Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Role of the redox state of the Pirin-bound cofactor on interaction with the master regulators of inflammation and other pathways

  • Tamim Ahsan ,

    Contributed equally to this work with: Tamim Ahsan, Sabrina Samad Shoily

    Roles Conceptualization, Formal analysis, Methodology

    Affiliation Molecular Biotechnology Division, National Institute of Biotechnology, Savar, Dhaka, Bangladesh

  • Sabrina Samad Shoily ,

    Contributed equally to this work with: Tamim Ahsan, Sabrina Samad Shoily

    Roles Conceptualization, Formal analysis, Writing – original draft

    Affiliation Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh

  • Tasnim Ahmed,

    Roles Validation, Writing – review & editing

    Affiliation Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh

  • Abu Ashfaqur Sajib

    Roles Conceptualization, Supervision, Writing – review & editing

    abu.sajib@du.ac.bd

    Affiliation Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh

Abstract

Persistent cellular stress induced perpetuation and uncontrolled amplification of inflammatory response results in a shift from tissue repair toward collateral damage, significant alterations of tissue functions, and derangements of homeostasis which in turn can lead to a large number of acute and chronic pathological conditions, such as chronic heart failure, atherosclerosis, myocardial infarction, neurodegenerative diseases, diabetes, rheumatoid arthritis, and cancer. Keeping the vital role of balanced inflammation in maintaining tissue integrity in mind, the way to combating inflammatory diseases may be through identification and characterization of mediators of inflammation that can be targeted without hampering normal body function. Pirin (PIR) is a non-heme iron containing protein having two different conformations depending on the oxidation state of the iron. Through exploration of the Pirin interactome and using molecular docking approaches, we identified that the Fe2+-bound Pirin directly interacts with BCL3, NFKBIA, NFIX and SMAD9 with more resemblance to the native binding pose and higher affinity than the Fe3+-bound form. In addition, Pirin appears to have a function in the regulation of inflammation, the transition between the canonical and non-canonical NF-κB pathways, and the remodeling of the actin cytoskeleton. Moreover, Pirin signaling appears to have a critical role in tumor invasion and metastasis, as well as metabolic and neuro-pathological complications. There are regulatory variants in PIR that can influence expression of not only PIR but also other genes, including VEGFD and ACE2. Disparity exists between South Asian and European populations in the frequencies of variant alleles at some of these regulatory loci that may lead to differential occurrence of Pirin-mediated pathogenic conditions.

Introduction

The immune system responds to stress or harmful stimuli, such as toxic compounds or pathogens, by initiating a cascade of inflammation, and thus restores homeostasis [1, 2]. However, pathological fueling of certain pathways and their systemic effects through interconnected networks of biological pathways and processes plays a key role in the onset or progression of immune‐mediated inflammatory diseases and leads to the emergence of multimorbidity [2, 3]. Abnormal regulation of one or more of the three key inflammatory pathways- Nuclear factor kappa B (NF-κB), Mitogen-activated protein kinase (MAPK), and Janus kinase/signal transducer and activator of transcription (JAK-STAT), results in progression to inflammation-mediated diseases [1].

Apparently, inhibition of NF-κβ may provide effective treatment option for diseases associated with its overactivation. However, as NF-κB transcription factors regulate the expression of genes involved in several critical physiological processes, including cell survival, growth, proliferation, oxidative stress responses, inhibition of apoptosis, inflammation and immune responses, direct inhibition of NF-κB signaling can lead to immunodeficiency and dysregulation of the associated physiological activities [4, 5]. Thus, bypass routes of suppressing over-activation of the NF-κB pathway can be the choice of treatment for chronic inflammation-mediated diseases.

Pirin (encoded by the gene PIR) is a highly conserved 290 amino acid long and 32 kDa nuclear protein. Pirin was discovered as an interactor of nuclear factor I/CCAAT box transcription factor (NFI/CTF1) in a yeast two-hybrid (Y2H) screening [6]. On the basis of sequence and structural homology, this protein, which consists of two antiparallel β-barrel domains with an iron cofactor located within the N-terminal domain, belongs to the functionally diverse cupin superfamily [7, 8]. Depending on the oxidation state of the bound iron (Fe2+ and Fe3+), there are two different conformations of Pirin which vary in their R-shaped surface area [9]. Pirin is detected at low levels in all human tissues, but the transcript levels are the highest in the heart and liver [6, 10]. PIR orthologs have been found in prokaryotic organisms, fungi, plants and other mammals [8]. Both human and bacterial Pirins were demonstrated to be functionally similar to quercetin 2,3-dioxygenase, which can use quercetin flavonoid as a substrate. This function of Pirin can be inhibited by the addition of the inhibitors of quercetin 2,3-dioxygenase [11].

Pirin is one of the regulators of NF-κB signaling in the nucleus, and this regulation is controlled by its iron center [9, 12]. Under oxidizing conditions, the Fe(III) form of Pirin binds transcription factor NF-κB p65 and enhances the interaction between DNA and NF-κB p65 [9, 13]. Additionally, Pirin coregulates the NF-κB transcription pathway through interaction with the oncoprotein B-cell CLL/lymphoma 3 (BCL3) [8]. In spite of being a member of the Inhibitor of Kappa-B (IκB) family of proteins, which contribute to repression of the NF-κB signaling cascade, BCL3 exerts both transactivation and transrepressor activities in the regulation of NF-κB associated pathways [14]. Thus, characterization of the molecular association of Pirin with NF-κB and the consequences of this interaction may reveal new targets for inhibition of pathogenic inflammation.

Over-expression of PIR in breast cancer cells play roles in tumorigenesis and cancer progression through induction of the E2F1 pathway [15]. Pirin was also demonstrated to be associated with metastasis of melanoma and cervical cancer cells [16, 17]. Abnormal pattern of sub-cellular localization of Pirin was observed in a subset of melanomas [7]. The potential role of Pirin regulated pathways in cancer progression and/or metastasis may be elucidated by analysis of its interaction with other proteins and associated biological processes. Variants in the PIR gene have been linked to interethnic disparities in COVID-19 case fatality rates [18].

Although established as a regulator of inflammation [9], there are barely any studies solely focusing on the pathways of Pirin and its role in disease pathogenesis. The existing databases lack information regarding the regulatory and functional roles of Pirin. Understanding how a protein interacts with other proteins in a protein-protein interaction (PPI) network is important for figuring out its overall role and how it controls other proteins [19, 20]. In this study, we have utilized protein-protein interactions and network-based methods to functionally characterize Pirin. We also explored the potentially pathogenic variants that may modulate PIR expression and their frequencies in populations worldwide.

Materials and methods

Determination of interaction partners of Pirin

Protein-protein interaction (PPI) databases- BioGRID (v4.4.201) [21], IMEx [22], IntAct (v1.0.2) [23], MINT [24], STRING (v11.5) [25] and Mentha [26] were searched with the term “PIR” to retrieve interacting partners of Pirin as well as evidence of the interactions. The Biological General Repository for Interaction Datasets (BioGRID) is an open access biomedical repository that houses comprehensively curated protein, genetic, and chemical interactions as well as post-translational modifications for humans and all major model organism species [21]. The IntAct molecular interaction database populates data derived from literature curation or direct user deposition, and it actively contributes to the International Molecular Exchange Consortium (IMEx) partners via a sophisticated web-based curation platform [23, 27]. Mentha is a PPI resource that brings together protein interactions from primary databases that are in compliance with IMEx curation policies [26]. The Molecular INTeraction database (MINT) is a public PPI database that stores molecular interactions that have been reported in peer-reviewed journals [24]. The Search Tool for Retrieval of Interacting Genes/Proteins (STRING) database integrates known as well as predicted interactions, including both physical (direct) and functional (indirect) associations [25, 28].

Analysis of binding between Pirin and its direct interactors

In this study, Y2H system was considered as the experimental evidence of direct PPI, as this genetic system enables the detection of direct interaction between proteins [29, 30]. Based on this criterion BCL3, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (NFKBIA, also known as IKBA), nuclear factor I/X (NFIX), and SMAD family member 9 (SMAD9) were identified as the direct interactors of Pirin (Table 1). The domain architectures of the direct interactors were obtained from either the UniProt Knowledgebase (UniProt-KB) or Pfam 35.0 database (depending on availability) [31, 32]. X-ray crystallographic structures of BCL3 (PDB ID: 1K1A), NFKBIA (PDB ID: 1IKN), and SMAD9 (PDB ID: 6FZT) were retrieved from the Protein Data Bank (PDB) [33]. Since crystallographic structure of NFIX was not available, its predicted structure was downloaded from AlphaFold2 [34]. Although X-ray crystallographic structures of both the Fe2+ (PDB code: 1J1L) and Fe3+ bound (PDB code: 4GUL) conformations of Pirin are available [35], the crystal structure of the Fe2+ bound Pirin contained six modified methionine residues. To maintain uniformity, structures of both the conformations were modeled using template-based modeling at SWISS-MODEL server [36]. Amino acid sequence of Pirin (UniProt accession: O00625-1) was collected from UniProt [31].

To get insight into the association of direct interaction partners with Pirin, the putative interacting domains of the direct interactors were docked to modeled structures of both ferrous (Fe2+) and ferric (Fe3+) conformations of Pirin protein using the HawkDock web server [37]. To calculate the binding free energies, Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) analysis was performed on the HawkDock server [37, 38].

The top 10 docked models were re-ranked using the DOCKSCORE web server [39]. In DOCKSCORE, weights are assigned to multiple parameters of the putative interface between the two protein partners, including surface area, short contacts, evolutionary conservation, spatial clustering as well as the presence of positively charged and hydrophobic residues. By considering these weights the normalized weighted score is calculated, using which a Z-score for each pose is provided to facilitate identification of native or near native pose from protein-protein docked poses [39]. The best models were chosen based on their pose, binding free energy from MM/GBSA analysis and Z-scores from DOCKSCORE. For these models, 2D interaction map was constructed using the structural analysis tool iCn3D [40]. Hotspot residues that contribute the most to binding between the proteins in these docked structures were identified using SpotOn [41]. For SMAD9 and NFIX, selected poses were superimposed on DNA-bound conformation of MH1 domains to ensure that Pirin was not interacting with DNA-binding residues. PIR-NFKBIA docked structure was superimposed on the NFKBIA-NF-κB complex (PDB ID: 1IKN) to confirm that Pirin binding site did not considerably overlap with binding sites of the other proteins in NFKBIA.

Construction and visualization of PPI network

Using PIR and its identified interactors as input, network analysis was performed with NetworkAnalyst 3.0 [42]. The network was constructed based on the PPI data in the IMEx database (using the default parameters and minimum network). The network was visualized with the Cytoscape (v3.8.0) software [43].

Enrichment analysis of Pirin and its interaction partners

Pathway enrichment.

Pathway enrichment analysis facilitates gaining mechanistic insight and makes interpretations easier by summarizing a large list of genes as a smaller list of pathways that are enriched in the determined gene list more than would be expected by chance [44]. Pathway enrichment was performed on all nodes of the minimum network through NetworkAnalyst 3.0 using each of the Kyoto Encyclopedia of Genes and Genomes (KEGG) [45] and Reactome [46] databases as the source for annotated pathways [42].

Integrated functional network formation.

To attain an integrated view of the pathways and processes associated with Pirin signaling, a network with the KEGG and Reactome pathways as well as Gene Ontology (GO): Biological process (BP) was formed using ClueGO v2.5.8 and CluePedia v1.5.8 plug-in of Cytoscape [47, 48]. For enrichment, a two-sided hypergeometric test with Benjamini-Hochberg correction was performed, GO terms were merged, and a kappa score of 0.5 was used as the threshold value. ClueGo facilitates formation of functionally organized network of pathway terms and biological processes enriched from precompiled annotation files for a given set of genes [47]. CluePedia allows visualization of functionally connected genes and their shared pathways in a ClueGo constructed network [48].

Disease enrichment.

Diseases associated with Pirin as well as its interactors were enriched using Enrichr [49, 50] with DisGeNET as the source database. Enrichr is a comprehensive enrichment and functional annotation web server. DisGeNET (v7.0) is a knowledge management platform that integrates and standardizes data regarding association of diseases with genes and variants from various sources [51]. The gene-disease associations (GDA) network was based on expert-curated data. Additionally, the DisGeNET (v7.0) [51] app at the Cytoscape [43] was used for visualizing associations of diseases with the direct interactors of Pirin as well as the RELA gene (encodes NF-κB p65) as binding of p65 TF to DNA is modulated by Pirin [9].

Retrieval of regulatory variants.

PIR variants with potential regulatory roles were identified from rVarBase (no filters applied) [52]. Ensembl Variant Effect Predictor (VEP) [53] was used to collect the features (chromosomal location, consequences with respect to PIR and impact) of these variants. Among these, expression quantitative trait loci (eQTLs) or the variants that can influence the expression of genes in at least one tissue were identified through Genotype-Tissue Expression (GTEx) portal v8 [54]. Frequencies of the eQTLs in five super-populations (African, admixed American, East Asian, European and South Asian) were determined from the Phase 3 haplotype data from the 1000 Genomes Project [55] using the LDhap tool at the open source LDlink suite [56].

Result

Interactors of Pirin protein

Through exploration of six PPI databases, 69 unique direct and indirect interaction partners of Pirin were identified (Table 1). The direct interactors of Pirin (shown in bold font in Table 1) identified by Y2H system are BCL3, NFKBIA, NFIX, and SMAD9.

Docked structures of Pirin and its direct partners

The models chosen based on pose, binding free energy, and Z-scores of the docked structures of Pirin and its direct interaction partners as well as the residues that significantly contribute to the interactions are shown in Figs 14. The binding free energy and Z-scores, respectively, from MM/GBSA and DOCKSCORE analysis of the selected models, are given in Table 2. All the domains of these interactors are given in S1 Table. All seven ankyrin repeat domains of BCL3 are required for binary interaction between Pirin and BCL3 [8]. Therefore, ankyrin repeats of BCL3 and NFKBIA were used for docking with Pirin. Such domains are not present in NFIX and SMAD9, but the Mad homology 1 (MH1) domain is commonly present in both these proteins (S1 Table). As stable interactions between proteins are mediated by domains, this common domain was selected for docking of NFIX and SMAD9 to Pirin. Fe2+ bound conformation of Pirin appeared to be the conformation that may bind to the direct interacting partners.

thumbnail
Fig 1. Interactions between Pirin and BCL3.

(a) Ankyrin repeats of BCL3 (blue) docked to Pirin (hotpink) with interface shown in yellow; (b) Hotspot (Pirin residues = yellow, partner residues = orange) within the interface; and (c) 2D interaction map where the vertical and horizontal axis represent the Pirin and BCL3 residues, respectively.

https://doi.org/10.1371/journal.pone.0289158.g001

thumbnail
Fig 2. Interactions between Pirin and NFKBIA.

(a) Ankyrin repeats of NFKBIA (blue) docked to Pirin (hotpink) with interface shown in yellow; (b) Hotspot (Pirin residues = yellow, partner residues = orange) within the interface; and (c) 2D interaction map where the vertical and horizontal axis represent the Pirin and NFKBIA residues, respectively.

https://doi.org/10.1371/journal.pone.0289158.g002

thumbnail
Fig 3. Interactions between Pirin and NFIX.

(a) MH1 domain of NFIX (blue) docked to Pirin (hotpink) with interface shown in yellow; (b) Hotspot (Pirin residues = yellow, partner residues = orange) within the interface; and (c) 2D interaction map where the vertical and horizontal axis represent the Pirin and NFIX residues, respectively.

https://doi.org/10.1371/journal.pone.0289158.g003

thumbnail
Fig 4. Interactions between Pirin and SMAD9.

(a) MH1 domain of SMAD9 (blue) docked to Pirin with interface shown in yellow; (b) Hotspot (Pirin residues = yellow, partner residues = orange) within the interface; and (c) 2D interaction map where the vertical and horizontal axis represent the Pirin and SMAD9 residues, respectively.

https://doi.org/10.1371/journal.pone.0289158.g004

thumbnail
Table 2. Binding free energies and z-scores of the selected models.

https://doi.org/10.1371/journal.pone.0289158.t002

Integrated view of Pirin and its interaction partners through network building

As the first-order interaction network created by NetworkAnalyst 3.0 was very dense (with more than 2000 nodes) due to a large number of query genes (seeds), it was trimmed to a minimum network that retained only the seeds and their connecting nodes for improving performance and ease of further analysis [42, 57]. This minimum interaction network, representing comprehensive Pirin signaling, contains 141 nodes and the first degree interactors of Pirin are highlighted in pink (Fig 5). Along with BCL3, NFIX, NFKBIA and SMAD9, the other first degree interaction partners of Pirin in the network are Signal transducing adaptor molecule (STAM), Proteasome 20S subunit beta 2 (PSMB2), Chloride nucleotide-sensitive channel 1A (CLNS1A), Histone deacetylase 2 (HDAC2), Protein phosphatase 2 catalytic subunit alpha (PPP2CA), SGT1 homolog MIS12 kinetochore complex assembly cochaperone (SUGT1), Spen family transcriptional repressor (SPEN), Ubiquitin-like 7 (UBL7), and Rho GDP dissociation inhibitor alpha (ARHGDIA). All of these, except HDAC2, were query genes given as input.

thumbnail
Fig 5. PPI network among Pirin and its interaction partners (represented by gene symbols).

First degree interaction partners of Pirin (undirected) are highlighted in pink.

https://doi.org/10.1371/journal.pone.0289158.g005

Functional analysis of Pirin and its interaction partners

Enriched pathways and GO terms.

The statistically significant biological process or pathway terms associated with Pirin and its 69 direct and indirect interaction partners (Table 3) obtained through ClueGO analysis are shown in Fig 6. Among the pathways enriched for all 141 genes (nodes) of the minimum network formed using Pirin and its interaction partners as input, top 20 based on the KEGG and Reactome databases are given in Fig 7.

thumbnail
Fig 6. ClueGO generated functional network of Pirin.

GO:BP terms and pathway terms are represented by triangles and ellipses, respectively. Interactors of Pirin are linked to enriched biological processes and KEGG and REACTOME pathways. Overrepresented functional categories are shown in the pie chart where the asterisks indicate significance of the terms (** p- value < 0.001).

https://doi.org/10.1371/journal.pone.0289158.g006

thumbnail
Fig 7. Scatter plots for top 20 enriched pathways based on KEGG (A) and Reactome (B) pathway databases.

The x-axis represents the rich factor of each pathway and y-axis shows the pathways. The Rich factor refers to the ratio of number of target genes (nodes of the minimum network) annotated in a pathway term to all gene numbers annotated in that pathway term. The larger the Rich factor, the higher the degree of pathway enrichment. The size and color of the dots represent the number of candidate genes mapped to the indicated pathway and q value, respectively. A lower q value, that is the corrected p-value ranging from 0 ~ 1, indicates greater pathway enrichment.

https://doi.org/10.1371/journal.pone.0289158.g007

The pathway involving activation of Wiskott-Aldrich syndrome protein (WASP) and WASP-family verprolin-homologous protein (WAVE) by the Rho family of GTPases (RHO GTPase) is most significantly associated with Pirin and its interaction partners (Fig 6). The correlation of Pirin signaling with actin filament fragmentation, regulation of actin dynamics for phagocytic cup formation, signaling by the B cell receptor (BCR), activation of NF-κB in B cells, gene and protein expression by JAK-STAT signaling after interleukin-12 (IL12) stimulation, nucleotide sugar biosynthetic process, metabolism of tyrosine and pyruvate, signaling by PTK6 and FGFR2, regulation of intrinsic apoptotic signaling pathway, regulation of DNA binding, and negative regulation of transposition is significantly highlighted by the ClueGo integrative analysis.

In KEGG pathway enrichment (Fig 7a) for the extended Pirin interactome (Pirin, its interaction partners and further nodes required to connect the seed nodes in minimum network (Fig 5)), the “ErbB signaling pathway” is the most significantly over-represented (q value < 0.005). ErbB signaling pathway influences cell proliferation, survival, differentiation, and migration (KEGG pathway entry: hsa04012). ErbB family of receptor tyrosine kinases (RTKs) comprises four distinct receptors: the epidermal growth factor receptor (EGFR/ErbB1/HER1), ErbB2 (neu, HER2), ErbB3 (HER3) and ErbB4 (HER4) [58]. Alteration of the pathways regulated by these receptors play key role in development of multiplicity of cancers as well as invasion of tumor cells [58, 59]. The highest number of genes (n = 18) are mapped to “pathways in cancer” in the KEGG database (Fig 7a). Node genes were also clustered in other carcinogenic pathways, including chronic myeloid leukemia (CML), renal cell carcinoma, viral carcinogenesis, bladder cancer, and prostate cancer. Endocrine resistance is also enriched which is a major clinical issue encountered in treating metastatic hormone receptor-positive breast cancer with endocrine therapy [60]. Pathways associated with microbial infection, which are Shigellosis or intestinal Shigella infection [61], bacterial invasion of epithelial cells, pathogenic Escherichia coli infection, viral myocarditis and prion diseases, were enriched as well.

Fc gamma receptor (FCGR) mediated phagocytosis is enriched in both KEGG and Reactome analyses (Fig 7). Terms related to platelet activation and aggregation are over-represented in the Reactome database enrichment (Fig 7b). These include “Integrin alpha IIb/beta 3 signaling” [62], “Platelet activation, signaling and aggregation”, “Platelet Aggregation (Plug Formation)”. Furthermore, involvement of the Pirin signaling in immune system pathways is also highlighted via enrichment of “Chemokine signaling pathway”, “Immune system”, “Innate immune system”, “CD28 dependent Vav1 pathway” and “CD28 co-stimulation” [63].

Enriched diseases.

Enrichment analysis was performed to identify the contribution or possible association of Pirin and its interaction partners with diseases. Thirty nine of the enriched diseases with adjusted p-value <0.05 were considered as significant and are shown in Fig 8a. To further narrow down the exploration, a GDA network for Pirin, BCL3, NFIX, NFKBIA, SMAD9 and RELA was obtained from Cytoscape based on curated data in DisGeNET (Fig 8b).

thumbnail
Fig 8. Diseases significantly enriched for Pirin and its interaction partners.

(a) Diseases over-represented for minimum network by DisGeNET at EnrichR reflecting all interactor connected signaling of Pirin. The x-axis shows the significantly enriched disease terms and the y-axis represents the negative log (base 10) of the adjusted p-value. The number of genes (n) assigned to the disease term are shown with each bar. (b) GDA network with score 0 ~ 1 for Pirin and proteins that directly interact with it. Nodes are colored according to disease classes which are indicated on the right side.

https://doi.org/10.1371/journal.pone.0289158.g008

Liver carcinoma is the most significantly enriched disease for the Pirin interactome (Fig 8a). Both breast carcinoma and malignant neoplasm of breast are enriched for 30 of the query genes. Other over-represented malignancies for the query genes include malignant neoplasm of mouth, malignant neoplasm of prostate, lip and oral cavity carcinoma, secondary malignant neoplasm of lymph node, and prostate carcinoma. Multiple neoplasms (benign or malignant tumor) are correlated to RELA, NFKBIA and SMAD9 (Fig 8b).

The second most significantly enriched disease is Alzheimer’s disease (AD) (Fig 8a). Neuroinflammation contributes to onset and progression of this neurodegenerative disorder [64, 65]. Along with Alzheimer’s disease, multiple other degenerative brain disorders are correlated to at least 3 of the query genes. The broader umbrella term motor neuron disease (MND) or atrophy as well as a number of neurodegenerative conditions described by the terms, including spinal muscular atrophy and the Guam form of amyotrophic lateral sclerosis (ALS), are enriched as well [6668]. Several forms of Parkinsonian disorder or parkinsonism are also enriched significantly. Such neurodegenerative disorders are characterized by rigidity, bradykinesia, resting tremor, and postural instability [6971] and the major hallmark of Parkinson’s disease (PD) is chronic inflammation [72].

A transient ischemic attack (TIA), that frequently precedes ischemic stroke, is an episode of neurological dysfunction resulting from inadequate blood flow to focal brain, spinal cord, or retina, without acute infarction [73, 74]. Multiple forms of transient neurological dysfunctions are highly clustered for the Pirin interactome (Fig 8a). Furthermore, Ramsay Hunt Paralysis Syndrome, a neurological disorder caused by varicella-zoster virus reactivation and replication at the facial nerve, correlates to interaction partners of Pirin. These enrichments indicate towards involvement of Pirin signaling in neuroinflammation and pathological neuro-alterations.

Not many diseases were connected to Pirin directly (Fig 8b). One of the reasons may be a lack of studies on this protein. Skin and connective tissue diseases are connected to Pirin, RELA and NFKBIA. RELA is also associated with endocrine system and metabolic diseases, including non-insulin dependent diabetes mellitus, diabetic angiopathies and diabetic nephropathy as well as digestive system diseases, such as biliary cirrhosis, hepatomegaly, hepatic insufficiency, experimental and alcoholic liver cirrhosis, ulcerative colitis and cholestasis. SMAD9 is correlated to various respiratory tract diseases and different types of pulmonary hypertension.

Regulatory variants of PIR gene

Using the rVarBase [52] database, 78 variants potentially modifying PIR gene expression were identified (Table 3). Among these, five, twenty five and forty eight are located in the 5’ UTR, upstream and intron regions of PIR gene, respectively. Genetic variants residing in noncoding regions can be significantly associated with expression of corresponding genes [75]. The target genes for majority of the intron variants are PIR and BMX non-receptor tyrosine kinase (BMX).

rs180708754, a 3’ UTR variant of PIR gene, is located within the target site of a miRNA (Table 3). According to mirSNP database [76], this SNP can break the binding of hsa-miR-3673 and hsa-miR-4694-3p whereas it enhances the binding of hsa-miR-5680 (data not shown). TargetScan (Release 7.1) [77] predicted PIR to be one of the target genes for both miR-4694-3p and hsa-miR-5680.

The twenty five upstream variants of PIR gene are overlapped by both Transcription factor (TF) binding region and chromatin interactive region (Table 3). Variants of TF binding region may affect gene expression by altering binding of TFs, which are molecular switches regulating the amount and timing of gene transcription via sequence-specific binding [75]. As distal regulatory elements interact with their target genes through long-range chromatin loop bridges [78, 79], variants within chromatin interactive regions can plausibly influence gene expression through deviating from the natural interaction. The potential target genes for the upstream regulatory variants of PIR are PIR itself, ACE2, BMX, and Carbonic Anhydrase 5B Pseudogene 1 (CA5BP1).

Nine variants (rs565388697, rs539142342, rs369918088, rs190912217, rs187777697, rs1567894, rs372563960, rs183910461, and rs148530113) reside on a CpG island (Table 3). Discrete CpG dinucleotide motifs are the sites for DNA methylation- a type of epigenetic modification, and genetic variants located within or close to high density CpG sites can be mediators of gene expression [80, 81].

eQTLs in PIR and their geographic distribution

The GTEx project provides a comprehensive picture of expression quantitative trait loci (eQTL) that can alter gene expression profiles in a number of tissues by assessing the effects of genetic variations on transcriptomes across > 50 tissues of 838 individuals [54]. Among the identified regulatory PIR gene variants, nine (rs4830964, rs2094, rs2095, rs908005, rs1567894, rs2271550, rs5980162, rs6629104 and rs6629105) could be identified through GTEx portal as modulators of expression of PIR and Vascular Endothelial Growth Factor D (VEGFD) genes across multiple tissues (Table 4). rs6629104 and rs6629105 can affect expression of both ACE2 and PIR in nucleus accumbens of basal ganglia within the brain (S2 Table). rs6629105 T allele increases expression of ACE2 in nucleus accumbens (S1 Fig). rs4830964, rs2094, rs1567894, and rs6629104 regulates expression of carbonic anhydrase 5B pseudogene 1 (CA5BP1) in the lung. Variant allele frequencies (VAFs) at these loci in five super-populations (African, Admixed American, East Asian, European and South Asian) are given in Table 3. The frequencies of variant alleles at rs4830964, rs5980162, rs2095, rs2094 and rs1567894 are relatively low in South Asian populations compared to the other populations. The variant allele at rs2271550 is present with > 0.3 frequency only in the European population.

thumbnail
Table 4. GTEx validated eQTLs and their frequencies in five super-populations.

https://doi.org/10.1371/journal.pone.0289158.t004

Discussion

Persistent cellular stress induced perpetuation and uncontrolled amplification of inflammatory response results in a shift from tissue repair toward collateral damage, significant alterations of tissue functions, and derangements of homeostasis which in turn can lead to a large number of acute and chronic pathological conditions, such as chronic heart failure, atherosclerosis, myocardial infarction, neurodegenerative diseases, diabetes, rheumatoid arthritis, and cancer [82]. Keeping the vital role of balanced inflammation in maintaining tissue integrity in mind, the way to combating inflammatory diseases may be through identification and characterization of mediators of inflammation that can be targeted without hampering normal body function. In our study, we have demonstrated Pirin protein to have role in multiple important processes including inflammation, rearrangement of actin cytoskeleton as well as metastasis, ischemic attack and neurodegenerative diseases among others. We have analyzed regulatory variants of Pirin gene to have insight into its expression.

Mode of interaction of Pirin with its direct interaction partners and associated consequences

The four proteins that directly interact with Pirin are NFIX, BCL3, NFKBIA and SMAD9 (Table 1). Pirin was identified through Y2H system based screening of the interaction partners of NFIX [6, 83]. NFIX is a master regulator for metastasis of lung cancer [84]. The iron cofactor is required for interaction of Pirin with NFIX and BCL3 [8]. The Fe2+ bound conformation of Pirin appears to bind to the putative binding regions of the direct interactors with higher affinities and has more resemblance to the native pose in comparison with the Fe3+ bound conformation (Figs 14 and Table 2).

Regulation of NF-κB signaling pathway.

The NF-κB family of transcription factors (TFs) consists of five members- p65 (RelA), RelB, c-Rel, p50 (NF-κB1), and p52 (NF-κB2), which form transcriptionally active homodimers (except Rel B) and heterodimers [5, 85, 86]. In unstimulated cells, NF-κB dimers are sequestered in an inactive form through association with one of three typical inhibitor of NF-κB (IκB) proteins: IκBα (encoded by NFKBIA), IκBβ (NFKBIB), and IκBɛ (NFKBIE), or the precursor proteins p100 (NFKB2) and p105 (NFKB1) or the atypical IκB proteins: IκBζ, BCL3 and IκBNS via the ankyrin repeat domains of these inhibitors [85, 87, 88]. Upon stimulation of cells by agents, such as TNFA, IL1 or numerous pathogens or pathogenic proteins, like members of the Poxviridae, N-protein of SARS-CoV-2 as well as pathogenic bacteria, the IκB kinase (IKK) complex is activated that in turn phosphorylates IκB molecules on two serine residues and mediates their ubiquitination by the SCF E3 ligase and subsequent degradation by the 26S proteasome [4, 87, 89, 90]. As a result of degradation of the IκB inhibitors, NF-κB dimer is freed and able to translocate into the nucleus to initiate a transcriptional response [4].

Various NF-κB complexes, predominantly the p65/p50 heterodimers activated via IκBα-degradation regulate transcription of target genes in the canonical pathway, while the RelB/p52 heterodimer formed via inducible proteasomal processing of p100 to p52 drives a transcriptional response in the non-canonical pathway [88, 91]. Sequestration by IκBα is important for modulating NF-κB signaling as functional polymorphisms in the NFKBIA promoter and 3′ untranslated regions are significantly associated with cancers, such as HBV-induced hepatocarcinogenesis [92, 93]. Co-fractionation process indicated interaction (indirect) between Pirin and ubiquitin-conjugating enzyme E2 L3 (UBE2L3) (Table 1), which is a ubiquitin-conjugating enzyme with roles in ubiquitination cascade of proteins for generating signals for 26S proteasome dependent protein degradation (KEGG: hsa04120) [45]. Pirin signaling includes two subunits of 26S proteasome that are proteasome subunit alpha type-7 (PSMA7) and proteasome subunit, beta type, 2 (PSMB2) [94], among which PSMB2 is a first degree interactor of Pirin (Table 1 and Fig 5). Pirin may contribute to NF-κB signaling through direct interaction with IκBα (NFKBIA) and subsequently mediating its proteasomal degradation. The nuclear localization of Pirin contradicts such role, but presence of Pirin in cytoplasm in a subset of melanomas has been reported previously [7]. Additionally, despite an exclusively cytosolic steady state localization of IkBα/p50 complexes, these are constantly shuttled between the cytosol and nucleus using the nuclear export sequence (NES) of IkBα and the nuclear localization sequence (NLS) of p50 [5]. Thus, it might not be unlikely that over-expression of Pirin dysregulate sequestration of NF-κB by IκBα and lead to associated diseases.

The carboxy-terminal transactivation domains, that activate transcription at the κB-sites in target genes, are present in RelA, RelB and c-Rel, but absent in the p50 and p52 homodimers [86]. BCL3 is a transactivation domain containing oncoprotein that is located predominantly in the nucleus [95]. BCL3 provides transactivation domains to p50 and p52 homodimers by interacting with these complexes and promote transcription of NF-κB target genes [96]. Without any direct association with p50, Pirin enhances the amount of the DNA binding activity of p50-BCL3 by interacting with the ankyrin repeat domains of BCL3 and forming a quaternary complex [8, 97].

Along with activation, binding of BCL3 to p50 and p52 homodimers can lead to repression of a subset of NF-κB regulated genes via stabilizing repressive p50 homodimers or recruiting co-repressors [96]. However, BCL3 can also dissociate repressive p50 homodimers from κB-sites on DNA by sequestering these in a complex, which allows p65/p50 heterodimers to activate transcription at these sites and mediate canonical signaling pathway [96, 97]. Such removal of p50 and p52 homodimers from bound DNA is regulated by unphosphorylated BCL3 that plays inhibitory roles similar to a typical IκB family member [98]. One of the first degree interactors of Pirin is PPP2CA (Fig 5), which is a serine/threonine phosphatase with activities critical for maintaining healthy cellular functions and suppressing tumor [99]. Pirin may interact with BCL3 and PPP2CA simultaneously to mediate the shift from non-canonical to canonical NF-κB signaling through dephosphorylation of BCL3, although still there is no study confirming any such role.

Shifting between different modes of NF-κB signaling pathway.

Change in the oxidation state causes structural change in Pirin through altering a R-shaped surface loop that includes the area surrounding the N-terminal metal-binding cavity and the interface between the two cupin domains [9]. This R-shaped surface region interacts with the C-terminal Rel homology domain of p65 and the Fe3+ conformation enhances the binding of p65 to the κB-target site [9]. As the iron cofactor plays an important role in interaction between BCL3 and Pirin [8], change in the distance of the metal ligand from the surface by change in oxidation state [9] can be the reason why BCL3 does not interact with same energy with both the conformations of Pirin (Fig 1 and Table 2). It appears that the Fe(II) conformation mediates the non-canonical NF-κB through strengthening the function of p50-BCL3 on κB-sites and the Fe(III) conformation with no such role on p50-BCL3-DNA complex contributes critically to the p65 regulated canonical pathway with a key role of redox state of cell. This may explain the shift between different NF-κB pathways with full reversibility [9].

Transcriptional co-regulation.

BCL3 expression is elevated in various hematopoietic and solid cancers, including breast and hepatocellular carcinomas [95, 100]. This oncoprotein acts as a bridging factor between NF-κB and nuclear co-regulators [97]. The same conformation, but completely different regions (as observed by overlaying the docked structures in Chimera 1.15rc [101], of Pirin interacts with NFIX and BCL3 (Figs 1 and 3). NFI family members can modulate activation and repression of transcription of genes [6]. Pirin may connect NFIX to BCL3-p50 or BCL3-p52 homodimer bound genes, but definitive conclusions regarding effect of NFIX on transcription of κB-genes await further study. Based on the in silico data (Table 1 and Fig 4), SMAD9 directly interacts with Pirin. SMAD9 is a transcriptional repressor of bone morphogenetic protein (BMP) signaling [102]. Rare sequence variants in SMAD9 were reported to contribute to pathogenesis of pulmonary arterial hypertension [103]. SMAD9 protein is associated with colorectal neoplasms, familial pulmonary hypertension and pulmonary arterial hypertension (Fig 7).

Functional and disease enrichment for Pirin signaling

Actin cytoskeleton remodeling and regulation of metastasis of cancer cells.

Pirin signaling is significantly involved in regulation of actin dynamics, actin filament fragmentation as well as Rho GTPases activated WASPs and WAVEs pathway (Figs 6 and 7). Through activating Wiskott–Aldrich syndrome family of proteins, such as WASP and WAVE1/2, Rho GTPases stimulate formation of lamellipodia and filopodia that are involved in directional motility of cells and invasiveness and metastasis of cancer cells [104]. Pirin signaling includes (Table 1) Wiskott-Aldrich syndrome protein family member 2 (WASF2) and RAC1 which are associated with lamellipodia assembly (Fig 5). Pirin also interacts with cytoplasmic protein NCK1, which mediates cancer metastasis [105, 106] as well as functions as an intracellular messenger leading to angiogenesis in the ERBB signaling pathway (KEGG entry: hsa04012) [45]. RAC1, together with NCK, mediates activation of the Arp2/3 complex leading to polymerization of the actin cytoskeleton to form lamellipodia typical of mesenchymal movements as well as tumor metastasis and angiogenesis [107, 108]. So, role of Pirin in cancers appears to be more centered to migration and invasion of tumor cells, instead of growth and initiation.

The most enriched KEGG [45] pathways and DisGeNET [51] analyzed diseases for interactors of Pirin are associated with cancers (Figs 7 and 8). Earlier reports also suggest a role Pirin in different cancers [16, 17, 109, 110]. Interrupting the interaction between Pirin and BCL3 with a Pirin inhibitor has been shown to inhibit melanoma cell metastasis via suppression of snail homolog 2 (SNAI2) [16]. Pirin can also mediate metastasis of cervical cancer cells independent of BCL3-SNAI2 signaling [17]. In oral and cervical cells, PIR gene silencing with small interfering RNA (siRNA) was shown to increase E-cadherin transcripts as well as reduce Vimentin, Slug, Zeb and Snail transcripts in oral and cervical cancer cells [109, 110]. Furthermore, high-risk human papillomavirus (HR-HPV) oncoproteins enhance levels of Pirin in both epithelial cervical and oral cancer cells [109]. In oral tumor cells, HPV16 E7 oncoprotein mediated induction of the EGFR/PI3K/AKT1/NRF2 pathway and recruitment of NRF2 in the PIR promoter was found to lead to Pirin/NF-κB activation which in turn enhanced epithelial to mesenchymal transition (EMT) and cell migration [10, 111].

Pirin is involved in a pro-fibrotic signaling pathway depending on myocardin-related transcription factor (MRTF) and serum response factor (SRF) which are activated downstream of the Rho GTPases [112]. Along with these enhancers of metastasis, Pirin also interacts with destrin (DSTN), cofilin (CFL) and gelsolin-like actin-capping protein CAPG (Table 1), which have opposite effects and play a role in actin filament fragmentation/depolymerization (Fig 6). Cofilin reverses the process of polymerization by converting F-actin filaments into G-actin monomers, but this reversal can be inactivated by RAC1 [107]. Rho GDP dissociation inhibitor (GDI) alpha (ARHGDIA) has a first degree association with Pirin (Fig 5). ARHGDIA is an ubiquitously expressed interactor of several Rho GTPases, including RAC1 and it blocks activation of Rho proteins through sequestration of the inactive GDP-bound Rho proteins in the cytosol and inhibiting the switch to active GTP-bound states [113]. Association between Pirin and ARHGDIA may impede this blockage and allow Rho GTPases to exert their function. Pirin also interacts with breast cancer anti-estrogen resistance 1 (BCAR1) protein that activates RAC1 (Fig 5). The exact mechanism of how interaction with these proteins contribute to Pirin mediated metastasis of cancer cells is yet to be known, but Pirin signaling appears to play a key role in cancer invasiveness and migration.

Role in neuropathological complications.

Various neurodegenerative conditions, including Alzheimer’s disease, motor neuron disease, Parkinsonism, Ramsay Hunt paralysis syndrome and amyotrophic lateral sclerosis (Guam form) are enriched for Pirin and its partnering proteins (Fig 8a). Abundant abnormal aggregates of neuronal cytoskeletal proteins are signatures of many neurodegenerative diseases [114]. Balanced cofilin activity is a prerequisite for actin turnover and proper central nervous system (CNS) functions [115].

Piwi-like proteins, including as PIWIL1, PIWIL2, and PIWIL4, are among the Pirin interactors (Table 1). These proteins are associated with negative regulation of transposition (Fig 6). These proteins play a vital role in maintaining piRNA activity, which is increased in neuropathological conditions [116]. In previous studies, PIWIL1, PIWIL2, and PIWIL4 have been reported to function in cancer cell proliferation, metastasis, and invasion [117, 118].

Involvement in infection associated body processes and complications.

For Pirin and its interaction partners, disease conditions correlated to pathogenic infections, such as Tuberculosis, Hepatitis B and viral diseases, as well as the body’s response to the infections, including regulation of actin dynamics for phagocytic cup formation and Fc gamma receptor (FCGR) dependent phagocytosis, are enriched (Figs 6 and 8). Phagocytic cups, an actin-based structure at the plasma membrane of a phagocyte, are essential for phagocytosis and digestion of pathogens, and WASP plays a key role in their formation [119, 120]. In addition, activation of RAC1 and inhibition of cofilin is part of the regulation of actin cytoskeleton in FCGR mediated phagocytosis (KEGG entry: hsa04666) [45], which may be mediated by Pirin, as mentioned previously. Furthermore, the relation of viral infection to the development of neurological disorders such as Parkinson’s disease, Alzheimer’s disease, and multiple sclerosis is well known [121]. For example, SARS‐CoV‐2 can trigger cellular processes involved in acute and subacute neurodegeneration by entering the brain [122, 123]. Involvement of Pirin signaling in actin cytoskeleton reorganization, phagocytic cup formation, and interaction with actin filament modulating proteins suggest association of excessive Pirin production with the development of neurological complications, in presence or absence of infection.

Oxidative stress induced regulation of cell death.

Basal expression of PIR can be modulated by nuclear factor (erythroid-derived 2)-like 2 (NRF2; encoded by the NFE2L2 gene) transcription factor through functional antioxidant response elements (AREs) in its promoter region [124, 125]. Knockdown of NRF2 in HeLa cells resulted in decreased PIR mRNA and protein levels [126]. NRF2 is triggered in mammals as a protective response to cellular oxidative, inflammatory and electrophilic stress [124, 127] may lead to excessive Pirin production.

Regulation of intrinsic apoptotic signaling pathway, oxidative stress induced cell death and response to endoplasmic reticulum stress are significantly enriched for Pirin interactome and interactors. For example, NCK1, superoxide dismutase 1 (SOD1), Parkinson protein 7 (PARK7), peptidylprolyl isomerase A (PPIA), receptor for activated C kinase (RACK1), and HtrA serine peptidase 2 (HTRA2) are associated with these processes (Fig 6). Upregulation of Pirin in the airway epithelium in response to the acute oxidative stress imposed by cigarette smoke was found to induce apoptosis [128]. Additionally, Pirin has been shown to play role in resistance to ferroptosis, which is an iron-dependent non-apoptotic cell death, in human pancreatic cancer cells [129]. On the other hand, Pirin was reported to be a negative regulator of senescence in melanocytic cells [130]. Thus, Pirin interactome is involved in regulation of apoptotic or non-apoptotic cell death but the mode of regulation cannot be inferred conclusively.

Role in diabetic complications.

Oxidative stress has a major contribution to the pathogenesis both microvascular and cardiovascular diabetic complications [131]. As mentioned earlier, ferric conformation of Pirin plays a vital role in NF-κβ p65 pathway activation. The canonical NF-κB pathway mediated by P65/p50 heterodimer directly induces the production of proinflammatory cytokines, such as TNFA and IL6 [88]. Proinflammatory NF-κβ pathway plays a critical role in pathophysiology of diabetes as well as associated vascular complications, such as diabetic retinopathy, diabetic nephropathy and cardiomyopathy [132, 133]. Pirin, RELA (p65) and NFKBIA are significantly associated with endocrine system diseases (Fig 8b). Pirin hyperactivated by oxidative stress may contribute to pathological inflammation involved in diabetic complications.

Involvement other conditions.

Transient ischemic attacks (TIAs) were enriched for Pirin signaling (Fig 8a). Recruitment of platelets to the ischemic region and their aggregation can result in TIAs [134, 135]. Reactome pathway enrichment analysis (Fig 7b) demonstrated involvement of Pirin interactome in platelet activation and aggregation as well as αIIb/β3 receptor signaling that is essential for platelet aggregation [62].

Additionally, glucose, amino acid and nucleotide sugar metabolism is significantly associated with Pirin interactome (Fig 6). Pirin is also associated to osteoporosis (Fig 8b) and NF-κB contributes to bone formation impairment in osteoporosis [136].

It appears that Pirin is associated with a myriad of inflammatory and neurodegenerative diseases as well as metastasis of cells through interaction with other proteins.

Abnormal expression of Pirin by regulatory variants

Pirin production can be modulated by expression modifying (modifier) regulatory variants (Tables 3 and 4). CpG island downstream from the transcription start site (TSS) of the PIR gene is crucial for its expression [126]. For regulatory variants rs565388697, rs539142342, rs369918088, rs190912217, rs187777697, rs1567894, rs372563960, rs183910461, and rs148530113, the CpG island was found to be the related regulatory element (Table 3). rs180708754 can influence the binding of miRNA to the PIR gene.

PIR gene upstream variant rs4830964 and intron variant rs2094 may regulate expression of not only PIR gene, but also vascular endothelial growth factor D (VEGFD), carbonic anhydrase 5B pseudogene 1 (CA5BP1), Fanconi anemia group B protein (FANCB) and Transmembrane protein 27 (TMEM27) (Tables 2 and 3). Related regulatory elements for these two variants are TF binding region and chromatin interactive region. Both rs5980162 and rs1567894 are intron variants that regulate expression of PIR and VEGFD across various tissues (S2 Table). Variant alleles at rs4830964, rs2094, rs5980162 and rs1567894 exist with > 0.3 frequency in African, admixed American, East Asian and European populations but with < 0.3 frequency in South Asian populations (Table 4). This may cause disparity in PIR expression and lead to differential prevalence of Pirin mediated inflammatory conditions and aberrant actin cytoskeleton remodeling involved diseases (as discussed previously) among these populations. For example, NF-κB pathway, that is regulated by Pirin [9, 13], plays a central role in pro-inflammatory cytokine response observed in COVID-19 patients [137] and interestingly, COVID-19 death rate have been comparatively lower in the South Asian countries compared to the other nations in Europe, United states and Asia [138].

Expression of both PIR and ACE2 can be regulated by rs6629104 and rs6629105 in nucleus accumbens of basal ganglia within the brain (Table 2 and S2 Table). mRNA abundance is an associated trait for these two intron variants of PIR (Table 2). Dysregulated PIR expression in brain regions may be associated with neurodegenerative diseases (as discussed above) as well as with neurological tumors [10]. Variant alleles at rs6629104 and rs6629105 are present with > 0.4 frequency in South- and East Asian populations, but with comparatively lower frequencies in African and European populations. ACE2 expression is enhanced by rs6629105 T allele in Nucleus accumbens (S1 Fig). Increased ACE2 expression in brain regions can be correlated to enhanced neuro-invasion of SARS-CoV-2 and associated neurological symptoms [139].

This study provides mechanistic insights into how Pirin’s conformational alterations regulate its interaction with key inflammatory regulators, as well as its function in tumor invasion, and other metabolic and neuropathological complications. Further in vivo and in vitro studies will shed more light on such interactions. Because of its important role in critical pathways that are associated with multiple disease conditions, Pirin might be considered as an important drug target, particularly as a bypass route to suppress over-activation of the NF-κB pathway for treating chronic inflammation-mediated diseases. As the frequency of certain regulatory variants of the PIR gene varies among populations of different ethnicities, additional research on the variants of Pirin and associated phenotypic changes is necessary to determine their connection with disease conditions. These variants may be taken into consideration while developing drugs or predicting outcomes. These PIR variants may also be taken into consideration while explaining the global incidences of associated disease conditions.

Conclusion

Dysregulated inflammatory networks stand at the forefront of highly prevalent human diseases. Identification of novel inhibitory target(s) for pathogenic inflammation is highly needed. Due to the lack of studies on the functions of the Pirin protein, an interactome-based approach was applied for the characterization of its functionality. Both conformations, Fe(II)- and Fe(III)-bound, of this protein are intricately associated with the NF-κB inflammatory pathway. Along with its role in the regulation of NF-κB activity, Pirin signaling appears to play a role in cytoskeleton remodeling, metastasis and invasion of tumors, platelet aggregation, and stroke. Pirin may be an effective target for anti-inflammatory and anti-metastatic treatments. There are regulatory variants in the PIR gene that not only regulate the expression of PIR but also the ACE2 gene, which is the targeted binding site of SARS-CoV-2.

Supporting information

S1 Table. Domains of binding partners of Pirin.

https://doi.org/10.1371/journal.pone.0289158.s001

(DOCX)

S2 Table. eQTLs in PIR and their regulated genes across different tissues.

https://doi.org/10.1371/journal.pone.0289158.s002

(DOCX)

S1 Fig. Violin plot representing normalized expression of PIR and ACE2 genes in nucleus accumbens region of brain depending on the genotype at rs6629105.

The number of subjects is shown under each genotype. The median value of the gene expression at each genotype is indicated by the white lines in the black box plots.

https://doi.org/10.1371/journal.pone.0289158.s003

(PNG)

Acknowledgments

The authors are thankful to the Ministry of Science and Technology, Bangladesh, for its support.

References

  1. 1. Chen L, Deng H, Cui H, Fang J, Zuo Z, et al. (2018) Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 9: 7204–7218. pmid:29467962
  2. 2. Bennett JM, Reeves G, Billman GE, Sturmberg JP (2018) Inflammation–nature’s way to efficiently respond to all types of challenges: Implications for understanding and managing “the Epidemic” of chronic diseases. Frontiers in Medicine 5: 316. pmid:30538987
  3. 3. Furman D, Campisi J, Verdin E, Carrera-Bastos P, Targ S, et al. (2019) Chronic inflammation in the etiology of disease across the life span. Nature Medicine 25: 1822–1832. pmid:31806905
  4. 4. Gilmore TD, Herscovitch M (2006) Inhibitors of NF-κB signaling: 785 and counting. Oncogene 25: 6887–6899.
  5. 5. Oeckinghaus A, Ghosh S (2009) The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb Perspect Biol 1: a000034. pmid:20066092
  6. 6. Wendler WM, Kremmer E, Förster R, Winnacker EL (1997) Identification of pirin, a novel highly conserved nuclear protein. Journal of Biological Chemistry 272: 8482–8489. pmid:9079676
  7. 7. Licciulli S, Luise C, Zanardi A, Giorgetti L, Viale G, et al. (2010) Pirin delocalization in melanoma progression identified by high content immuno-detection based approaches. BMC Cell Biol 11: 5. pmid:20089166
  8. 8. Pang H, Bartlam M, Zeng Q, Miyatake H, Hisano T, et al. (2004) Crystal structure of human pirin: an iron-binding nuclear protein and transcription cofactor. Journal of Biological Chemistry 279: 1491–1498. pmid:14573596
  9. 9. Liu F, Rehmani I, Esaki S, Fu R, Chen L, et al. (2013) Pirin is an iron-dependent redox regulator of NF-κB. Proc Natl Acad Sci U S A 110: 9722–9727.
  10. 10. Perez-Dominguez F, Carrillo-Beltrán D, Blanco R, Muñoz JP, León-Cruz G, et al. (2021) Role of Pirin, an oxidative stress sensor protein, in epithelial carcinogenesis. Biology (Basel) 10: 116. pmid:33557375
  11. 11. Adams M, Jia Z (2005) Structural and biochemical analysis reveal Pirins to possess quercetinase activity. Journal of Biological Chemistry 280: 28675–28682. pmid:15951572
  12. 12. Barman A, Hamelberg D (2016) Fe(II)/Fe(III) redox process can significantly modulate the conformational dynamics and electrostatics of Pirin in NF-κB regulation. ACS Omega 1: 837–842.
  13. 13. Adeniran C, Hamelberg D (2017) Redox-specific allosteric modulation of the conformational dynamics of κB DNA by Pirin in the NF-κB supramolecular complex. Biochemistry 56: 5002–5010.
  14. 14. Maldonado V, Melendez-Zajgla J (2011) Role of Bcl-3 in solid tumors. Molecular Cancer 10: 152. pmid:22195643
  15. 15. Suleman M, Chen A, Ma H, Wen S, Zhao W, et al. (2019) PIR promotes tumorigenesis of breast cancer by upregulating cell cycle activator E2F1. Cell Cycle 18: 2914–2927. pmid:31500513
  16. 16. Miyazaki I, Simizu S, Okumura H, Takagi S, Osada H (2010) A small-molecule inhibitor shows that pirin regulates migration of melanoma cells. Nature Chemical Biology 6: 667–673. pmid:20711196
  17. 17. Komai K, Niwa Y, Sasazawa Y, Simizu S (2015) Pirin regulates epithelial to mesenchymal transition independently of Bcl3-Slug signaling. FEBS Letters 589: 738–743. pmid:25680527
  18. 18. Shoily SS, Ahsan T, Fatema K, Sajib AA (2021) Disparities in COVID-19 severities and casualties across ethnic groups around the globe and patterns of ACE2 and PIR variants. Infect Genet Evol 92: 104888. pmid:33933634
  19. 19. Porras P, Barrera E, Bridge A, del-Toro N, Cesareni G, et al. (2020) Towards a unified open access dataset of molecular interactions. Nature Communications 11: 6144. pmid:33262342
  20. 20. Kerrien S, Aranda B, Breuza L, Bridge A, Broackes-Carter F, et al. (2012) The IntAct molecular interaction database in 2012. Nucleic Acids Research 40: D841–D846. pmid:22121220
  21. 21. Oughtred R, Stark C, Breitkreutz B-J, Rust J, Boucher L, et al. (2019) The BioGRID interaction database: 2019 update. Nucleic Acids Research 47: D529–D541. pmid:30476227
  22. 22. Orchard S, Kerrien S, Abbani S, Aranda B, Bhate J, et al. (2012) Protein interaction data curation: the International Molecular Exchange (IMEx) consortium. Nature Methods 9: 345–350. pmid:22453911
  23. 23. Orchard S, Ammari M, Aranda B, Breuza L, Briganti L, et al. (2014) The MIntAct project—IntAct as a common curation platform for 11 molecular interaction databases. Nucleic Acids Research 42: D358–D363. pmid:24234451
  24. 24. Licata L, Briganti L, Peluso D, Perfetto L, Iannuccelli M, et al. (2012) MINT, the molecular interaction database: 2012 update. Nucleic Acids Research 40: D857–D861. pmid:22096227
  25. 25. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, et al. (2019) STRING v11: Protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Research 47: D607–D613. pmid:30476243
  26. 26. Calderone A, Castagnoli L, Cesareni G (2013) mentha: A resource for browsing integrated protein-interaction networks. Nature Methods 10: 690–691. pmid:23900247
  27. 27. del Toro N, Shrivastava A, Ragueneau E, Meldal B, Combe C, et al. (2022) The IntAct database: efficient access to fine-grained molecular interaction data. Nucleic Acids Research 50: D648–D653. pmid:34761267
  28. 28. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, et al. (2021) The STRING database in 2021: customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Research 49: D605–D612. pmid:33237311
  29. 29. Yang J-S, Garriga-Canut M, Link N, Carolis C, Broadbent K, et al. (2018) rec-YnH enables simultaneous many-by-many detection of direct protein–protein and protein–RNA interactions. Nature Communications 9: 3747. pmid:30217970
  30. 30. Brückner A, Polge C, Lentze N, Auerbach D, Schlattner U (2009) Yeast two-hybrid, a powerful tool for systems biology. Int J Mol Sci 10: 2763–2788. pmid:19582228
  31. 31. The UniProt C (2021) UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Research 49: D480–D489. pmid:33237286
  32. 32. Mistry J, Chuguransky S, Williams L, Qureshi M, Salazar Gustavo A, et al. (2021) Pfam: The protein families database in 2021. Nucleic Acids Research 49: D412–D419. pmid:33125078
  33. 33. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, et al. (2000) The Protein Data Bank. Nucleic Acids Research 28: 235–242. pmid:10592235
  34. 34. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, et al. (2021) Highly accurate protein structure prediction with AlphaFold. Nature 596: 583–589. pmid:34265844
  35. 35. Berman H, Henrick K, Nakamura H (2003) Announcing the worldwide Protein Data Bank. Nature Structural & Molecular Biology 10: 980–980.
  36. 36. Waterhouse A, Bertoni M, Bienert S, Studer G, Tauriello G, et al. (2018) SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Research 46: W296–W303. pmid:29788355
  37. 37. Weng G, Wang E, Wang Z, Liu H, Zhu F, et al. (2019) HawkDock: a web server to predict and analyze the protein-protein complex based on computational docking and MM/GBSA. Nucleic Acids Res 47: W322–W330. pmid:31106357
  38. 38. Hou T, Wang J, Li Y, Wang W (2011) Assessing the performance of the MM/PBSA and MM/GBSA methods. 1. The accuracy of binding free energy calculations based on molecular dynamics simulations. Journal of Chemical Information and Modeling 51: 69–82. pmid:21117705
  39. 39. Malhotra S, Mathew OK, Sowdhamini R (2015) DOCKSCORE: A webserver for ranking protein-protein docked poses. BMC Bioinformatics 16: 127. pmid:25902779
  40. 40. Wang J, Youkharibache P, Marchler-Bauer A, Lanczycki C, Zhang D, et al. (2022) iCn3D: From web-based 3D viewer to structural analysis tool in batch mode. Frontiers in Molecular Biosciences 9: 831740. pmid:35252351
  41. 41. Moreira IS, Koukos PI, Melo R, Almeida JG, Preto AJ, et al. (2017) SpotOn: High accuracy identification of protein-protein interface hot-spots. Scientific Reports 7: 8007. pmid:28808256
  42. 42. Zhou G, Soufan O, Ewald J, Hancock REW, Basu N, et al. (2019) NetworkAnalyst 3.0: A visual analytics platform for comprehensive gene expression profiling and meta-analysis. Nucleic Acids Res 47: W234–W241. pmid:30931480
  43. 43. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, et al. (2003) Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res 13: 2498–2504. pmid:14597658
  44. 44. Reimand J, Isserlin R, Voisin V, Kucera M, Tannus-Lopes C, et al. (2019) Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap. Nature Protocols 14: 482–517. pmid:30664679
  45. 45. Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M (2021) KEGG: Integrating viruses and cellular organisms. Nucleic Acids Res 49: D545–D551. pmid:33125081
  46. 46. Jassal B, Matthews L, Viteri G, Gong C, Lorente P, et al. (2020) The reactome pathway knowledgebase. Nucleic Acids Res 48: D498–D503. pmid:31691815
  47. 47. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, et al. (2009) ClueGO: A Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25: 1091–1093. pmid:19237447
  48. 48. Bindea G, Galon J, Mlecnik B (2013) CluePedia Cytoscape plugin: Pathway insights using integrated experimental and in silico data. Bioinformatics 29: 661–663. pmid:23325622
  49. 49. Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, et al. (2016) Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 44: W90–97. pmid:27141961
  50. 50. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, et al. (2013) Enrichr: Interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14: 128. pmid:23586463
  51. 51. Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J, Ronzano F, Centeno E, et al. (2020) The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Research 48: D845–D855. pmid:31680165
  52. 52. Guo L, Du Y, Qu S, Wang J (2016) rVarBase: An updated database for regulatory features of human variants. Nucleic Acids Res 44: D888–D893. pmid:26503253
  53. 53. McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GRS, et al. (2016) The Ensembl Variant Effect Predictor. Genome Biology 17: 122. pmid:27268795
  54. 54. GTex-Consortium, Aguet F, Anand S, Ardlie KG, Gabriel S, et al. (2020) The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369: 1318–1330. pmid:32913098
  55. 55. Auton A, Abecasis GR, Altshuler DM, Durbin RM, Abecasis GR, et al. (2015) A global reference for human genetic variation. Nature 526: 68–74. pmid:26432245
  56. 56. Machiela MJ, Chanock SJ (2015) LDlink: A web-based application for exploring population-specific haplotype structure and linking correlated alleles of possible functional variants. Bioinformatics 31: 3555–3557. pmid:26139635
  57. 57. Xia J, Gill EE, Hancock REW (2015) NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nature Protocols 10: 823–844. pmid:25950236
  58. 58. Appert-Collin A, Hubert P, Crémel G, Bennasroune A (2015) Role of ErbB receptors in cancer cell migration and invasion. Frontiers in Pharmacology 6: 283. pmid:26635612
  59. 59. Yarden Y, Sliwkowski MX (2001) Untangling the ErbB signalling network. Nature Reviews Molecular Cell Biology 2: 127–137. pmid:11252954
  60. 60. D’Souza A, Spicer D, Lu J (2018) Overcoming endocrine resistance in metastatic hormone receptor-positive breast cancer. Journal of Hematology & Oncology 11: 80.
  61. 61. Marteyn B, Gazi A, Sansonetti P (2012) Shigella: A model of virulence regulation in vivo. Gut microbes 3: 104–120. pmid:22356862
  62. 62. Payrastre B, Missy K, Trumel C, Bodin S, Plantavid M, et al. (2000) The integrin αIIb/β3 in human platelet signal transduction. Biochemical Pharmacology 60: 1069–1074.
  63. 63. Riha P, Rudd CE (2010) CD28 co-signaling in the adaptive immune response. Self Nonself 1: 231–240. pmid:21487479
  64. 64. Heppner FL, Ransohoff RM, Becher B (2015) Immune attack: The role of inflammation in Alzheimer disease. Nature Reviews Neuroscience 16: 358–372. pmid:25991443
  65. 65. Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, et al. (2018) Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y) 4: 575–590. pmid:30406177
  66. 66. Verber NS, Shepheard SR, Sassani M, McDonough HE, Moore SA, et al. (2019) Biomarkers in motor neuron disease: A state of the art review. Frontiers in Neurology 10: 291. pmid:31001186
  67. 67. Foster LA, Salajegheh MK (2019) Motor neuron disease: Pathophysiology, diagnosis, and management. The American Journal of Medicine 132: 32–37. pmid:30075105
  68. 68. Ahlskog JE, Litchy WJ, Petersen RC, Waring SC, Esteban-Santillan C, et al. (1999) Guamanian neurodegenerative disease: Electrophysiologic findings. Journal of the Neurological Sciences 166: 28–35. pmid:10465496
  69. 69. Jankovic J (2008) Parkinson’s disease: Clinical features and diagnosis. Journal of Neurology, Neurosurgery & Psychiatry 79: 368. pmid:18344392
  70. 70. Tomiyama H, Lesage S, Tan E-K, Jeon BS (2015) Familial Parkinson’s disease/Parkinsonism. BioMed Research International 2015: 736915. pmid:25961036
  71. 71. Saito M, Maruyama M, Ikeuchi K, Kondo H, Ishikawa A, et al. (2000) Autosomal recessive juvenile parkinsonism. Brain and Development 22: 115–117.
  72. 72. Herrero M-T, Estrada C, Maatouk L, Vyas S (2015) Inflammation in Parkinson’s disease: Role of glucocorticoids. Frontiers in Neuroanatomy 9: 32. pmid:25883554
  73. 73. Johnston DC, Hill MD (2004) The patient with transient cerebral ischemia: A golden opportunity for stroke prevention. Cmaj 170: 1134–1137. pmid:15051699
  74. 74. Easton JD, Saver JL, Albers GW, Alberts MJ, Chaturvedi S, et al. (2009) Definition and evaluation of transient ischemic attack: a scientific statement for healthcare professionals from the American Heart Association/American Stroke Association Stroke Council; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Radiology and Intervention; Council on Cardiovascular Nursing; and the Interdisciplinary Council on Peripheral Vascular Disease. The American Academy of Neurology affirms the value of this statement as an educational tool for neurologists. Stroke 40: 2276–2293. pmid:19423857
  75. 75. Tseng CC, Wong MC, Liao WT, Chen CJ, Lee SC, et al. (2021) Genetic variants in transcription factor binding sites in humans: Triggered by natural selection and triggers of diseases. Int J Mol Sci 22: 4187. pmid:33919522
  76. 76. Liu C, Zhang F, Li T, Lu M, Wang L, et al. (2012) MirSNP, a database of polymorphisms altering miRNA target sites, identifies miRNA-related SNPs in GWAS SNPs and eQTLs. BMC Genomics 13: 661. pmid:23173617
  77. 77. Agarwal V, Bell GW, Nam JW, Bartel DP (2015) Predicting effective microRNA target sites in mammalian mRNAs. Elife 4: e05005. pmid:26267216
  78. 78. Sobhy H, Kumar R, Lewerentz J, Lizana L, Stenberg P (2019) Highly interacting regions of the human genome are enriched with enhancers and bound by DNA repair proteins. Scientific Reports 9: 4577. pmid:30872630
  79. 79. Peng Y, Xiong D, Zhao L, Ouyang W, Wang S, et al. (2019) Chromatin interaction maps reveal genetic regulation for quantitative traits in maize. Nature Communications 10: 2632. pmid:31201335
  80. 80. Wang J, Ma X, Zhang Q, Chen Y, Wu D, et al. (2021) The interaction analysis of SNP variants and DNA methylation identifies novel methylated pathogenesis genes in congenital heart diseases. Frontiers in Cell and Developmental Biology 9: 665514. pmid:34041244
  81. 81. Gibbs JR, van der Brug MP, Hernandez DG, Traynor BJ, Nalls MA, et al. (2010) Abundant quantitative trait loci exist for DNA methylation and gene expression in human brain. PLOS Genetics 6: e1000952. pmid:20485568
  82. 82. Lugrin J, Rosenblatt-Velin N, Parapanov R, Liaudet L (2014) The role of oxidative stress during inflammatory processes. Biological Chemistry 395: 203–230. pmid:24127541
  83. 83. Brzóska K, Stępkowski TM, Kruszewski M (2011) Putative proto-oncogene Pir expression is significantly up-regulated in the spleen and kidney of cytosolic superoxide dismutase-deficient mice. Redox Report 16: 129–133. pmid:21801495
  84. 84. Rahman NIA, Abdul Murad NA, Mollah MM, Jamal R, Harun R (2017) NFIX as a master regulator for lung cancer progression. Frontiers in Pharmacology 8: 540. pmid:28871224
  85. 85. Hayden MS, Ghosh S (2008) Shared principles in NF-κB signaling. Cell 132: 344–362.
  86. 86. Rico-Rosillo G, Vega-Robledo GB (2011) The involvement of NF-κB transcription factor in asthma. Rev Alerg Mex 58: 107–111. pmid:21967970
  87. 87. Bhatt D, Ghosh S (2014) Regulation of the NF-κB-mediated transcription of inflammatory genes. Frontiers in Immunology 5: 71.
  88. 88. Yu H, Lin L, Zhang Z, Zhang H, Hu H (2020) Targeting NF-κB pathway for the therapy of diseases: Mechanism and clinical study. Signal Transduction and Targeted Therapy 5: 209.
  89. 89. Wu Y, Ma L, Cai S, Zhuang Z, Zhao Z, et al. (2021) RNA-induced liquid phase separation of SARS-CoV-2 nucleocapsid protein facilitates NF-κB hyper-activation and inflammation. Signal Transduction and Targeted Therapy 6: 167.
  90. 90. Rahman MM, McFadden G (2011) Modulation of NF-κB signalling by microbial pathogens. Nature Reviews Microbiology 9: 291–306.
  91. 91. Sun S-C (2011) Non-canonical NF-κB signaling pathway. Cell Research 21: 71–85.
  92. 92. Cheng C-W, Su J-L, Lin C-W, Su C-W, Shih C-H, et al. (2013) Effects of NFKB1 and NFKBIA gene polymorphisms on hepatocellular carcinoma susceptibility and clinicopathological features. PLOS ONE 8: e56130. pmid:23457512
  93. 93. Zhang Q, Ji XW, Hou XM, Lu FM, Du Y, et al. (2014) Effect of functional nuclear factor-kappaB genetic polymorphisms on hepatitis B virus persistence and their interactions with viral mutations on the risk of hepatocellular carcinoma. Annals of Oncology 25: 2413–2419. pmid:25223483
  94. 94. Tanaka K (2009) The proteasome: Overview of structure and functions. Proceedings of the Japan Academy, Series B 85: 12–36. pmid:19145068
  95. 95. Chang T-P, Vancurova I (2014) Bcl3 regulates pro-survival and pro-inflammatory gene expression in cutaneous T-cell lymphoma. Biochimica et Biophysica Acta (BBA)—Molecular Cell Research 1843: 2620–2630. pmid:25089799
  96. 96. Legge DN, Chambers AC, Parker CT, Timms P, Collard TJ, et al. (2020) The role of B-Cell Lymphoma-3 (BCL-3) in enabling the hallmarks of cancer: Implications for the treatment of colorectal carcinogenesis. Carcinogenesis 41: 249–256. pmid:31930327
  97. 97. Dechend R, Hirano F, Lehmann K, Heissmeyer V, Ansieau S, et al. (1999) The Bcl-3 oncoprotein acts as a bridging factor between NF-κB/Rel and nuclear co-regulators. Oncogene 18: 3316–3323.
  98. 98. Wang Y-F, Li Y, Kim D, Zhong X, Du Q, et al. (2017) Bcl3 phosphorylation by Akt, Erk2, and IKK is required for its transcriptional activity. Molecular Cell 67: 484–497.e485. pmid:28689659
  99. 99. Mazhar S, Taylor SE, Sangodkar J, Narla G (2019) Targeting PP2A in cancer: Combination therapies. Biochimica et Biophysica Acta (BBA)—Molecular Cell Research 1866: 51–63. pmid:30401535
  100. 100. Puvvada SD, Funkhouser WK, Greene K, Deal A, Chu H, et al. (2010) NF-ĸB and Bcl-3 activation are prognostic in metastatic colorectal cancer. Oncology 78: 181–188.
  101. 101. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, et al. (2004) UCSF Chimera—A visualization system for exploratory research and analysis. Journal of Computational Chemistry 25: 1605–1612. pmid:15264254
  102. 102. Tsukamoto S, Mizuta T, Fujimoto M, Ohte S, Osawa K, et al. (2014) Smad9 is a new type of transcriptional regulator in bone morphogenetic protein signaling. Scientific Reports 4: 7596. pmid:25534700
  103. 103. Gräf S, Haimel M, Bleda M, Hadinnapola C, Southgate L, et al. (2018) Identification of rare sequence variation underlying heritable pulmonary arterial hypertension. Nature Communications 9: 1416. pmid:29650961
  104. 104. Lane J, Martin T, Weeks HP, Jiang WG (2014) Structure and role of WASP and WAVE in Rho GTPase signalling in cancer. Cancer Genomics—Proteomics 11: 155. pmid:24969695
  105. 105. Zhang F, Lu Y-x, Chen Q, Zou H-m, Zhang J-m, et al. (2017) Identification of NCK1 as a novel downstream effector of STAT3 in colorectal cancer metastasis and angiogenesis. Cellular Signalling 36: 67–78. pmid:28455144
  106. 106. He P, Sheng J, Qi J, Bai X, Li J, et al. (2022) STAT3-induced NCK1 elevation promotes migration of triple-negative breast cancer cells via regulating ERK1/2 signaling. Molecular Biology Reports 49: 267–278. pmid:34846647
  107. 107. Bid HK, Roberts RD, Manchanda PK, Houghton PJ (2013) RAC1: An amerging therapeutic option for targeting cancer angiogenesis and metastasis. Molecular Cancer Therapeutics 12: 1925–1934.
  108. 108. Møller LL, Klip A, Sylow L (2019) Rho GTPases—emerging regulators of glucose homeostasis and metabolic health. Cells 8: 434. pmid:31075957
  109. 109. Carrillo D, Muñoz JP, Huerta H, Leal G, Corvalán A, et al. Upregulation of PIR gene expression induced by human papillomavirus E6 and E7 in epithelial oral and cervical cells. Open Biology 7: 170111. pmid:29118270
  110. 110. Aedo‑Aguilera V, Carrillo‑Beltrán D, Calaf GM, Muñoz JP, Guerrero N, et al. (2019) Curcumin decreases epithelial‑mesenchymal transition by a Pirin‑dependent mechanism in cervical cancer cells. Oncol Rep 42: 2139–2148.
  111. 111. Carrillo-Beltrán D, Muñoz JP, Guerrero-Vásquez N, Blanco R, León O, et al. (2020) Human papillomavirus 16 E7 promotes EGFR/PI3K/AKT1/NRF2 signaling pathway contributing to PIR/NF-κB activation in oral cancer cells. Cancers 12: 1904.
  112. 112. Lisabeth EM, Kahl D, Gopallawa I, Haynes SE, Misek SA, et al. (2019) Identification of Pirin as a molecular target of the CCG-1423/CCG-203971 series of antifibrotic and antimetastatic compounds. ACS Pharmacology & Translational Science 2: 92–100.
  113. 113. Liang L, Li Q, Huang LY, Li DW, Wang YW, et al. (2014) Loss of ARHGDIA expression is associated with poor prognosis in HCC and promotes invasion and metastasis of HCC cells. Int J Oncol 45: 659–666. pmid:24859471
  114. 114. Cairns NJ, Lee VMY, Trojanowski JQ (2004) The cytoskeleton in neurodegenerative diseases. The Journal of Pathology 204: 438–449. pmid:15495240
  115. 115. Namme JN, Bepari AK, Takebayashi H (2021) Cofilin signaling in the CNS physiology and neurodegeneration. International Journal of Molecular Sciences 22: 10727. pmid:34639067
  116. 116. Huang X, Wong G (2021) An old weapon with a new function: PIWI-interacting RNAs in neurodegenerative diseases. Translational Neurodegeneration 10: 9. pmid:33685517
  117. 117. Han YN, Li Y, Xia SQ, Zhang YY, Zheng JH, et al. (2017) PIWI proteins and PIWI-interacting RNA: Emerging roles in cancer. Cellular Physiology and Biochemistry 44: 1–20. pmid:29130960
  118. 118. Sun T, Han X (2019) The disease-related biological functions of PIWI-interacting RNAs (piRNAs) and underlying molecular mechanisms. ExRNA 1: 21.
  119. 119. Uribe-Querol E, Rosales C (2017) Control of phagocytosis by microbial pathogens. Frontiers in Immunology 8: 1368. pmid:29114249
  120. 120. Tsuboi S, Meerloo J (2007) Wiskott-Aldrich Syndrome protein is a key regulator of the phagocytic cup formation in macrophages. Journal of Biological Chemistry 282: 34194–34203. pmid:17890224
  121. 121. Wouk J, Rechenchoski DZ, Rodrigues BCD, Ribelato EV, Faccin-Galhardi LC (2021) Viral infections and their relationship to neurological disorders. Archives of Virology 166: 733–753. pmid:33502593
  122. 122. Bouali-Benazzouz R, Benazzouz A (2021) Covid-19 infection and Parkinsonism: Is there a link? Movement Disorders 36: 1737–1743. pmid:34080714
  123. 123. Beauchamp LC, Finkelstein DI, Bush AI, Evans AH, Barnham KJ (2020) Parkinsonism as a third wave of the COVID-19 pandemic? Journal of Parkinson’s Disease 10: 1343–1353. pmid:32986683
  124. 124. Chorley BN, Campbell MR, Wang X, Karaca M, Sambandan D, et al. (2012) Identification of novel NRF2-regulated genes by ChIP-Seq: Influence on retinoid X receptor alpha. Nucleic Acids Research 40: 7416–7429. pmid:22581777
  125. 125. Hübner R-H, Schwartz JD, De BP, Ferris B, Omberg L, et al. (2009) Coordinate control of expression of Nrf2-modulated genes in the human small airway epithelium is highly responsive to cigarette smoking. Molecular Medicine 15: 203–219. pmid:19593404
  126. 126. Brzóska K, Stępkowski TM, Kruszewski M (2014) Basal PIR expression in HeLa cells is driven by NRF2 via evolutionary conserved antioxidant response element. Molecular and Cellular Biochemistry 389: 99–111. pmid:24390086
  127. 127. Calabrese EJ, Kozumbo WJ, Kapoor R, Dhawan G, Lara PC, et al. (2021) Nrf2 activation putatively mediates clinical benefits of low-dose radiotherapy in COVID-19 pneumonia and acute respiratory distress syndrome (ARDS): Novel mechanistic considerations. Radiotherapy and Oncology 160: 125–131. pmid:33932453
  128. 128. Gelbman BD, Heguy A, O’Connor TP, Zabner J, Crystal RG (2007) Upregulation of Pirin expression by chronic cigarette smoking is associated with bronchial epithelial cell apoptosis. Respiratory Research 8: 10. pmid:17288615
  129. 129. Hu N, Bai L, Dai E, Han L, Kang R, et al. (2021) Pirin is a nuclear redox-sensitive modulator of autophagy-dependent ferroptosis. Biochemical and Biophysical Research Communications 536: 100–106. pmid:33373853
  130. 130. Licciulli S, Luise C, Scafetta G, Capra M, Giardina G, et al. (2011) Pirin inhibits cellular senescence in melanocytic cells. The American Journal of Pathology 178: 2397–2406. pmid:21514450
  131. 131. Giacco F, Brownlee M (2010) Oxidative stress and diabetic complications. Circ Res 107: 1058–1070. pmid:21030723
  132. 132. Suryavanshi SV, Kulkarni YA (2017) NF-κβ: A potential target in the management of vascular complications of diabetes. Frontiers in Pharmacology 8: 798.
  133. 133. Patel S, Santani D (2009) Role of NF-kappa B in the pathogenesis of diabetes and its associated complications. Pharmacol Rep 61: 595–603. pmid:19815941
  134. 134. Yilmaz G, Granger DN (2008) Cell adhesion molecules and ischemic stroke. Neurological Research 30: 783–793. pmid:18826804
  135. 135. Al-Mefty O, Marano G, Rajaraman S, Nugent GR, Rodman N (1979) Transient ischemic attacks due to increased platelet aggregation and adhesiveness: Ultrastructural and functional correlation. Journal of Neurosurgery 50: 449–453.
  136. 136. Chang J, Wang Z, Tang E, Fan Z, McCauley L, et al. (2009) Inhibition of osteoblastic bone formation by nuclear factor-κB. Nature Medicine 15: 682–689.
  137. 137. Kircheis R, Haasbach E, Lueftenegger D, Heyken WT, Ocker M, et al. (2020) NF-κB pathway as a potential target for treatment of critical stage COVID-19 patients. Frontiers in Immunology 11: 598444.
  138. 138. Guhathakurata S, Saha S, Kundu S, Chakraborty A, Banerjee JS (2021) South Asian countries are less fatal concerning COVID-19: A fact-finding procedure integrating machine learning & multiple criteria decision-making (MCDM) technique. Journal of The Institution of Engineers (India): Series B 102: 1249–1263.
  139. 139. Reiken S, Sittenfeld L, Dridi H, Liu Y, Liu X, et al. (2022) Alzheimer’s-like signaling in brains of COVID-19 patients. Alzheimer’s & Dementia 18: 955–965. pmid:35112786