Recombinant Human Protein-arginine deiminase type-2 (PADI2)

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant PADI2

Recombinant PADI2 is produced using heterologous expression systems such as Baculovirus-infected Sf9 insect cells or other mammalian systems . The full-length protein (1–665 amino acids) has a molecular weight of ~75 kDa and is purified to ≥61% purity . Its enzymatic activity is calcium-dependent, requiring 10 mM CaCl₂ for optimal function .

Table 1: Kinetic Parameters of PADI2

ParameterValueConditionsSource
kcat/Km (AcH4–15)1.4 × 10⁴ M⁻¹min⁻¹pH 7.5, 10 mM CaCl₂
pH optimum7.5–7.6Bell-shaped profile
Calcium dependenceEC₅₀ = 0.5–1.0 mMRequired for activation

Physiological Functions

  • Myelination: Citrullinates myelin basic protein (MBP) in neuronal cells .

  • Gene regulation: Citrullinates histone H3 at R26, promoting estrogen receptor (ER)-mediated transcription .

  • Bone formation: Stabilizes RUNX2 by citrullinating R381, preventing proteasomal degradation .

Table 2: Diseases Linked to PADI2 Dysregulation

DiseaseMechanismKey FindingsSource
Multiple sclerosisHyper-citrullination of MBPDisrupts myelin structure
Rheumatoid arthritis (RA)Synovial citrullination triggers ACPAPAD2 found in RA synovial fluid
Breast cancerHER2/ERBB2-driven PADI2 overexpressionCorrelates with luminal subtype

Inhibitor Development

Second-generation inhibitors like BB-Cl-amidine exhibit:

  • Enhanced potency: kinact/KI = 61,600 M⁻¹min⁻¹ for PADI2 .

  • Selectivity: >100-fold selectivity over PAD4 .

Preclinical Efficacy

  • Breast cancer: Cl-amidine reduces MCF10DCIS tumor growth by 3-fold in xenografts .

  • RA: Inhibitors suppress extracellular citrullinated antigen release .

Research Advancements

  • Citrullinome profiling: LC-MS/MS identified 10 citrullination sites on RUNX2, with R381 critical for stability .

  • ATP-induced activation: Mast cell PADI2 is activated via P2X7 receptors, linking purinergic signaling to autoimmunity .

Expression and Purification Challenges

  • Yield: Recombinant PADI2 is typically obtained at 4 mg/L .

  • Stability: Requires storage at -80°C with reducing agents (e.g., DTT, 2-mercaptoethanol) .

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format that is currently in stock. However, if you have a specific format requirement, please indicate it in your order notes. We will prepare the product according to your request.
Lead Time
Delivery time may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timeframes.
Note: All our proteins are shipped with standard blue ice packs by default. If you require dry ice shipping, please communicate with us in advance as additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%. Customers can use this as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer components, storage temperature, and the protein's intrinsic stability.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type is determined during the production process. If you have a specific tag type preference, please inform us, and we will prioritize the development of that tag.
Synonyms
KIAA0994; OTTHUMP00000044625; PAD 2; PAD H19 ; PAD-H19; PAD2; PADI 2; Padi2; PADI2 protein; PADI2_HUMAN; PDI 2; PDI2; Peptidlyarginine deiminase type II; Peptidyl arginine deiminase II; Peptidyl arginine deiminase type II; Peptidylarginine deiminase II; Protein arginine deiminase; Protein arginine deiminase type 2; Protein arginine deiminase type II; Protein-arginine deiminase type II; Protein-arginine deiminase type-2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-665aa
Mol. Weight
77.5kD
Protein Length
Full length
Purity
>85% (SDS-PAGE)
Research Area
Epigenetics and Nuclear Signaling
Species
Homo sapiens (Human)
Target Names
PADI2
Target Protein Sequence
MLRERTVRLQYGSRVEAVYVLGTYLWTDVYSAAPAGAQTFSLKHSEHVWVEVVRDGEAEEVATNGKQRWLLSPSTTLRVTMSQASTEASSDKVTVNYYDEEGSIPIDQAGLFLTAIEISLDVDADRDGVVEKNNPKKASWTWGPEGQGAILLVNCDRETPWLPKEDCRDEKVYSKEDLKDMSQMILRTKGPDRLPAGYEIVLYISMSDSDKVGVFYVENPFFGQRYIHILGRRKLYHVVKYTGGSAELLFFVEGLCFPDEGFSGLVSIHVSLLEYMAQDIPLTPIFTDTVIFRIAPWIMTPNILPPVSVFVCCMKDNYLFLKEVKNLVEKTNCELKVCFQYLNRGDRWIQDEIEFGYIEAPHKGFPVVLDSPRDGNLKDFPVKELLGPDFGYVTREPLFESVTSLDSFGNLEVSPPVTVNGKTYPLGRILIGSSFPLSGGRRMTKVVRDFLKAQQVQAPVELYSDWLTVGHVDEFMSFVPIPGTKKFLLLMASTSACYKLFREKQKDGHGEAIMFKGLGGMSSKRITINKILSNESLVQENLYFQRCLDWNRDILKKELGLTEQDIIDLPALFKMDEDHRARAFFPNMVNMIVLDKDLGIPKPFGPQVEEECCLEMHVRGLLEPLGLECTFIDDISAYHKFLGEVHCGTNVRRKPFTFKWWHMVP
Uniprot No.

Target Background

Function
Catalyzes the deimination of arginine residues within proteins.
Gene References Into Functions
  1. The mRNA expression of PADI2, PADI4, and Sp1 is upregulated in rheumatoid arthritis bone marrow CD34+ cells, independent of systemic inflammation or treatment regimen. PMID: 29148420
  2. These data suggest that overexpression of the human PAD2 transgene in the epidermis of transgenic mice increases the malignant conversion rate of benign tumors by promoting an inflammatory microenvironment. PMID: 28766045
  3. Brain gene expression of PADI2, ZNF385A, PSD2, and A2ML1, as well as DNA methylation dysregulations, are implicated in the alteration of brain tissue properties associated with late-life cognitive decline, beyond the influence of common neuropathologic conditions. PMID: 29084334
  4. Peptidyl arginine deiminase 2 (PADI2) is essential for the activation of androgen receptor (AR) signaling under androgen-deprived conditions. PMID: 28819028
  5. Data suggest that protein-arginine deiminase 2 (PADI2) suppresses the proliferation of colonic epithelial cells through the catalysis of protein citrullination. Consequently, downregulation of PADI2 expression might contribute to colon carcinogenesis. PMID: 28403548
  6. Downregulation of PADI2 is an early event in the pathogenesis of colorectal cancer, associated with poor prognosis. This suggests a potential role of citrullination in modulating tumor cells and their microenvironment. PMID: 27280713
  7. Multiple proteins citrullinated by hypoxia-induced PADs were identified. Furthermore, the extracellular domain of vascular endothelial growth factor receptor 2 was citrullinated by human PAD2 in vitro. These findings may contribute to understanding the pathophysiology of malignant gliomas from the perspective of protein citrullination. PMID: 27818200
  8. Deimination of myelin basic protein (MBP) by peptidylarginine deiminase (PAD) prevents its binding to the proteasome and decelerates its degradation by the proteasome in mammalian cells. The potential anticancer drug tetrazole analogue of chloramidine 2, at concentrations greater than 1 microM, inhibits the enzymatic activity of PAD in vitro. PMID: 27599511
  9. This study demonstrates that miR-4728 downregulates PADI2, a novel rheumatoid arthritis risk gene. PMID: 26927695
  10. We identified the presence of PADI3 mRNA expression in synovial tissue and PADI2 and PADI4 mRNA expressions in fibroblast-like synoviocytes from patients with rheumatoid arthritis. PMID: 26255191
  11. Protein arginine deiminase 2 binds six calcium ions in an ordered fashion. PMID: 25621824
  12. PAD2 activity was significantly higher in cell-free synovial fluid of rheumatoid arthritis patients compared to osteoarthritis patients. PMID: 26245941
  13. PAD2 activity was detected in synovial fluid samples from patients with rheumatoid arthritis. PMID: 25475141
  14. Studies report increased levels of extracellular PAD2 in the lungs of smokers. PMID: 25897949
  15. PADI2 and vimentin participate in the apoptotic mechanisms of activated T lymphocytes. PMID: 24850148
  16. These studies provide the first genetic evidence that PAD2 functions as an oncogene and suggest that PAD2 may promote tumor progression by enhancing inflammation within the tumor microenvironment. PMID: 25213324
  17. PAD2 appears to utilize a substrate-assisted mechanism of catalysis where the positively charged substrate guanidinium depresses the pKa of the nucleophilic cysteine. PMID: 24989433
  18. Data suggest peptidylarginine deiminase 2 (PAD2) as a potential biomarker in various inflammatory diseases. PMID: 24384061
  19. PAD2 and PAD4 have distinct substrate specificities. PMID: 24594197
  20. These findings suggest that PAD2 and citrullinated proteins may play a crucial role in the brain pathology of prion diseases. [review] PMID: 23022892
  21. Our observations show increased levels of protein deimination, but not PAD2, in age-related macular degeneration retinas and retinal pigment epithelium, suggesting a reduced rate of turnover of deiminated proteins. PMID: 23562679
  22. PAD2 binds directly to the promoters of the PTN and MAGEA12 genes, and the likely mechanism by which PAD2 regulates the expression of these genes is via citrullination of arginine residues 2-8-17 on histone H3 tails. PMID: 22911765
  23. Contact between stimulated T cells and monocyte-macrophages or cytokine-activated monocyte-macrophages constitutes a highly likely source of PAD2 and PAD4, which are observed in inflamed synovial tissues. PMID: 22614825
  24. Normal human and canine mammary epithelium showed strong cytoplasmic and nuclear expression of PAD2, but there was reduced PAD2 expression in mammary carcinomas from both species. PMID: 22520816
  25. 17beta-estradiol stimulation induces the recruitment of PAD2 to target promoters by ERalpha, whereby PAD2 then citrullinates H3R26, leading to local chromatin decondensation and transcriptional activation. PMID: 22853951
  26. Defective regulation of PAD2 in the peripheral blood, without the immunological shelter of the blood-brain barrier, may contribute to the development of autoimmune responses in MS. PMID: 21878453
  27. This is the first report demonstrating that, similar to primary open angle glaucoma, normal tension glaucoma also possesses elevated levels of both PAD2 and protein-bound citrulline. PMID: 20806090
  28. PAD2 activation and aberrant citrullinated proteins could play a role in pathogenesis and have value as a marker for the postmortem classification of neurodegenerative diseases. PMID: 20013286
  29. PADI2 does not contribute to genetic susceptibility to schizophrenia. PMID: 19478818
  30. PAD2 is expressed in human monocytic leukemia THP-1 cells during differentiation into macrophages. PMID: 19564157
  31. Molecular cloning and gene organization; expressed by all the living epidermal layers, suggesting that PAD type II is functionally important during terminal differentiation of epidermal keratinocytes. PMID: 12392711
  32. The first report to demonstrate a measurable response in the amounts of peptidylarginine deiminase type II mRNA, protein, and activity in human astrocytes by prolonged hypoxic exposure. PMID: 15555572
  33. hPADI2 and hPADI4 have distinct roles under physiological and pathological conditions. PMID: 15629448
  34. The amount of peptidyl arginine deiminase type II enzyme and citrullinated myelin basic protein was increased in multiple sclerosis. PMID: 17469138
  35. PAD-2 & PAD-4 are the only isotypes expressed in synovial tissue in rheumatoid arthritis and other arthritides. Inflammatory cells are the primary source, but PAD-4 also originates from hyperplastic synoviocytes. Both isotypes are likely involved in citrullination of fibrin. PMID: 17968929
  36. These data provide new structure-function dimensions for chemokines in leukocyte mobilization, disclosing an anti-inflammatory role for PAD. PMID: 18645041
  37. The citrullinating enzyme PAD-4 was detected in synovial fluid from patients with rheumatoid arthritis and spondylarthritides. PMID: 18668562
  38. Results describe the in vitro kinetic properties of the human peptidylarginine deiminase isoform 2 (hPAD2) and explore the putative inhibitory action of the methyl ester side chain of paclitaxel. PMID: 18923545

Show More

Hide All

Database Links

HGNC: 18341

OMIM: 607935

KEGG: hsa:11240

STRING: 9606.ENSP00000364635

UniGene: Hs.33455

Protein Families
Protein arginine deiminase family
Subcellular Location
Cytoplasm.
Tissue Specificity
Detected in keratinocytes in epidermis (at protein level).

Q&A

What is PADI2 and what is its primary enzymatic function?

PADI2 (Protein Arginine Deiminase 2) is an enzyme that catalyzes citrullination, a post-translational modification that converts positively charged arginine residues to neutral citrulline. This process effectively neutralizes the positive charge of a guanidinium group by replacing it with a neutral urea . PADI2 belongs to the PAD family, which includes PADs 1-4 and 6, and is believed to be the ancestral homologue based on sequence similarity across mammalian species and genomic organization of the PAD2 gene . Though initially known primarily for its role in myelination, PADI2 has more recently been linked to other cellular processes including gene transcription and macrophage extracellular trap formation .

How does PADI2 differ from other PAD family members in terms of cellular localization and substrate specificity?

PADI2 and PADI4 exhibit distinct cellular localization patterns and substrate preferences:

  • Cellular localization: PADI2 has a predominantly cytoplasmic distribution, while PADI4 is mainly nuclear .

  • Substrate specificity: When activated during cell death pathways (such as perforin-mediated cytotoxicity), PADI2 demonstrates a strong preference for citrullinating substrates above 31kDa. In contrast, PADI4 induces a less prominent but more widespread pattern of substrate citrullination, targeting molecules across a broader range of molecular weights .

These differences in localization and substrate preference contribute to the distinct patterns of citrullination observed in cells expressing these enzymes, which may have important implications for their respective roles in disease pathogenesis.

What is the catalytic mechanism of PADI2?

PADI2 utilizes a substrate-assisted mechanism of catalysis that differs from other PAD isozymes. In this mechanism:

  • The positively charged substrate guanidinium group appears to depress the pKa of the active site cysteine (Cys647).

  • This depression of pKa facilitates thiol deprotonation, which is necessary for nucleophilic attack on the substrate.

  • Studies with 2-chloroacetamidine, a positively charged inactivator, demonstrate this predicted effect, showing a shift in pKa that supports a substrate-assisted mechanism rather than a reverse-protonation mechanism .

This contrasts with PAD4, which shows no such pKa shift with 2-chloroacetamidine. The requirement for substrate binding to facilitate thiol deprotonation may provide greater protection against nonspecific inactivation of PADI2 by reactive oxygen or nitrogen species, which could be particularly important in highly oxidative environments such as those generated by activated macrophages where PADI2 is highly expressed .

What are the recommended methods for cloning and expressing recombinant human PADI2?

For successful expression of recombinant human PADI2, researchers should consider the following methodological approach:

  • Gene cloning: The human PADI2 gene can be cloned into an expression vector such as pET16B using NdeI/XhoI restriction sites after PCR amplification .

  • Expression system: E. coli is commonly used for expression, with optimal conditions typically involving induction at moderate temperatures.

  • Purification strategy: A His-tag purification approach is often employed, utilizing the vector's N-terminal His-tag for metal affinity chromatography.

  • Quality control: Verification of enzyme activity using citrullination assays with standardized substrates such as benzoyl-arginine ethyl ester (BAEE) or specific protein substrates like histone H3.

  • Storage considerations: Purified enzyme should be stored with reducing agents to protect the active site cysteine, and calcium should be excluded during storage to prevent autoactivation.

This expression system allows for the production of functional PADI2 that can be used for enzymatic studies, inhibitor screening, and structural analyses .

How can researchers effectively measure PADI2 enzymatic activity in experimental settings?

Several complementary methods can be employed to measure PADI2 enzymatic activity:

  • Colorimetric assays: These detect citrulline formation using specific chemical reactions, such as the COLDER (Colorimetric Detection of Citrulline) assay, which measures the release of ammonia during citrullination.

  • Immunoblotting with anti-modified citrulline antibodies: The anti-modified citrulline-Senshu method involves chemical modification of citrulline residues followed by detection with specific antibodies. This approach provides a visual representation of citrullinated proteins across a range of molecular weights .

  • Mass spectrometry: For precise identification of citrullination sites, mass spectrometry can detect the mass shift of +0.984 Da that occurs when arginine is converted to citrulline.

  • Peptide/protein substrates: Using defined substrates such as histone H3 (especially targeting R26) allows for specific assessment of PADI2 activity in various experimental conditions .

  • Calcium dependence analysis: Since PADI2 is calcium-dependent, activity measurements at varying calcium concentrations can provide insights into enzyme regulation and activation thresholds.

When selecting a method, researchers should consider the specific research question, sensitivity requirements, and whether global citrullination or specific substrate targeting is being investigated .

What is the evidence linking PADI2 to autoimmune diseases like rheumatoid arthritis?

PADI2 plays a significant role in the pathogenesis of rheumatoid arthritis (RA) through several mechanisms:

  • Generation of autoantigens: During cell death induced by cytotoxic pathways (such as perforin-mediated damage or complement attack), PADI2 activation leads to hypercitrullination of cellular proteins. In experimental models using PAD-expressing cells and cytotoxic assays, PADI2 generates a distinct pattern of citrullinated proteins that are recognized by RA autoantibodies .

  • Pattern specificity: PADI2 preferentially citrullinates substrates above 31kDa, creating a unique signature of modified proteins. Importantly, sera from RA patients recognize specific autoantigen patterns generated by PADI2 that are distinct from those generated by PADI4, suggesting these enzymes may have different contributions to disease pathogenesis .

  • Synovial fluid findings: Cells from the synovial fluid of RA patients exhibit hypercitrullination patterns that can be reproduced in experimental models of membranolytic cell death involving PADI2 activation .

  • Inhibitor effects: The PADI inhibitor Cl-amidine shows efficacy in various disease models, suggesting therapeutic potential in targeting PADI2 activity .

These findings indicate that PADI2-mediated citrullination generates specific autoantigens recognized by RA autoantibodies, establishing a mechanistic link between this enzyme and autoimmune pathology .

How does PADI2 contribute to breast cancer progression and what are the potential therapeutic implications?

PADI2 appears to play an important role in breast cancer progression through several mechanisms:

  • Expression correlation: PADI2 mRNA expression is highly correlated with HER2/ERBB2 (p = 2.2 × 10^6) in luminal breast cancer cell lines, suggesting a potential relationship with this oncogenic pathway .

  • Expression changes during cancer progression: Using the MCF10AT model of breast cancer progression, PADI2 expression increases during the transition from normal mammary epithelium to fully malignant breast carcinomas. A strong peak of PADI2 expression and activity is observed in the MCF10DCIS cell line, which models human comedo-DCIS lesions .

  • Epigenetic regulation: PADI2 is recruited to estrogen receptor α (ERα) promoters where it citrullinates histone H3 at R26. This modification triggers localized chromatin decondensation, facilitating ERα binding to its promoters and driving the transcription of more than 200 genes under estrogen receptor control .

  • Therapeutic targeting: The PADI inhibitor Cl-amidine strongly suppresses the growth of MCF10DCIS monolayers and tumor spheroids in culture. Preclinical studies in nude mice demonstrated that Cl-amidine also suppressed the growth of xenografted MCF10DCIS tumors by more than 3-fold .

  • Cell cycle effects: Cell cycle array analysis of Cl-amidine treated MCF10DCIS cells revealed that PADI inhibition strongly affects the expression of several cell cycle genes implicated in tumor progression, including p21, GADD45α, and Ki67 .

These findings suggest that PADI2 could serve as an important biomarker for HER2/ERBB2+ tumors and that PADI2 inhibitors represent promising candidates for breast cancer therapy, particularly for subtypes with high PADI2 expression .

How is PADI2 activity regulated at the molecular level?

PADI2 activity is regulated through several molecular mechanisms:

  • Calcium dependence: PADI2 is a calcium-dependent enzyme. Crystal structure studies of the related PAD4 have revealed five calcium-binding sites, none of which adopt an EF-hand motif. Calcium binding induces conformational changes that generate the active site cleft necessary for substrate binding and catalysis .

  • Substrate-assisted catalysis: Unlike other PAD isozymes, PADI2 employs a substrate-assisted mechanism where the positively charged substrate guanidinium group depresses the pKa of the active site cysteine (Cys647). This mechanism may provide greater protection against nonspecific inactivation by reactive oxygen or nitrogen species in oxidative environments such as activated macrophages .

  • Subcellular localization: PADI2's predominantly cytoplasmic distribution restricts its access to certain substrates, particularly nuclear proteins, unless cellular compartmentalization is disrupted during processes like cell death .

  • Cell death pathways: PADI2 becomes hyperactivated during specific forms of cell death, particularly those involving membranolytic pathways such as perforin-mediated cytotoxicity and complement membrane attack complex formation. This activation leads to widespread protein citrullination .

  • Enzyme recruitment: In certain contexts, such as gene regulation, PADI2 can be recruited to specific chromatin locations (e.g., ERα promoters) to perform targeted citrullination of histones .

Understanding these regulatory mechanisms is crucial for developing targeted approaches to modulate PADI2 activity in research and potential therapeutic applications .

What structural features distinguish PADI2 from other PAD family members and how do these differences affect inhibitor design?

The structural features that distinguish PADI2 from other PAD family members have significant implications for inhibitor design:

These structural and mechanistic differences provide the foundation for rational design of PADI2-selective inhibitors, which are critical tools for elucidating the specific biological roles of this isozyme and may ultimately be useful for treating diseases in which PADI2 activity is dysregulated .

How can researchers distinguish between citrullination mediated by PADI2 versus other PAD isozymes in complex biological samples?

Distinguishing between citrullination mediated by PADI2 versus other PAD isozymes in complex biological samples requires a multi-faceted approach:

  • Isozyme-specific substrate patterns: PADI2 shows a strong preference for citrullinating substrates above 31kDa, while PADI4 induces a more widespread pattern of citrullination across a broader range of molecular weights. Analyzing the molecular weight distribution of citrullinated proteins can provide initial insights into the PAD isozyme responsible .

  • Subcellular fractionation: Since PADI2 is predominantly cytoplasmic while PADI4 is mainly nuclear, subcellular fractionation followed by analysis of citrullinated proteins in each fraction can help attribute modifications to specific isozymes .

  • Isozyme-selective inhibitors: The development of PADI2-selective inhibitors, such as the benzimidazole-based derivatives of Cl-amidine, allows researchers to specifically block PADI2 activity while leaving other PAD isozymes functional. Comparing citrullination patterns in the presence and absence of these selective inhibitors can help identify PADI2-specific substrates .

  • Genetic approaches: Using siRNA knockdown, CRISPR-Cas9 gene editing, or cells from knockout animals to specifically deplete individual PAD isozymes provides another method to attribute citrullination events to specific enzymes.

  • Recombinant enzyme validation: After identifying potential PADI2-specific substrates in complex samples, validation using recombinant PADI2 and PADI4 with purified substrate proteins can confirm isozyme specificity.

  • Mass spectrometry analysis: Advanced mass spectrometry approaches can identify specific citrullination sites on proteins and determine whether these sites are preferentially modified by PADI2 or other PAD isozymes .

These complementary approaches allow researchers to attribute specific citrullination events to PADI2 versus other PAD isozymes, providing insights into their distinct roles in both normal biology and disease processes .

What are the challenges and solutions in developing selective PADI2 inhibitors for research and potential therapeutic applications?

Developing selective PADI2 inhibitors presents several challenges but also promising solutions:

Challenges:

  • Structural similarity: The high degree of sequence and structural similarity between PAD family members makes it difficult to achieve isozyme selectivity.

  • Calcium dependence: All PAD enzymes are calcium-dependent, which complicates the development of activation-specific inhibitors.

  • Bioavailability: Many PAD inhibitors are peptide-based or contain reactive warheads, which can limit their cellular uptake and in vivo stability.

  • Target validation: Confirming that observed biological effects result specifically from PADI2 inhibition rather than off-target effects requires extensive controls.

Solutions and Approaches:

  • Structural modifications of existing scaffolds: Modifications at both the N-terminus and C-terminus of the Cl-amidine scaffold have successfully yielded benzimidazole-based derivatives with >100-fold increases in PADI2 potency and selectivity .

  • Mechanism-based design: Exploiting the substrate-assisted mechanism unique to PADI2 provides an avenue for developing isozyme-selective inhibitors. Compounds that specifically interact with this mechanism may preferentially inhibit PADI2 .

  • Structure-activity relationship studies: Systematic exploration of chemical modifications can identify key features that enhance PADI2 selectivity while reducing affinity for other PAD isozymes.

  • Cellular efficacy optimization: Designing inhibitors with enhanced membrane permeability improves their utility for cellular and in vivo studies.

  • Target engagement assays: Developing methods to confirm PADI2 inhibition in cellular and in vivo contexts ensures that observed effects result from on-target activity.

These approaches have already yielded promising PADI2-selective inhibitors that will be critical for elucidating the biological roles of this isozyme and may ultimately prove useful for treating diseases in which PADI2 activity is dysregulated, including multiple sclerosis, rheumatoid arthritis, and breast cancer .

What are common pitfalls in PADI2 activity assays and how can researchers overcome them?

Researchers working with PADI2 activity assays should be aware of several common pitfalls and their solutions:

  • Calcium concentration variability:

    • Pitfall: Inconsistent calcium concentrations can lead to variable PADI2 activation.

    • Solution: Standardize calcium concentrations in assay buffers and include calcium calibration curves to understand the activation threshold for your specific experimental system.

  • Oxidation of the active site cysteine:

    • Pitfall: The active site cysteine (Cys647) in PADI2 is susceptible to oxidation, which can inactivate the enzyme.

    • Solution: Include reducing agents in storage and assay buffers, minimize freeze-thaw cycles, and consider performing experiments under low-oxygen conditions when possible.

  • Substrate specificity interference:

    • Pitfall: When using complex protein mixtures as substrates, differential accessibility of arginine residues can complicate interpretation.

    • Solution: Use defined peptide substrates in parallel with complex protein substrates to establish baseline enzyme activity.

  • Detection method limitations:

    • Pitfall: Anti-modified citrulline antibodies may have different affinities for citrullinated epitopes depending on surrounding sequence context.

    • Solution: Combine multiple detection methods (e.g., antibody-based detection, colorimetric assays, and mass spectrometry) to comprehensively assess citrullination.

  • Background citrullination:

    • Pitfall: Endogenous PADs in biological samples can create background citrullination that confounds interpretation.

    • Solution: Include appropriate controls such as calcium chelators (EGTA) or PAD inhibitors, and consider using PAD-knockout cells or tissues when available .

  • Distinguishing between PAD isozymes:

    • Pitfall: In complex biological samples, citrullination could be mediated by multiple PAD isozymes.

    • Solution: Use isozyme-selective inhibitors, subcellular fractionation, or recombinant enzyme validation to attribute citrullination to specific PAD isozymes .

By anticipating these challenges and implementing appropriate controls and experimental design, researchers can obtain more reliable and interpretable results from PADI2 activity assays.

How can researchers effectively validate PADI2 as a therapeutic target in disease models?

Comprehensive validation of PADI2 as a therapeutic target requires a multi-faceted approach:

  • Genetic validation:

    • Employ CRISPR-Cas9 gene editing or RNA interference to deplete PADI2 in disease-relevant cell models

    • Generate conditional or tissue-specific PADI2 knockout animal models

    • Assess whether genetic depletion recapitulates the effects of pharmacological inhibition

  • Pharmacological validation:

    • Utilize PADI2-selective inhibitors with appropriate controls for selectivity

    • Employ dose-response studies to establish correlation between inhibition level and therapeutic effect

    • Compare effects of PADI2-selective inhibitors with pan-PAD inhibitors like Cl-amidine

  • Biomarker development:

    • Identify disease-specific substrates of PADI2

    • Develop assays to monitor citrullination of these substrates in biological samples

    • Correlate changes in biomarker levels with disease progression and therapeutic response

  • Mechanism elucidation:

    • For breast cancer: Monitor effects on estrogen receptor signaling and histone H3R26 citrullination

    • For rheumatoid arthritis: Assess impact on autoantigen generation and autoantibody reactivity

    • For multiple sclerosis: Evaluate effects on myelination and inflammatory processes

  • Efficacy demonstration:

    • In cellular models: Assess impact on disease-relevant phenotypes (e.g., proliferation for cancer models)

    • In animal models: Evaluate disease progression using established metrics (e.g., tumor growth, arthritis score)

    • Document dose-dependent effects that correlate with PADI2 inhibition levels

  • Safety assessment:

    • Evaluate potential on-target toxicity in tissues where PADI2 plays physiological roles

    • Monitor for off-target effects on other PAD isozymes or unrelated proteins

    • Assess impact on normal cellular functions where PADI2 activity may be required

  • Combination approaches:

    • Test PADI2 inhibitors in combination with standard-of-care therapies

    • Evaluate potential synergistic effects or mechanisms of resistance

Evidence supporting PADI2 as a therapeutic target is particularly strong in breast cancer, where the PADI inhibitor Cl-amidine suppresses the growth of MCF10DCIS monolayers and tumor spheroids in culture, and reduces xenografted tumor growth by more than 3-fold in preclinical mouse studies . Similar validation approaches can be applied to other disease contexts where PADI2 activity is implicated .

What are the emerging roles of PADI2 in cellular processes beyond established functions?

Recent research has uncovered several emerging roles for PADI2 that extend beyond its established functions:

  • Epigenetic regulation: PADI2 plays a significant role in gene regulation by citrullinating histone H3 at R26. This modification triggers localized chromatin decondensation, facilitating transcription factor binding. In breast cancer cells, PADI2-mediated histone citrullination leads to increased transcription of more than 200 genes under estrogen receptor control, suggesting a broader role in epigenetic programming .

  • Macrophage extracellular trap formation: Similar to the neutrophil extracellular traps (NETs) formed by neutrophils through PAD4 activity, PADI2 appears to be involved in the formation of macrophage extracellular traps (METs). This process may contribute to innate immune responses but could also promote inflammation in pathological contexts .

  • Cell death pathway modulation: The activation of PADI2 during specific forms of cell death, particularly those involving membranolytic pathways, suggests a potential role in regulating cell death processes or in the clearance of dying cells .

  • Generation of disease-specific autoantigens: The ability of PADI2 to generate distinct patterns of citrullinated proteins that are recognized by autoantibodies in rheumatoid arthritis suggests a role in shaping the autoimmune response in this disease .

  • Cancer progression: Beyond its role in breast cancer through estrogen receptor signaling, the correlation of PADI2 expression with HER2/ERBB2 suggests potential involvement in additional oncogenic pathways. The increased expression of PADI2 during the transition from normal mammary epithelium to malignant breast carcinomas points to a role in cancer progression .

These emerging roles highlight the diverse functions of PADI2 in cellular processes and disease pathogenesis, suggesting that continued investigation will likely uncover additional biological roles for this enzyme .

How might advances in structural biology and computational approaches enhance our understanding of PADI2 function and facilitate drug discovery?

Advances in structural biology and computational approaches offer promising avenues for enhancing our understanding of PADI2 and accelerating drug discovery:

  • High-resolution structures: While crystal structures of related PAD4 have been determined , obtaining high-resolution structures of PADI2 in different conformational states (calcium-free, calcium-bound, and substrate-bound) would provide critical insights into its activation mechanism and substrate recognition. Techniques such as cryo-electron microscopy could capture dynamic states that are difficult to crystallize.

  • Molecular dynamics simulations: Computational modeling of PADI2 dynamics can reveal conformational changes associated with calcium binding and substrate recognition. These simulations can identify transient binding pockets and allosteric sites that might be targeted for selective inhibition.

  • Virtual screening and docking: Using the unique structural features of PADI2, virtual screening of compound libraries can identify novel chemical scaffolds with potential selectivity. Structure-based drug design approaches can optimize these scaffolds for improved potency and pharmacokinetic properties.

  • Quantum mechanics/molecular mechanics (QM/MM) approaches: Since PADI2 employs a substrate-assisted catalytic mechanism , QM/MM simulations can provide insights into the energetics of this process and identify transition states that could be targeted by transition-state analogue inhibitors.

  • Machine learning for selectivity prediction: By analyzing the structural differences between PAD isozymes and their interactions with known inhibitors, machine learning algorithms can identify features that confer selectivity and guide the design of PADI2-selective compounds.

  • Protein-protein interaction mapping: Identifying the interaction partners of PADI2 in different cellular contexts can reveal regulatory mechanisms and potential indirect targeting strategies.

  • Systems biology approaches: Integration of structural information with genomic, proteomic, and metabolomic data can provide a systems-level understanding of PADI2 function in health and disease.

These advanced approaches would complement the successful development of benzimidazole-based derivatives of Cl-amidine that have already achieved >100-fold increases in PADI2 potency and selectivity . By providing deeper insights into PADI2 structure and function, these methods could facilitate the development of next-generation PADI2-selective inhibitors with improved potency, selectivity, and drug-like properties.

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.