion to identify phenotypes specifically linked to scaffolding functions.

How does the relationship between PARP1 and cellular metabolism impact DNA repair capacity?

The interplay between PARP1 and cellular metabolism represents a frontier in PARP1 research:

  • NAD⁺ consumption: PARP1 is the most abundant PARP family member, accounting for 90% of cellular NAD⁺ used by this enzyme family . This significant NAD⁺ consumption links PARP1 activity directly to cellular metabolic state.

  • Resveratrol connection: PARP1 appears to be resveratrol's primary functional target through interaction with tyrosyl tRNA synthetase (TyrRS). Under stress conditions, TyrRS translocates to the nucleus and stimulates NAD⁺-dependent auto-poly-ADP-ribosylation of PARP1, altering its functions from a chromatin architectural protein to a DNA damage responder and transcription regulator .

  • Age-related NAD⁺ decline: The age-associated decrease in NAD⁺ levels may impact PARP1 activity and subsequently DNA repair capacity, potentially contributing to the aging phenotype.

Research approaches to address this question include:

  • Metabolic profiling of cells with different PARP1 activity levels

  • Investigation of how nutrient availability affects PARP1-mediated DNA repair

  • Analysis of how NAD⁺ precursors impact PARP1 function in aging models

  • Study of the reciprocal regulation between PARP1 and metabolic enzymes

What explains the apparent contradictions in PARP1 experimental data across different model systems?

Researchers have noted inconsistencies in PARP1 data across different experimental systems, which may be attributed to several factors:

  • PARP1's dual role as catalyst and substrate: PARP1 functions simultaneously as an enzyme and as a target for auto-modification, making it challenging to interpret experimental results .

  • Variation in experimental conditions: Different buffer conditions, DNA structures, NAD⁺ concentrations, and presence of regulatory proteins can significantly alter PARP1 activity and function.

  • Stoichiometry considerations: The number of PARP1 molecules involved in the auto-modification reaction may vary between experimental setups .

  • Species-specific differences: While PARP1 is highly conserved, subtle differences between species may contribute to inconsistent results when comparing data from different model organisms.

  • Technical limitations: Variations in detection methods, purification procedures, and experimental timescales can lead to apparent contradictions.

To address these contradictions, researchers can:

  • Standardize experimental conditions

  • Directly compare different model systems within the same study

  • Use multiple complementary techniques to validate findings

  • Carefully control the stoichiometry of reaction components

  • Consider the influence of regulatory proteins such as HPF1

How can emerging technologies advance our understanding of PARP1 dynamics in living systems?

Emerging technologies offer new opportunities to study PARP1 in more physiologically relevant contexts:

  • Live-cell single-molecule tracking: To observe PARP1 movement and DNA interaction kinetics in real-time within living cells, providing insights into the "monkey bar" mechanism in vivo .

  • Cryo-electron microscopy: To visualize PARP1-DNA complexes and conformational changes associated with PARP1 activation at near-atomic resolution.

  • CRISPR-based genomic approaches: For precise modification of PARP1 and its regulatory partners to study structure-function relationships in endogenous contexts.

  • Optical control technologies: Using optogenetic tools to achieve spatiotemporal control of PARP1 activity in specific cellular compartments.

  • Mass spectrometry-based interactomics: To comprehensively identify PARP1 interaction partners and PARylation targets under different cellular conditions.

  • Integrative multi-omics approaches: Combining transcriptomics, proteomics, metabolomics, and genomics to understand PARP1's global impact on cellular function.

  • Computational modeling: Developing mathematical models of PARP1 kinetics and interaction networks to predict behavior under various conditions and generate testable hypotheses.

These technological advances will help resolve longstanding questions about PARP1 function and potentially identify new therapeutic opportunities targeting PARP1 and its regulatory network.

Product Science Overview

Structure and Function

PARP1 is primarily located in the cell nucleus, although a cytosolic fraction has also been reported . The enzyme uses NAD+ to synthesize poly ADP-ribose (PAR) and transfers PAR moieties to proteins in a process known as ADP-ribosylation . This modification is essential for various cellular functions, including:

  • DNA Repair: PARP1 acts as a first responder to DNA damage, detecting single-strand and double-strand breaks and facilitating the choice of repair pathways . It contributes to repair efficiency by ADP-ribosylating histones, leading to chromatin decompaction, and by interacting with multiple DNA repair factors .
  • Chromatin Remodeling: PARP1’s role in chromatin remodeling is crucial for gene expression regulation .
  • Cell Differentiation and Proliferation: PARP1 is involved in cell differentiation, proliferation, and tumor transformation .
  • Inflammation and Disease: The enzyme is implicated in the induction of inflammation and the pathophysiology of diseases such as type I diabetes .
Recombinant PARP1

Human recombinant PARP1 is produced using recombinant DNA technology, which involves inserting the PARP1 gene into a suitable expression system, such as bacteria or yeast, to produce the enzyme in large quantities. This recombinant enzyme is used in various research applications, including studies on DNA repair mechanisms, drug development, and cancer research.

Therapeutic Implications

PARP1 has emerged as a significant therapeutic target, particularly in cancer treatment. Inhibitors of PARP1 have shown promise in treating cancers with defects in DNA repair pathways, such as BRCA-mutated breast and ovarian cancers . By inhibiting PARP1, these drugs exploit the concept of synthetic lethality, where the inhibition of PARP1 in cells already deficient in DNA repair leads to cell death.

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.