Next Article in Journal
Frequency and Clinical Significance of Elevated IgG4 in Rheumatoid Arthritis: A Systematic Review
Next Article in Special Issue
Mitomycin C in Homologous Recombination Deficient Metastatic Pancreatic Cancer after Disease Progression on Platinum-Based Chemotherapy and Olaparib
Previous Article in Journal
Different Metabolites of the Gastric Mucosa between Patients with Current Helicobacter pylori Infection, Past Infection, and No Infection History
Previous Article in Special Issue
Dynamic Interplay between Cockayne Syndrome Protein B and Poly(ADP-Ribose) Polymerase 1 during Oxidative DNA Damage Repair
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Base Excision Repair in Innate Immune Cells and Its Relevance for Cancer Therapy

1
Division of Pharmacology and Toxicology, Dell Pediatric Research Institute, College of Pharmacy, The University of Texas at Austin, 1400 Barbara Jordan Blvd. R1800, Austin, TX 78723, USA
2
Department of Cell Systems & Anatomy, University of Texas Health, San Antonio, TX 78229, USA
3
Biology and Environment Science Department, College of Art and Science, University of New Haven, 300 Boston Post Rd., West Haven, CT 06516, USA
4
Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(3), 557; https://doi.org/10.3390/biomedicines10030557
Submission received: 25 December 2021 / Revised: 18 February 2022 / Accepted: 24 February 2022 / Published: 26 February 2022
(This article belongs to the Special Issue Evolving Landscape of DNA Repair in Cancer Therapy)

Abstract

:
Innate immunity is critical for immediate recognition and elimination of invading pathogens or defense against cancer cell growth. Dysregulation of innate immune systems is associated with the pathogenesis of different types of inflammatory diseases, including cancer. In addition, the maintenance of innate immune cells’ genomic integrity is crucial for the survival of all organisms. Oxidative stress generated from innate immune cells may cause self-inflicted DNA base lesions as well as DNA damage on others neighboring cells, including cancer cells. Oxidative DNA base damage is predominantly repaired by base excision repair (BER). BER process different types of DNA base lesions that are presented in cancer and innate immune cells to maintain genomic integrity. However, mutations in BER genes lead to impaired DNA repair function and cause insufficient genomic integrity. Moreover, several studies have implicated that accumulation of DNA damage leads to chromosomal instability that likely activates the innate immune signaling. Furthermore, dysregulation of BER factors in cancer cells modulate the infiltration of innate immune cells to the tumor microenvironment. In the current review, the role of BER in cancer and innate immune cells and its impact on innate immune signaling within the tumor microenvironment is summarized. This is a special issue that focuses on DNA damage and cancer therapy to demonstrate how BER inhibitor or aberrant repair modulates innate inflammatory response and impact immunotherapy approaches. Overall, the review provides substantial evidence to understand the impact of BER in innate immune response dynamics within the current immune-based therapeutic strategy.

1. Introduction

Innate immunity is triggered in response to pathogen infection or local lesions to promote infection clearance or wound-healing processes [1,2,3]. In addition, innate immune response can be initiated by non-professional immune cells (like epithelial cells, endothelial cells, and fibroblasts) and professional antigen presenting cells (like neutrophils, macrophages, and dendritic cells) [2]. Mechanistically, the activation of innate immune response depends on pattern-recognition receptors (PRRs) that sense danger associated molecular patterns (DAMPs) or pathogen-associated molecular patterns (PAMPs) [4,5]. Pathogen-derived nucleic acids constitute a major class of PAMPs that operate in specific subcellular localizations and recognized by a vast array of PRRs [6]. Due to the abundance of nucleic acids in cells, their corresponding PRRs are evolved to be highly regulated and compartmentalized [7]. Most of the PRRs exhibit high substrate specificity to detect DAMPS [8]. Specific groups of pathogens are recognized via PRRs expressed mainly by cells of the innate immune system. For example, many viruses, bacteria, and intracellular parasites trigger type 1 immunity, with elevations in the expression of specific cytokines [9]. In contrast, multicellular pathogens, including helminths, stimulate a type 2 response, with elevations in IL-4 and IL-13 [10]. Recently published data have also shown that self-released nucleic acids derived from DNA repair deficient cells [1,11] or oxidative mitochondrial DNA damage [2,12] have been considered as DAMPs and trigger innate immune cells response. It is critical to consider both the cell lineage and the specific activation state when assessing the function of a cell of the immune system in response to a specific pathogen or stimuli.
There are different types of innate immune cells such as monocytes, macrophages, neutrophils, and dendritic cells (DC) that can be stimulated by endogenous oxidative stress or exogenous agents such as pathogen [3]. Macrophages originate either from bone marrow-derived monocytes [13,14,15] or from precursor cells derived from the yolk sac or fetal liver during development [16,17,18]. In addition, macrophages play a significant role in maintaining tissue homeostasis, defending against pathogens, and facilitating wound healing [19]. Moreover, macrophages release different types of cytokines, chemokines, and other immune factors to attract other cells to the infection site or tissue injury site [20]. Similarly, monocytes-derived DC also play critical roles in innate immunity [21,22]. DC have PRR and PAMPs, which activate Toll-like receptor (TLR) pathways, type C lectins, release proinflammatory cytokines, and stimulate the innate immune system [23]. Moreover, DCs in the periphery capture and processes antigens, expresses co-stimulatory molecules and produces cytokines, and migrates to lymphoid organs [21]. In addition, DC are equipped with robust oxidative DNA damage repair machinery to maintain its DNA integrity [24]. Neutrophils are another major innate immune cell in the peripheral blood and are produced in the bone marrow from stem cells [25,26]. Neutrophils are recruited to the tumor bed and contribute to further amplifying inflammatory response and releasing metabolites such as hydrogen peroxide that may cause reactive oxygen species (ROS) induced DNA damage [27,28]. Overall, the major common features of neutrophil, monocyte/macrophage activation is the generation of ROS and reactive nitrogen intermediates (RNI), such as O−2 and nitric oxide (NO), respectively [29,30,31]. NO is produced by inducible nitric oxide synthase (iNOS) from L-arginine and oxygen, while O−2 is catalyzed by NADPH oxidase [32,33,34]. For instance, activated macrophage induced ROS and RNI caused DNA damage, including 8-nitroguanine and 8-oxo-7,8-dihydro-2′-deoxyguanosine (8-oxodG) [35,36]. Those DNA base lesions can lead to base loss or DNA single-strand breaks (SSBs) [37,38,39,40]. Furthermore, cells harbor SSBs progress into S-phase encounter DNA-replication apparatus and convert to double-strand breaks (DSBs) [41,42]. In addition, spontaneous ROS-associated DNA base damage and DSBs can result from RNA transcription, DNA replication, and/or genotoxic stress during infection [43,44,45]. Those DNA damaged sites likely release DNA fragments into cytosolic compartment of the cells and would potentially be recognized as DAMP to trigger innate immune response [43]. Notably, a recent discovery has shown that innate immune cells such as macrophages have two important self-protective mechanisms against oxidative stress associated DNA damage/DAMP-associated innate immune cells activation, including maintaining redox balance via generating antioxidant enzymes such as superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase, and glutathione reductase (GR) [46,47]. The second mechanism is associated with inherent DNA repair pathways to process oxidative DNA damage and restore immune system homeostasis [6,48].
An overall summary of what has been so far discovered and established on DNA damage and repair in innate immune cells biology is provided within this review. In addition, we highlight what remains outstanding and which questions are still unanswered. The key findings of studies that have been carried out in oxidative DNA damage repair linking to innate inflammatory response are provided. The various aspects of BER dynamics to modulate the macrophages population that are present in high numbers in the tumor microenvironment (tumor-associated macrophage) and its role in how BER contributes to cGAS/STING inflammatory pathways is included. Finally, we outline the potential use of some of the DNA repair targeted therapy that modulates innate immune signaling and remodels the inflammatory response.

2. Base Excision Repair in Innate Immune Cells

Excessive production of ROS and RNI in innate immune cells, including neutrophils, dendritic cells, eosinophil, and macrophage, leads to oxidative-stress-related DNA base damage, DNA adducts, and SSBs, which may further result in mutations [49,50,51]. BER is the main repair pathway against oxidized DNA bases to maintain genomic integrity [49,52,53,54,55]. Mammalian cells harbor two sub-BER pathways that are dependent on the size of the oxidized DNA base they process and the key enzyme involved in the repair process [56]. The two sub-pathways are known as short-patch BER (SP-BER) and long-patch BER (LP-BER) [57,58]. Short-patch BER engages in repairing one nucleotide gap [44,45], while the long-patch BER involves processing and repairing 2–12 nucleotides gaps. Both BER mechanisms are initiated by DNA glycosylase that recognizes and removes the DNA base lesion. In SP-BER, AP-endonuclease 1 (APE1) cleaved the DNA backbone to generate a 3′-OH terminus at the damage site, followed by DNA polymerase beta adding one nucleotide and the nick sealed by a DNA ligase in a complex with XRCC1 [59]. In comparison, LP-BER utilizes both DNA polymerase beta and other DNA replication enzymes, such as DNA Pol δ and DNA Pol ε, to conduct strand-displacement DNA synthesis. The displaced single-stranded DNA structure or 5′-DNA flap off-load by flap endonuclease I (FEN1) [60] followed by sealing of the DNA nicks by Ligase I or Ligase III [61].
Several studies have shown that neutrophils and monocytes are hypersensitive to accumulated DNA damage derived from oxidative stress due to their insufficient BER function [24,62]. The unrepaired DNA base damage derived from oxidative stress and accumulated BER intermediates in monocytes and triggers DNA damage response (DDR) to halt the cell cycle or induce cell death [50]. Low BER and DSB repair activity observed in neutrophils and monocytes are results from under-expression of BER proteins, including XRCC1, ligase IIIα, ligase I, poly(ADP-ribose) polymerase-1 (PARP1), and repair proteins [25,37,63]. In addition, ROS induces DNA single- and double-strand breaks, leading to activation of the DDR pathways that trigger monocytes apoptosis, whereas macrophages and DCs are resistant to ROS-associated DNA damage and apoptotic cell death [12,24]. In addition, eosinophils harbor efficient DNA repair activity, including BER [62]. Overall, these observations are supported by the presence of a sufficient level of BER proteins, including XRCC1, Ligase I, and DNA polymerase beta (Figure 1) in macrophages, but not in monocytes [50]. In accordance with this, the relevant evidence on how BER factors are also involved in macrophage plasticity is discussed in the next section.

3. BER Modulates DNA Damage Induced Innate Immune Inflammatory Response

Several studies have shown that oxidative DNA damage initiates an innate immune inflammatory response via activation of a variety of transcription factors, such as nuclear factor kappa B (NF-κB), STAT, and interferon regulatory factors [51,64,65]. BER function is critical to protect the cells from genomic instability and/or inflammation [37,66]. In particular, BER is expressed in selected types of innate immune cells that may regulate the innate immune response [67]. Few studies highlight the role of oxidative DNA damage repair in innate immune cells; however, detailed mechanistic insight into how loss-of-function BER drives or contributes to DNA-damage-induced inflammatory response remains unknown. The Cancer Genome Atlas (TCGA) data set have shown that somatic BER gene mutations are found in 30% of tumors, and other also indicated that germline variant of BER genes (POLB, APEX1, MUTYH, XRCC1 OGG1, TDG) leads to compromised repair capacity and increases the risk of inflammatory associated diseases, including cancer [68]. Due to impaired function of oxidized DNA damage repair and/or increased accumulation of ROS induced DNA base damage that can trigger DNA damage response (DDR), including activation of Ataxia-telangiectasia mutated (ATM) kinase, ATR, and stimulate nuclear factor kappa B (NF-κB) transcription factors to drive the expression of proinflammatory genes [51,69,70,71]. Moreover, some of the BER proteins such as OGG1 and APE1 can act as transcription factors and are involved in epigenetic regulation of expression of inflammatory cytokines mediated by NF-κB to foster inflammatory response [65,72]. In contrast, BER deficiency (e.g., OGG1) suppresses the expression of proinflammatory genes [72,73,74,75]. However, in some cases, loss of OGG1 function enhances inflammatory response to mitigate bacterial infection [63]. Excessive level of unrepaired DNA damage in the nucleus- and factors-associated DNA end resection during double-strand break repair, including the Bloom syndrome (BLM) helicase and exonuclease 1 (EXO1), plays a major role in generating DNA fragments, and the cytoplasmic 3′–5′ exonuclease Trex1 is required for their degradation. However, the lack of DNA degrading nucleases enzyme in the cytosol likely contributes to increasing cytosolic DNA [43,76]. The cytosolic damaged DNA is recognized by the innate immune system mediated by different types of receptors or DNA sensors [77,78], which subsequently triggers inflammatory immune response [79,80,81]. Moreover, DNA fragments released from apoptotic cells [82,83] can activate innate immune cells in a Toll-like receptor 9 (TLR9) [84] and cGAS/STING dependent fashion [2].
Multiple studies have shown that BER prevents DNA damage derived from oxidative stress in innate immune cells and modulates the inflammatory response [73]. BER deficiency leads to accumulated intrinsic oxidized DNA bases, BER intermediates (AP sites, SSBs), and DNA-replication-associated DSBs [85]. Alternatively, DNA damage derived from oxidative stress leads to activation of DNA damage response (DDR) and modulate inflammatory response [67,86]. DDR signaling enhances the DNA repair capacity of the cells by inducing DNA repair transcriptionally or post-translation modification to counter DNA damage [87]. However, several BER knockout mouse models are found to develop inflammatory associated diseases, including cancer and autoimmunity [1,88,89]. For instance, mice are susceptible to inflammation when they harbor aberrant DNA glycosylases function (e.g., OGG1, MUTYH, NEIL2, and NEIL3) since they are unable to remove oxidized DNA bases [66,90,91,92,93,94]. Moreover, mouse carrying mutation in dRP lyase domain of DNA polymerase beta displays higher innate immune inflammatory response [37]. Another study found that a nuclease-deficient Flap endonuclease-1 (FEN1) mutant mouse model accumulates apoptotic-associated cytosolic DNA that forms complex with IgG to promote chronic inflammation [95,96]. However, it is unknown whether the cytosolic DNA in BER-deficient cells is predominantly generated from DNA damage product from nuclei or mitochondrial DNA that likely activates innate immune signaling and promotes the inflammatory response. Recent work has implicated chromosol instability contribute to inducing innate immune inflammatory pathways [97]. Detection of DNA in the cytoplasmic compartment of the cells induces a type I interferons (IFNs) response to amplify the innate immune signaling-based inflammatory response [89]. DNA leakage from damaged nuclear and/or mitochondrial DNA has the potential to induce innate immune signaling through binding to a cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS) DNA sensor [98]. These products catalyze the production of cyclic GMP-AMP (cGAMP) and further lead to the activation of stimulators of interferon genes (STING) [98]. Previously, other studies have shown that cGAS and STING have been identified as intracellular DNA sensors that activate the interferon pathway in response to virus infection and intercellular pathogens [81,99]. STING is a signaling molecule, often located at the endoplasmic reticulum (ER), and it is essential for controlling the induction of type 1 interferons (IFN) and inflammatory mediators [100,101,102]. In addition, interferon-inducible protein 16 (IFN16) binds to dsDNA and promotes STING dependent activity [100,103,104] of TBK1 phosphorylation that subsequently leads to activation of transcription factors interferon regulatory factor 3 (IRF3) and Nuclear factor-kappa B (NF-kB), which triggers innate immune gene transcription [76,88,89,105,106]. In particular, macrophages harbor robust DNA-sensing activity [107]. Macrophage has the ability to activate intrinsic cGAS/STING pathways due to persistence of endogenous oxidative stress-associated DNA base damage and promote proinflammatory response [108]. Alternatively, dsDNA released from oxidative stress-associated DNA damage product may also trigger activation of cGAS/STING a (Figure 2). Moreover, exposure to exogenous DNA damaging agents such as radiation and chemotherapy (e.g., paclitaxel, BSO) triggers the death of tumor cells and the production of oxidized DNA base damage, which activates innate immune cells, including macrophage and dendritic cells via the STING-dependent signaling pathway [43,109].

4. Role of BER in Macrophage Plasticity

As an essential component of innate immunity, macrophages can inhibit or promote cell proliferation and tissue repair [110]. Macrophages originate from peripheral blood monocytes and harbor heterogeneous phenotypes [17]. Moreover, macrophages can adjust their phenotype in response to various extracellular signals [111,112]. M1 macrophages secrete high levels of proinflammatory cytokines and increased concentrations of superoxide anions (O2), oxygen radicals, and nitrogen radicals to increase their cytotoxicity activity, as required in early phases of tissue repair [113]. Conversely, M2 macrophages produce an extracellular matrix and reduce proinflammatory cytokine levels to promote tissue repair [100]. More specifically, tumor-associated macrophages (TAMs) account for more than 50% of tumor-infiltrating cells in the tumor microenvironment (TME) [114,115]. TAM is derived from two different sources including tissue-resident macrophage and macrophage derived from monocytes [101,102]. TAM is the most abundant cell type among the tumor-promoting stromal cells in solid tumors [103,104]. TME consists of cytokines/chemokines produced by tumor cells or stroma cells, and it is critical to shaping the macrophage functional plasticity [116,117,118]. In particular, macrophage chemoattractant molecules [e.g., C-C Motif Chemokine Ligand 2 (CCL2)/monocyte chemoattractant protein 1 (MCP-1), colony-stimulating factor 1 (CSF-1)] play a major role to recruit TAMs into the tumor microenvironment. TAMs are categorized into M1 and M2 phenotypes [119]. The switch between M1 (anti-tumorigenesis) and M2 (pro-tumorigenesis) is known as “macrophage polarization” [120]. The relationship between TAMs and cancer cells has been studied intensively [121,122], and it is well established that TAMs play a major role in tumor development and progression [123,124]. Several studies have shown that increased TAM infiltration negatively correlated with the overall survival of cancer patients [125,126,127]. TAMs promote tumor progression via enhancing cancer cell genetic instability, facilitating cancer stem cells replication and survival, supporting metastasis, influencing adaptive immunity response, and promoting chemoresistance [128]. Specifically, M2 macrophages play key roles in cancer development by promoting angiogenesis in tumors and accelerating metastasis, as well as fostering resistance to different cancer therapy [128,129,130]. In addition, M2 macrophages exhibit an anti-inflammatory response to promote tumor progression, while M1 macrophages support proinflammation response hostile to tumor cells. In contrast, finding from several preclinical studies demonstrated that infiltration of M1 in the tumor microenvironment is associated with better overall survival of patients [126].
Genetic variants of BER genes may result in differences in immune system components, including the abundance and activation states of immune-cell types, the expression of immunomodulatory molecules, and regulation of immune-related genes [131]. Several studies have identified somatic and germline variants of BER genes and have provided mechanistic insight their impact in tumor initiation [132,133,134,135,136]. Germline genetic variation in genes that code for DNA repair may influence an individual’s innate immune function. Moreover, recent studies have shown that germline may influence which mutations are selected for in the growing tumor [137,138]. In contrast, few studies have shown that macrophage carrying DNA repair defect accumulates DNA damage that derives metabolic reprogramming and eventually leads to chronic inflammation [139]. However, it is unknown that human germline variant of BER genes impacts macrophage functional plasticity. In comparison, altered BER function in cancer cells may contribute to unbalanced recruitment of macrophages in the tumor microenvironment. There are several mouse models established with altered BER functions to understand mechanisms of inflammation-associated cancer pathogenesis. For example, MUTYH knockout mice demonstrated that BER has a capacity to modulate inflammatory response via macrophages infiltration in the tumor microenvironment [103]. Furthermore, mouse models studies provide mechanistic insight into how TAMs are recruited to primary tumors and metastatic tumors [140]. In line with this, a high number of infiltrations of M2 macrophage in the TME is associated with poor overall survival of patient and worse disease outcome [141,142]. In addition, BER genes, including MPG and NEIL2, are overexpressed and have a positive correlation with the infiltration of M2 macrophage in the tumor microenvironment (TIMER 2.0, CIBERSORT) (Figure 3). However, experimental evidence has shown that increased TAM infiltration enhances the DNA repair capacity of the tumor cells and fosters low treatment response [143]. Furthermore, treating tumor with oxidative DNA damaging chemotherapeutic drug, such as Paclitaxel, promotes the rewire of TAMs toward tumoricidal phenotypes through activation of TLR4 [144].
Different strategies to target TAMs have been proposed including depleting and/or reprogramming TAMs to improve clinically approved chemotherapeutic drugs response [145,146]. Therefore, using TAM as a therapeutic target in the tumor microenvironment will likely provide a novel approach to prevent the progression of solid tumors and prevent metastatic disease [128]. One of the therapeutic strategies focused on targeting BER genes such as APE1 and PARP1 promotes attracting antitumor macrophage in tumors microenvironment and enhancing antitumor immune response [103,119,147,148,149]. In particular, targeting the Colony Stimulating Factor1 (CSF1)–Colony Stimulating Factor1 Receptor (CSF-1R) axis has received significant consideration for DNA repair targeted therapeutic purpose to overcome immunosuppressive macrophages [150]. CSF1 (also known as M-CSF) plays a critical role in promoting the differentiation of monocyte to macrophage lineages [151]. CSF1 is abundantly expressed by several tumor types; this ligand-receptor pathway has been extensively investigated in tumor models and constitutes a paradigm of TAM-cancer cell interaction [1,152]. Moreover, DNA damaging agents such as radiation therapy induce the expression of CSF1, which results in CSFR1 dependent infiltration of immunosuppressive macrophages to the TME [153,154]. CSF-1R is expressed by macrophages and provides a potential target to eliminate TAM. Overexpression of CSF-1, the major lineage regulator for macrophages, is associated with poor prognosis in breast, ovarian, endometrial, prostate, hepatocellular, and colorectal cancer [15], as the intra-tumoral presence of CSF1R+ macrophages correlates with poor survival in various tumor types [127,155]. Moreover, our in silico analysis shows that overexpression of CSF1 and CSFR1 is positively correlated in tumors that harbor high expression of DNA repair genes such as APEX1 and PARP-1 (TCGA, TIMER 2.0, CIBERSORT). In line with this, exploiting the CSF1–CSF1-R axis of macrophage and DNA repair targeted therapy may likely provide an alternative platform to enhance therapeutic efficacy and may improve overall patient survival.

5. Targeting PARP1 Modulate Macrophage Mediated Tumor Inflammatory Response

Poly(ADP-ribose) polymerase-1 (PARP1) contributes to many physiological processes, including DNA replication and transcription, DNA repair, regulation of cell differentiation, and apoptosis [156]. PARP1 plays a significant role in BER pathway to repair SSBs [157]. In addition, PARP1 is also critical to repair replication-associated DNA damage during DNA synthesis [157,158,159]. Apart from its primary role in genome integrity maintenance, PARP1 appears to protect M1 macrophages from oxidative-stress-related cell death through transcriptionally regulating antioxidants such as MnSOD (SOD2), glutathione reductase, and thioredoxin reductase [160,161]. In addition, the level of PARP1 is reduced when macrophage is stimulated with lipopolysaccharide (LPS) and polarized to pro-M1 phenotype [160]. Further, PARP1 modulates macrophage polarization via inflammatory mediators such as high-mobility group box protein 1 (HMGB1) [162]. HMGB1 is a non-histone nuclear proteins and very potent inflammatory mediator [144,163]. HMGB1 is secreted by activated innate immune cells, including monocytes and macrophages [164], and is also released by necrotic cells [165,166]. In the setting of immune challenge, PARP1 PARylates HMGB1 promotes its acetylation, detaches from chromatin, and thus allows the translocation of HMGB1 from the nucleus to the cytoplasm [164,167]. Subsequently, the cytoplasmic HMGB1 can be secreted into the extracellular space as DAMP to induce inflammatory mediator such as cytokine and chemokine activities [168]. Moreover, HMGB1 interacts with the receptor for advanced glycation products (RAGE) and leads to macrophage polarization into M1 phenotype [169], whereas HMGB1 interacts with C1q complement to induce M2 polarization [170]. Conversely, the TCGA dataset shows that expression of PARP1 in tumors negatively correlated with type I interferon gene expression (CCL5, IFNβ, ISG15). The recent results reviewed in this article indicate that PARP1 inhibitors, such as Olaparib treatment in cancer cells, reduce polarization of adjacent macrophage to M2 phenotype via accumulation of unrepaired DNA damage and BER intermediates [150]. Moreover, PARP1 inhibition is associated with increased levels of cytosolic DNA, which can trigger cGAS-STING pathways to activate IRF3 and NF-κB dependent expression of several genes that mediate innate immune response [171,172]. This includes expression of type 1 interferons and T-cell-recruiting chemokines [C-C Motif Chemokine Ligand 5 (CCL5), C-X-C Motif Chemokine Ligand 10 (CXCL10)], leading to a higher level of tumor-infiltrating T-cells [171]. Additionally, PARP1′s role in sensing DNA damage mediates a non-canonical pathway of STING activation [173]. Upon binding DSBs, PARP1 recruits and activates ATM, which subsequently activates the ubiquitin ligase TRAF6 to translocate to the cytosol and interact with IRF16, which results in STING activation [174]. This alternative pathway stimulates STING-dependent activation of the transcription factor NF-κB to generate inflammatory response [175]. It is possible that PARP1 inhibitor exacerbates accumulation of unrepaired DNA damage in cells that harbor pathogenic germline variant of BER that caused impaired BER function (Figure 4). Overall, targeting PARP1 will likely stimulate cGAS/STING dependent on the innate immune signaling and enhance antitumor immune response that potentially contributes to maximizing immunotherapy treatment efficacy (Figure 4 labeled with blue).

6. Targeting APE1 Modulate Macrophage Mediated Tumor Inflammatory Response

Apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a multifunctional enzyme participating in both oxidative DNA damage repair and redox signaling in cancer [176,177]. APE1 enhances the affinity of transcription factors (e.g., activator protein 1 [AP-1], nuclear factor-κappa B [NF-κB], p53, and others) binding to DNA to modulate inflammatory response [178]. APE1 is overexpressed in several types of cancer and alters treatment outcomes [149,178,179,180,181]. Previous studies have shown that APE1 regulates innate inflammatory response in macrophages [182]. APE1 overexpression in tumors is associated with poor prognosis in cancer patients and upregulation of immunosuppressive expression of genes [e.g., program death ligand-1 (PD-L1)] [183]. In addition, APE1 overexpressed tumor positively correlates with pro-tumor M2 macrophage infiltration and is associated with poor overall survival (CIBERSORT, TIMER 2.0). In line with this, the release of inflammatory cytokines/chemokine by M2 macrophage played a major role in cancer promotion and progression [121], whereas anti-inflammatory drugs reduce the risk of cancer [184]. Several studies have suggested the potential of APE1 as a therapeutic target in cancer [178]. However, little has been investigated on the role of APE1 in regulating innate immune cells mediated antitumor inflammatory response. Furthermore, APE1 expression in tumors negatively correlated with Type I interferon genes expression such as IFNB, CCL5 expression (TCGA) implicated that its overexpression contributes to suppressing the antitumor immune response. However, a limited number of studies have shown that targeting APE1 with inhibitor (E3330) reduces macrophage-mediated NF-κB and AP-1 signaling, which leads to low expression of inflammatory mediators such as TNF-α, IL-6, IL-12, NO, and PGE2 [185,186,187]. That experimental evidence suggests that targeting APE1 (E3330) in cancer and innate immune cells likely provides an alternative therapeutic strategy to stimulate antitumor immunity and may also increase anti-PD-L1 therapeutic response in cancer (Figure 4, labeled with purple).

7. Exploiting BER as Innate Immune Modulator for Cancer Therapy

BER modulates the immune response in inflammation-associated human disease [188,189]. Inhibition of the BER pathway promotes the accumulation of oxidative-stress-related DNA damage in an acidic TME [190]. There are different types of immune-based therapy for cancer; however, the therapeutic efficacy depends on the interaction of cancer cells with different components of the TME, including the DNA repair capacity, the density of infiltration of tumor-associated macrophage, and the status of innate immune signaling. In the next section of this review, we discuss the potential contribution of some of the tumor-associated functions of DNA repair proteins and their role in the context of immune-based therapy. We discuss the two important scenarios that are associated with defective DNA damage repair and activation of immune response to enhance immune-based therapy.
(i) Exploiting BER defect associated with reactivation of cGAS/STING in TME. Several types of cancer cells are found to silence their intrinsic cGAS-STING signaling pathways in the TME by epigenetic hypermethylation [191]. Loss of cGAS-STING signaling facilitates tumor cells to escape immune surveillance, thereby promoting carcinogenesis and resistance towards immunotherapy. Further studies in mouse models have shown that loss of cGAS-STING pathway causes low expression of type I IFN genes, which leads to a low number of infiltrating CD3+ CD8+ T cells [192]. Recent evidence emerged on activating “cold tumor” to “hot tumor” through the restoration of cGAS/STING pathway to enhance immune-based therapy response. One viable strategy is to target the BER pathway. For example, PARP1 inhibitor impairs DNA repair capacity and enhances the accumulation of PARP1-DNA complex trapping and further triggers the release of cytosolic DNA for activation of cGAS/STING [162,193]. There have been multiple attempts in developing a number of synthetic STING agonists, including Cyclic dinucleotides (CDNs) (e.g., MK-1454) and non-CDN small molecule compounds (MK2118, SR717, TK676, MSA-1, and MSA-2) to restore activation of cGAS/STING pathways [194]. Further, divalent metal ion such as manganese ion plays an important role during oxidative stress and act asco-factor for many DNA repair enzymes, including in BER factors for activation of cGAS/STING signaling pathways. A newly discovered mechanism suggests manganese released from mitochondria to the cytosol activates cGAS/STING pathways [195] and enhances immunotherapy response [196].
(ii) Inhibition of BER factors enhances program cell death ligand-1 (PD-L1) expression in cancer cells. Cancer-specific BER defects are abundant in malignant tissues [197,198,199,200]. Immune checkpoint therapy has recently emerged as a promising next-generation cancer treatment. Several studies have shown that the PD-L1 level of expression in tumors is an important factor to influence the therapeutic efficacy of response of cancer patients [201,202]. Interestingly, traditional chemotherapeutic agents inducing DNA damage have been found to upregulate the expression of PD-L1 in many cancer types [203]. Furthermore, emerging evidence suggests that defects in DNA repair machinery lead to upregulation of PD-L1 [204]. Recently, Permata et al. showed that BER gene expressions are negatively correlated with PD-L1 expression in tumors and oxidative DNA damaging agents exacerbate the expression of PD-L1 [205]. Loss of BER that enhances the accumulation of SSBs and progress into the S phase of the cell cycle likely causes replication-associated DSBs in cancer cells [85,203]. BER defects/low expression show high microsatellite instability increased neoantigen production and PD-L1 expression in tumors [206]. In the future, exploring BER deficiency and oxidative stress DNA damage associated upregulation of PD-L1expression in tumors will likely provide an additional immune biomarker to introduce the immune-based therapeutic strategy.

8. Conclusions and Future Direction

In this review article, we presented a new direction to highlight the cross talk of DNA repair capacity of cancer cells and innate immune cells phenotypes. The overall mechanistic insight into the DNA repair capacity of cancer cells and innate immune cells can provide a fundamental framework to develop a therapeutic strategy that considers the innate immune cells’ response in the tumor microenvironment. Even though several BER germline and tumor-associated mutations were uncovered from different studies [68,135,198,200], their functional impact on innate immune cells such as macrophage and dendritic cells is unknown. In particular, macrophages phenotypes and functional plasticity are influenced by several factors, including oxidative DNA damage and repair capacity. Based on our in silico analysis, the positive correlation of overexpression of BER genes in tumor cell versus infiltration of M2 macrophage impacts the overall survival of patients. Within this context, and similar to other studies, we have shown that reprogramming M2 macrophage into an M1-like phenotype is a potential cancer therapeutic strategy [207,208]. Further, our comprehensive review can inform future studies to consider BER pathways as a potential target to modulate the innate immune signaling and promote the inflammatory response. The future may uncover potential mechanistic insight into how the DNA repair pathway modulates the innate immune response in terms of reprogramming the tumor microenvironment, restoring antitumor immunities, and enhancing cancer immunotherapy treatment response.

Author Contributions

S.Z. wrote part of the manuscript and conducted data extraction from the TCGA database. D.K. wrote the manuscript and conceptualized the overall connection of BER and immune cells dynamics. S.L.H. wrote the manuscript and provided comments. A.G.S. contributed during writing the manuscript and provided comments. M.S. provided read the manuscript and comments. All authors have read and agreed to the published version of the manuscript.

Funding

DK is supported by the National Institute of Health/National Cancer Center (R21CA 249346-01A1.) and AGA-R. Robert and Sally Funderburg of America Gastroenterology Association. The funding agencies or institutes had no role in study design, data collection and analysis, or preparation of the manuscript.

Acknowledgments

This work is supported by United States National Institutes of Health (NIH/National Cancer Institute (NCI)) grant award R21CA 249346-01A1.) and AGA-R. Robert and Sally Funderburg of America Gastroenterology Association award to cover the over all cost of publication and other related expense.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Mantovani, A.; Sica, A. Macrophages, innate immunity and cancer: Balance, tolerance, and diversity. Curr. Opin. Immunol. 2010, 22, 231–237. [Google Scholar] [CrossRef] [PubMed]
  2. Medzhitov, R.; Janeway, C., Jr. Innate immunity. N. Engl. J. Med. 2000, 343, 338–344. [Google Scholar] [CrossRef]
  3. Koenderman, L.; Buurman, W.; Daha, M.R. The innate immune response. Immunol. Lett. 2014, 162, 95–102. [Google Scholar] [CrossRef]
  4. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Medzhitov, R.; Janeway, C., Jr. Innate immune recognition: Mechanisms and pathways. Immunol. Rev. 2000, 173, 89–97. [Google Scholar] [CrossRef]
  6. Matzinger, P. The danger model: A renewed sense of self. Science 2002, 296, 301–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Barton, G.M.; Kagan, J.C. A cell biological view of Toll-like receptor function: Regulation through compartmentalization. Nat. Rev. Immunol. 2009, 9, 535–542. [Google Scholar] [CrossRef]
  8. Chuenchor, W.; Jin, T.; Ravilious, G.; Xiao, T.S. Structures of pattern recognition receptors reveal molecular mechanisms of autoinhibition, ligand recognition and oligomerization. Curr. Opin. Immunol. 2014, 26, 14–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Thakur, A.; Mikkelsen, H.; Jungersen, G. Intracellular Pathogens: Host Immunity and Microbial Persistence Strategies. J. Immunol. Res. 2019, 2019, 1356540. [Google Scholar] [CrossRef]
  10. Gause, W.C.; Wynn, T.A.; Allen, J.E. Type 2 immunity and wound healing: Evolutionary refinement of adaptive immunity by helminths. Nat. Rev. Immunol. 2013, 13, 607–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Coquel, F.; Silva, M.J.; Techer, H.; Zadorozhny, K.; Sharma, S.; Nieminuszczy, J.; Mettling, C.; Dardillac, E.; Barthe, A.; Schmitz, A.L.; et al. SAMHD1 acts at stalled replication forks to prevent interferon induction. Nature 2018, 557, 57–61. [Google Scholar] [CrossRef]
  12. Briegert, M.; Kaina, B. Human monocytes, but not dendritic cells derived from them, are defective in base excision repair and hypersensitive to methylating agents. Cancer Res. 2007, 67, 26–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Auffray, C.; Sieweke, M.H.; Geissmann, F. Blood monocytes: Development, heterogeneity, and relationship with dendritic cells. Annu. Rev. Immunol. 2009, 27, 669–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gordon, S. Macrophage neutral proteinases and chronic inflammation. Ann. N. Y. Acad. Sci. 1976, 278, 176–189. [Google Scholar] [CrossRef]
  15. Pollard, J.W. Trophic macrophages in development and disease. Nat. Rev. Immunol. 2009, 9, 259–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Yona, S.; Kim, K.W.; Wolf, Y.; Mildner, A.; Varol, D.; Breker, M.; Strauss-Ayali, D.; Viukov, S.; Guilliams, M.; Misharin, A.; et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 2013, 38, 79–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Gordon, S.; Taylor, P.R. Monocyte and macrophage heterogeneity. Nat. Rev. Immunol. 2005, 5, 953–964. [Google Scholar] [CrossRef] [PubMed]
  18. Raggatt, L.J.; Wullschleger, M.E.; Alexander, K.A.; Wu, A.C.; Millard, S.M.; Kaur, S.; Maugham, M.L.; Gregory, L.S.; Steck, R.; Pettit, A.R. Fracture healing via periosteal callus formation requires macrophages for both initiation and progression of early endochondral ossification. Am. J. Pathol. 2014, 184, 3192–3204. [Google Scholar] [CrossRef] [PubMed]
  19. Minutti, C.M.; Knipper, J.A.; Allen, J.E.; Zaiss, D.M. Tissue-specific contribution of macrophages to wound healing. Semin. Cell Dev. Biol. 2017, 61, 3–11. [Google Scholar] [CrossRef] [Green Version]
  20. Turner, M.D.; Nedjai, B.; Hurst, T.; Pennington, D.J. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochim. Biophys. Acta 2014, 1843, 2563–2582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef] [PubMed]
  22. Schraml, B.U.; Reis e Sousa, C. Defining dendritic cells. Curr. Opin. Immunol. 2015, 32, 13–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tan, Y.F.; Leong, C.F.; Cheong, S.K. Observation of dendritic cell morphology under light, phase-contrast or confocal laser scanning microscopy. Malays. J. Pathol. 2010, 32, 97–102. [Google Scholar] [PubMed]
  24. Bauer, M.; Goldstein, M.; Christmann, M.; Becker, H.; Heylmann, D.; Kaina, B. Human monocytes are severely impaired in base and DNA double-strand break repair that renders them vulnerable to oxidative stress. Proc. Natl. Acad. Sci. USA 2011, 108, 21105–21110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mayadas, T.N.; Cullere, X.; Lowell, C.A. The multifaceted functions of neutrophils. Annu. Rev. Pathol. 2014, 9, 181–218. [Google Scholar] [CrossRef] [Green Version]
  26. Borregaard, N. Neutrophils, from marrow to microbes. Immunity 2010, 33, 657–670. [Google Scholar] [CrossRef] [Green Version]
  27. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef] [PubMed]
  28. Ueha, S.; Shand, F.H.; Matsushima, K. Myeloid cell population dynamics in healthy and tumor-bearing mice. Int. Immunopharmacol. 2011, 11, 783–788. [Google Scholar] [CrossRef]
  29. Durackova, Z. Some current insights into oxidative stress. Physiol. Res. 2010, 59, 459–469. [Google Scholar] [CrossRef]
  30. Nathan, C.; Shiloh, M.U. Reactive oxygen and nitrogen intermediates in the relationship between mammalian hosts and microbial pathogens. Proc. Natl. Acad. Sci. USA 2000, 97, 8841–8848. [Google Scholar] [CrossRef] [Green Version]
  31. Kawanishi, S.; Hiraku, Y.; Pinlaor, S.; Ma, N. Oxidative and nitrative DNA damage in animals and patients with inflammatory diseases in relation to inflammation-related carcinogenesis. Biol. Chem. 2006, 387, 365–372. [Google Scholar] [CrossRef]
  32. Ischiropoulos, H.; Zhu, L.; Beckman, J.S. Peroxynitrite formation from macrophage-derived nitric oxide. Arch. Biochem. Biophys. 1992, 298, 446–451. [Google Scholar] [CrossRef]
  33. Zingarelli, B.; O’Connor, M.; Wong, H.; Salzman, A.L.; Szabo, C. Peroxynitrite-mediated DNA strand breakage activates poly-adenosine diphosphate ribosyl synthetase and causes cellular energy depletion in macrophages stimulated with bacterial lipopolysaccharide. J. Immunol. 1996, 156, 350–358. [Google Scholar]
  34. Xia, Y.; Zweier, J.L. Superoxide and peroxynitrite generation from inducible nitric oxide synthase in macrophages. Proc. Natl. Acad. Sci. USA 1997, 94, 6954–6958. [Google Scholar] [CrossRef] [Green Version]
  35. Ponath, V.; Kaina, B. Death of Monocytes through Oxidative Burst of Macrophages and Neutrophils: Killing in Trans. PLoS ONE 2017, 12, e0170347. [Google Scholar] [CrossRef]
  36. Ohshima, H.; Sawa, T.; Akaike, T. 8-nitroguanine, a product of nitrative DNA damage caused by reactive nitrogen species: Formation, occurrence, and implications in inflammation and carcinogenesis. Antioxid. Redox Signal. 2006, 8, 1033–1045. [Google Scholar] [CrossRef] [PubMed]
  37. Zhao, S.; Klattenhoff, A.W.; Thakur, M.; Sebastian, M.; Kidane, D. Mutation in DNA Polymerase Beta Causes Spontaneous Chromosomal Instability and Inflammation-Associated Carcinogenesis in Mice. Cancers 2019, 11, 1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. David, S.S.; O’Shea, V.L.; Kundu, S. Base-excision repair of oxidative DNA damage. Nature 2007, 447, 941–950. [Google Scholar] [CrossRef] [Green Version]
  39. Hegde, M.L.; Hazra, T.K.; Mitra, S. Early steps in the DNA base excision/single-strand interruption repair pathway in mammalian cells. Cell Res. 2008, 18, 27–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Yang, N.; Chaudhry, M.A.; Wallace, S.S. Base excision repair by hNTH1 and hOGG1: A two edged sword in the processing of DNA damage in gamma-irradiated human cells. DNA Repair 2006, 5, 43–51. [Google Scholar] [CrossRef]
  41. Eccles, L.J.; O’Neill, P.; Lomax, M.E. Delayed repair of radiation induced clustered DNA damage: Friend or foe? Mutat. Res. 2011, 711, 134–141. [Google Scholar] [CrossRef] [PubMed]
  42. Sage, E.; Harrison, L. Clustered DNA lesion repair in eukaryotes: Relevance to mutagenesis and cell survival. Mutat. Res. 2011, 711, 123–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gehrke, N.; Mertens, C.; Zillinger, T.; Wenzel, J.; Bald, T.; Zahn, S.; Tuting, T.; Hartmann, G.; Barchet, W. Oxidative damage of DNA confers resistance to cytosolic nuclease TREX1 degradation and potentiates STING-dependent immune sensing. Immunity 2013, 39, 482–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kubota, Y.; Nash, R.A.; Klungland, A.; Schar, P.; Barnes, D.E.; Lindahl, T. Reconstitution of DNA base excision-repair with purified human proteins: Interaction between DNA polymerase beta and the XRCC1 protein. EMBO J. 1996, 15, 6662–6670. [Google Scholar] [CrossRef] [PubMed]
  45. Wei, Y.F.; Robins, P.; Carter, K.; Caldecott, K.; Pappin, D.J.; Yu, G.L.; Wang, R.P.; Shell, B.K.; Nash, R.A.; Schar, P.; et al. Molecular cloning and expression of human cDNAs encoding a novel DNA ligase IV and DNA ligase III, an enzyme active in DNA repair and recombination. Mol. Cell. Biol. 1995, 15, 3206–3216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Wang, P.; Geng, J.; Gao, J.; Zhao, H.; Li, J.; Shi, Y.; Yang, B.; Xiao, C.; Linghu, Y.; Sun, X.; et al. Macrophage achieves self-protection against oxidative stress-induced ageing through the Mst-Nrf2 axis. Nat. Commun. 2019, 10, 755. [Google Scholar] [CrossRef] [PubMed]
  47. Tan, H.Y.; Wang, N.; Li, S.; Hong, M.; Wang, X.; Feng, Y. The Reactive Oxygen Species in Macrophage Polarization: Reflecting Its Dual Role in Progression and Treatment of Human Diseases. Oxid. Med. Cell Longev. 2016, 2016, 2795090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Gallucci, S.; Matzinger, P. Danger signals: SOS to the immune system. Curr. Opin. Immunol. 2001, 13, 114–119. [Google Scholar] [CrossRef]
  49. Krokan, H.E.; Nilsen, H.; Skorpen, F.; Otterlei, M.; Slupphaug, G. Base excision repair of DNA in mammalian cells. FEBS Lett. 2000, 476, 73–77. [Google Scholar] [CrossRef]
  50. Bauer, M.; Goldstein, M.; Heylmann, D.; Kaina, B. Human monocytes undergo excessive apoptosis following temozolomide activating the ATM/ATR pathway while dendritic cells and macrophages are resistant. PLoS ONE 2012, 7, e39956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. McCool, K.W.; Miyamoto, S. DNA damage-dependent NF-kappaB activation: NEMO turns nuclear signaling inside out. Immunol. Rev. 2012, 246, 311–326. [Google Scholar] [CrossRef] [PubMed]
  52. Ames, B.N. Dietary carcinogens and anticarcinogens. Oxygen radicals and degenerative diseases. Science 1983, 221, 1256–1264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Barnes, D.E.; Lindahl, T. Repair and genetic consequences of endogenous DNA base damage in mammalian cells. Annu. Rev. Genet. 2004, 38, 445–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Lindahl, T. Instability and decay of the primary structure of DNA. Nature 1993, 362, 709–715. [Google Scholar] [CrossRef] [PubMed]
  55. Dianov, G.L.; Souza-Pinto, N.; Nyaga, S.G.; Thybo, T.; Stevnsner, T.; Bohr, V.A. Base excision repair in nuclear and mitochondrial DNA. Prog. Nucleic Acid Res. Mol. Biol. 2001, 68, 285–297. [Google Scholar] [CrossRef]
  56. Wilson, S.H. Mammalian base excision repair and DNA polymerase beta. Mutat. Res. 1998, 407, 203–215. [Google Scholar] [CrossRef]
  57. Beard, W.A.; Horton, J.K.; Prasad, R.; Wilson, S.H. Eukaryotic Base Excision Repair: New Approaches Shine Light on Mechanism. Annu. Rev. Biochem. 2019, 88, 137–162. [Google Scholar] [CrossRef] [PubMed]
  58. Fortini, P.; Dogliotti, E. Base damage and single-strand break repair: Mechanisms and functional significance of short- and long-patch repair subpathways. DNA Repair 2007, 6, 398–409. [Google Scholar] [CrossRef] [PubMed]
  59. Tomkinson, A.E.; Chen, L.; Dong, Z.; Leppard, J.B.; Levin, D.S.; Mackey, Z.B.; Motycka, T.A. Completion of base excision repair by mammalian DNA ligases. Prog. Nucleic Acid Res. Mol. Biol. 2001, 68, 151–164. [Google Scholar] [CrossRef] [PubMed]
  60. Miller, A.S.; Balakrishnan, L.; Buncher, N.A.; Opresko, P.L.; Bambara, R.A. Telomere proteins POT1, TRF1 and TRF2 augment long-patch base excision repair in vitro. Cell Cycle 2012, 11, 998–1007. [Google Scholar] [CrossRef] [Green Version]
  61. Kim, Y.-J.; Wilson, D.M., 3rd. Overview of base excision repair biochemistry. Curr. Mol. Pharmacol. 2012, 5, 3–13. [Google Scholar] [CrossRef] [PubMed]
  62. Salati, S.; Bianchi, E.; Zini, R.; Tenedini, E.; Quaglino, D.; Manfredini, R.; Ferrari, S. Eosinophils, but not neutrophils, exhibit an efficient DNA repair machinery and high nucleolar activity. Haematologica 2007, 92, 1311–1318. [Google Scholar] [CrossRef] [Green Version]
  63. Qin, S.; Lin, P.; Wu, Q.; Pu, Q.; Zhou, C.; Wang, B.; Gao, P.; Wang, Z.; Gao, A.; Overby, M.; et al. Small-Molecule Inhibitor of 8-Oxoguanine DNA Glycosylase 1 Regulates Inflammatory Responses during Pseudomonas aeruginosa Infection. J. Immunol. 2020, 205, 2231–2242. [Google Scholar] [CrossRef] [PubMed]
  64. Li, G.; Yuan, K.; Yan, C.; Fox, J., 3rd; Gaid, M.; Breitwieser, W.; Bansal, A.K.; Zeng, H.; Gao, H.; Wu, M. 8-Oxoguanine-DNA glycosylase 1 deficiency modifies allergic airway inflammation by regulating STAT6 and IL-4 in cells and in mice. Free Radic. Biol. Med. 2012, 52, 392–401. [Google Scholar] [CrossRef] [Green Version]
  65. Pan, L.; Zhu, B.; Hao, W.; Zeng, X.; Vlahopoulos, S.A.; Hazra, T.K.; Hegde, M.L.; Radak, Z.; Bacsi, A.; Brasier, A.R.; et al. Oxidized Guanine Base Lesions Function in 8-Oxoguanine DNA Glycosylase-1-mediated Epigenetic Regulation of Nuclear Factor kappaB-driven Gene Expression. J. Biol. Chem. 2016, 291, 25553–25566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chakraborty, A.; Wakamiya, M.; Venkova-Canova, T.; Pandita, R.K.; Aguilera-Aguirre, L.; Sarker, A.H.; Singh, D.K.; Hosoki, K.; Wood, T.G.; Sharma, G.; et al. Neil2-null Mice Accumulate Oxidized DNA Bases in the Transcriptionally Active Sequences of the Genome and Are Susceptible to Innate Inflammation. J. Biol. Chem. 2015, 290, 24636–24648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Fontes, F.L.; Pinheiro, D.M.L.; Oliveira, A.H.S.d.; Oliveira, R.K.d.M.; Lajus, T.B.P.; Agnez-Lima, L.F. Role of DNA repair in host immune response and inflammation. Mutat. Res.-Rev. Mutat. Res. 2015, 763, 246–257. [Google Scholar] [CrossRef] [PubMed]
  68. Yamtich, J.; Nemec, A.A.; Keh, A.; Sweasy, J.B. A germline polymorphism of DNA polymerase beta induces genomic instability and cellular transformation. PLoS Genet. 2012, 8, e1003052. [Google Scholar] [CrossRef] [PubMed]
  69. Ting, L.; Lingyun, Z.; Donghyun, J.; Shao-Cong, S. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [Green Version]
  70. Kidane, D.; Chae, W.J.; Czochor, J.; Eckert, K.A.; Glazer, P.M.; Bothwell, A.L.M.; Sweasy, J.B. Interplay between DNA repair and inflammation, and the link to cancer. Crit. Rev. Biochem. Mol. Biol. 2014, 49, 116–139. [Google Scholar] [CrossRef]
  71. Li, N.; Banin, S.; Ouyang, H.; Li, G.C.; Courtois, G.; Shiloh, Y.; Karin, M.; Rotman, G. ATM is required for IkappaB kinase (IKKk) activation in response to DNA double strand breaks. J. Biol. Chem. 2001, 276, 8898–8903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ba, X.; Bacsi, A.; Luo, J.; Aguilera-Aguirre, L.; Zeng, X.; Radak, Z.; Brasier, A.R.; Boldogh, I. 8-oxoguanine DNA glycosylase-1 augments proinflammatory gene expression by facilitating the recruitment of site-specific transcription factors. J. Immunol. 2014, 192, 2384–2394. [Google Scholar] [CrossRef] [PubMed]
  73. Seifermann, M.; Ulges, A.; Bopp, T.; Melcea, S.; Schafer, A.; Oka, S.; Nakabeppu, Y.; Klungland, A.; Niehrs, C.; Epe, B. Role of the DNA repair glycosylase OGG1 in the activation of murine splenocytes. DNA Repair 2017, 58, 13–20. [Google Scholar] [CrossRef]
  74. Visnes, T.; Cazares-Korner, A.; Hao, W.; Wallner, O.; Masuyer, G.; Loseva, O.; Mortusewicz, O.; Wiita, E.; Sarno, A.; Manoilov, A.; et al. Small-molecule inhibitor of OGG1 suppresses proinflammatory gene expression and inflammation. Science 2018, 362, 834–839. [Google Scholar] [CrossRef] [Green Version]
  75. Mabley, J.G.; Pacher, P.; Deb, A.; Wallace, R.; Elder, R.H.; Szabo, C. Potential role for 8-oxoguanine DNA glycosylase in regulating inflammation. FASEB J. 2005, 19, 290–292. [Google Scholar] [CrossRef] [PubMed]
  76. Erdal, E.; Haider, S.; Rehwinkel, J.; Harris, A.L.; McHugh, P.J. A prosurvival DNA damage-induced cytoplasmic interferon response is mediated by end resection factors and is limited by Trex1. Genes Dev. 2017, 31, 353–369. [Google Scholar] [CrossRef] [Green Version]
  77. Soulat, D.; Bauch, A.; Stockinger, S.; Superti-Furga, G.; Decker, T. Cytoplasmic Listeria monocytogenes stimulates IFN-beta synthesis without requiring the adapter protein MAVS. FEBS Lett. 2006, 580, 2341–2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Stockinger, S.; Reutterer, B.; Schaljo, B.; Schellack, C.; Brunner, S.; Materna, T.; Yamamoto, M.; Akira, S.; Taniguchi, T.; Murray, P.J.; et al. IFN regulatory factor 3-dependent induction of type I IFNs by intracellular bacteria is mediated by a TLR- and Nod2-independent mechanism. J. Immunol. 2004, 173, 7416–7425. [Google Scholar] [CrossRef] [Green Version]
  79. Nociari, M.; Ocheretina, O.; Schoggins, J.W.; Falck-Pedersen, E. Sensing infection by adenovirus: Toll-like receptor-independent viral DNA recognition signals activation of the interferon regulatory factor 3 master regulator. J. Virol. 2007, 81, 4145–4157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Rasmussen, S.B.; Sorensen, L.N.; Malmgaard, L.; Ank, N.; Baines, J.D.; Chen, Z.J.; Paludan, S.R. Type I interferon production during herpes simplex virus infection is controlled by cell-type-specific viral recognition through Toll-like receptor 9, the mitochondrial antiviral signaling protein pathway, and novel recognition systems. J. Virol. 2007, 81, 13315–13324. [Google Scholar] [CrossRef] [Green Version]
  81. Stetson, D.B.; Medzhitov, R. Recognition of cytosolic DNA activates an IRF3-dependent innate immune response. Immunity 2006, 24, 93–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Riley, J.S.; Quarato, G.; Cloix, C.; Lopez, J.; O’Prey, J.; Pearson, M.; Chapman, J.; Sesaki, H.; Carlin, L.M.; Passos, J.F.; et al. Mitochondrial inner membrane permeabilisation enables mtDNA release during apoptosis. EMBO J. 2018, 37. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, Q.; Itagaki, K.; Hauser, C.J. Mitochondrial DNA is released by shock and activates neutrophils via p38 map kinase. Shock 2010, 34, 55–59. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Klattenhoff, A.W.; Thakur, M.; Chu, C.S.; Ray, D.; Habib, S.L.; Kidane, D. Loss of NEIL3 DNA glycosylase markedly increases replication associated double strand breaks and enhances sensitivity to ATR inhibitor in glioblastoma cells. Oncotarget 2017, 8, 112942–112958. [Google Scholar] [CrossRef] [PubMed]
  86. Kay, J.; Thadhani, E.; Samson, L.; Engelward, B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair 2019, 83. [Google Scholar] [CrossRef]
  87. Giglia-Mari, G.; Zotter, A.; Vermeulen, W. DNA damage response. Cold Spring Harb. Perspect. Biol. 2011, 3, a000745. [Google Scholar] [CrossRef]
  88. Wu, J.; Chen, Z.J. Innate Immune Sensing and Signaling of Cytosolic Nucleic Acids. Annu. Rev. Immunol. 2014, 32, 461–488. [Google Scholar] [CrossRef]
  89. Dhanwani, R.; Takahashi, M.; Sharma, S. Cytosolic sensing of immuno-stimulatory DNA, the enemy within. Curr. Opin. Immunol. 2018, 50, 82–87. [Google Scholar] [CrossRef] [Green Version]
  90. Liu, M.; Doublié, S.; Wallace, S.S. Neil3, the final frontier for the DNA glycosylases that recognize oxidative damage. Mutat. Res. 2013, 743–744, 4–11. [Google Scholar] [CrossRef] [Green Version]
  91. Zhou, J.; Liu, M.; Fleming, A.M.; Burrows, C.J.; Wallace, S.S. Neil3 and NEIL1 DNA glycosylases remove oxidative damages from quadruplex DNA and exhibit preferences for lesions in the telomeric sequence context. J. Biol. Chem. 2013, 288, 27263–27272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Massaad, M.J.; Zhou, J.; Tsuchimoto, D.; Chou, J.; Jabara, H.; Janssen, E.; Glauzy, S.; Olson, B.G.; Morbach, H.; Ohsumi, T.K.; et al. Deficiency of base excision repair enzyme NEIL3 drives increased predisposition to autoimmunity. J. Clin. Investig. 2016, 126, 4219–4236. [Google Scholar] [CrossRef]
  93. Sakumi, K.; Tominaga, Y.; Furuichi, M.; Xu, P.; Tsuzuki, T.; Sekiguchi, M.; Nakabeppu, Y. Ogg1 knockout-associated lung tumorigenesis and its suppression by Mth1 gene disruption. Cancer Res. 2003, 63, 902–905. [Google Scholar]
  94. Casorelli, I.; Pannellini, T.; De Luca, G.; Degan, P.; Chiera, F.; Iavarone, I.; Giuliani, A.; Butera, A.; Boirivant, M.; Musiani, P.; et al. The Mutyh base excision repair gene influences the inflammatory response in a mouse model of ulcerative colitis. PLoS ONE 2010, 5, e12070. [Google Scholar] [CrossRef] [PubMed]
  95. Zheng, L.; Dai, H.; Zhou, M.; Li, M.; Singh, P.; Qiu, J.; Tsark, W.; Huang, Q.; Kernstine, K.; Zhang, X.; et al. Fen1 mutations result in autoimmunity, chronic inflammation and cancers. Nat. Med. 2007, 13, 812–819. [Google Scholar] [CrossRef]
  96. He, L.; Zhang, Y.; Sun, H.; Jiang, F.; Yang, H.; Wu, H.; Zhou, T.; Hu, S.; Kathera, C.S.; Wang, X.; et al. Targeting DNA Flap Endonuclease 1 to Impede Breast Cancer Progression. EBioMedicine 2016, 14, 32–43. [Google Scholar] [CrossRef] [Green Version]
  97. Bakhoum, S.F.; Ngo, B.; Laughney, A.M.; Cavallo, J.A.; Murphy, C.J.; Ly, P.; Shah, P.; Sriram, R.K.; Watkins, T.B.K.; Taunk, N.K.; et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 2018, 553, 467–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Ishikawa, H.; Barber, G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 2008, 455, 674–678. [Google Scholar] [CrossRef]
  99. Motwani, M.; Pesiridis, S.; Fitzgerald, K.A. DNA sensing by the cGAS-STING pathway in health and disease. Nat. Rev. Genet. 2019, 20, 657–674. [Google Scholar] [CrossRef]
  100. Krzyszczyk, P.; Schloss, R.; Palmer, A.; Berthiaume, F. The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes. Front. Physiol. 2018, 9, 419. [Google Scholar] [CrossRef] [PubMed]
  101. Krishnan, V.; Schaar, B.; Tallapragada, S.; Dorigo, O. Tumor associated macrophages in gynecologic cancers. Gynecol. Oncol. 2018, 149, 205–213. [Google Scholar] [CrossRef]
  102. Gupta, V.; Yull, F.; Khabele, D. Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers 2018, 10, 366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [Green Version]
  104. Smallwood, H.S.; Lopez-Ferrer, D.; Eberlein, P.E.; Watson, D.J.; Squier, T.C. Calmodulin mediates DNA repair pathways involving H2AX in response to low-dose radiation exposure of RAW 264.7 macrophages. Chem. Res. Toxicol. 2009, 22, 460–470. [Google Scholar] [CrossRef] [PubMed]
  105. Paludan, S.R.; Bowie, A.G. Immune sensing of DNA. Immunity 2013, 38, 870–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Barber, G.N. STING: Infection, inflammation and cancer. Nat. Rev. Immunol. 2015, 15, 760–770. [Google Scholar] [CrossRef] [Green Version]
  107. Wassermann, R.; Gulen, M.F.; Sala, C.; Perin, S.G.; Lou, Y.; Rybniker, J.; Schmid-Burgk, J.L.; Schmidt, T.; Hornung, V.; Cole, S.T.; et al. Mycobacterium tuberculosis Differentially Activates cGAS- and Inflammasome-Dependent Intracellular Immune Responses through ESX-1. Cell Host Microbe 2015, 17, 799–810. [Google Scholar] [CrossRef] [Green Version]
  108. Rahman, I.; Marwick, J.; Kirkham, P. Redox modulation of chromatin remodeling: Impact on histone acetylation and deacetylation, NF-kappaB and pro-inflammatory gene expression. Biochem. Pharmacol. 2004, 68, 1255–1267. [Google Scholar] [CrossRef]
  109. Roux, C.; Jafari, S.M.; Shinde, R.; Duncan, G.; Cescon, D.W.; Silvester, J.; Chu, M.F.; Hodgson, K.; Berger, T.; Wakeham, A.; et al. Reactive oxygen species modulate macrophage immunosuppressive phenotype through the up-regulation of PD-L1. Proc. Natl. Acad. Sci. USA 2019, 116, 4326–4335. [Google Scholar] [CrossRef] [Green Version]
  110. Hirayama, D.; Iida, T.; Nakase, H. The Phagocytic Function of Macrophage-Enforcing Innate Immunity and Tissue Homeostasis. Int. J. Mol. Sci. 2017, 19, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Lavin, Y.; Winter, D.; Blecher-Gonen, R.; David, E.; Keren-Shaul, H.; Merad, M.; Jung, S.; Amit, I. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 2014, 159, 1312–1326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Roszer, T. Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediat. Inflamm. 2015, 2015, 816460. [Google Scholar] [CrossRef] [Green Version]
  113. Dale, D.C.; Boxer, L.; Liles, W.C. The phagocytes: Neutrophils and monocytes. Blood 2008, 112, 935–945. [Google Scholar] [CrossRef] [PubMed]
  114. Tsai, C.S.; Chen, F.H.; Wang, C.C.; Huang, H.L.; Jung, S.M.; Wu, C.J.; Lee, C.C.; McBride, W.H.; Chiang, C.S.; Hong, J.H. Macrophages from irradiated tumors express higher levels of iNOS, arginase-I and COX-2, and promote tumor growth. Int. J. Radiat. Oncol. Biol. Phys. 2007, 68, 499–507. [Google Scholar] [CrossRef]
  115. van Ravenswaay Claasen, H.H.; Kluin, P.M.; Fleuren, G.J. Tumor infiltrating cells in human cancer. On the possible role of CD16+ macrophages in antitumor cytotoxicity. Lab. Investig. 1992, 67, 166–174. [Google Scholar] [PubMed]
  116. Baghban, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; Kolahian, S.; Javaheri, T.; Zare, P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal. 2020, 18, 59. [Google Scholar] [CrossRef] [Green Version]
  117. Denton, A.E.; Roberts, E.W.; Fearon, D.T. Stromal Cells in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2018, 1060, 99–114. [Google Scholar] [CrossRef] [PubMed]
  118. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
  119. Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef]
  120. Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
  121. Bingle, L.; Brown, N.J.; Lewis, C.E. The role of tumour-associated macrophages in tumour progression: Implications for new anticancer therapies. J. Pathol. 2002, 196, 254–265. [Google Scholar] [CrossRef] [PubMed]
  122. Kimura, Y.N.; Watari, K.; Fotovati, A.; Hosoi, F.; Yasumoto, K.; Izumi, H.; Kohno, K.; Umezawa, K.; Iguchi, H.; Shirouzu, K.; et al. Inflammatory stimuli from macrophages and cancer cells synergistically promote tumor growth and angiogenesis. Cancer Sci. 2007, 98, 2009–2018. [Google Scholar] [CrossRef]
  123. Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol. Rev. 2008, 222, 155–161. [Google Scholar] [CrossRef]
  124. Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Mei, J.; Xiao, Z.; Guo, C.; Pu, Q.; Ma, L.; Liu, C.; Lin, F.; Liao, H.; You, Z.; Liu, L. Prognostic impact of tumor-associated macrophage infiltration in non-small cell lung cancer: A systemic review and meta-analysis. Oncotarget 2016, 7, 34217–34228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Yin, S.; Huang, J.; Li, Z.; Zhang, J.; Luo, J.; Lu, C.; Xu, H.; Xu, H. The Prognostic and Clinicopathological Significance of Tumor-Associated Macrophages in Patients with Gastric Cancer: A Meta-Analysis. PLoS ONE 2017, 12, e0170042. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, Q.W.; Liu, L.; Gong, C.Y.; Shi, H.S.; Zeng, Y.H.; Wang, X.Z.; Zhao, Y.W.; Wei, Y.Q. Prognostic significance of tumor-associated macrophages in solid tumor: A meta-analysis of the literature. PLoS ONE 2012, 7, e50946. [Google Scholar] [CrossRef] [Green Version]
  128. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef] [PubMed]
  129. Zajac, E.; Schweighofer, B.; Kupriyanova, T.A.; Juncker-Jensen, A.; Minder, P.; Quigley, J.P.; Deryugina, E.I. Angiogenic capacity of M1- and M2-polarized macrophages is determined by the levels of TIMP-1 complexed with their secreted proMMP-9. Blood 2013, 122, 4054–4067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Ostuni, R.; Kratochvill, F.; Murray, P.J.; Natoli, G. Macrophages and cancer: From mechanisms to therapeutic implications. Trends Immunol. 2015, 36, 229–239. [Google Scholar] [CrossRef] [PubMed]
  131. Sayaman, R.W.; Saad, M.; Thorsson, V.; Hu, D.; Hendrickx, W.; Roelands, J.; Porta-Pardo, E.; Mokrab, Y.; Farshidfar, F.; Kirchhoff, T.; et al. Germline genetic contribution to the immune landscape of cancer. Immunity 2021, 54, 367–386.e8. [Google Scholar] [CrossRef]
  132. Zhu, J.; Jia, W.; Wu, C.; Fu, W.; Xia, H.; Liu, G.; He, J. Base Excision Repair Gene Polymorphisms and Wilms Tumor Susceptibility. EBioMedicine 2018, 33, 88–93. [Google Scholar] [CrossRef]
  133. Mattar, M.M.; Zekri, A.N.; Hussein, N.; Morsy, H.; Esmat, G.; Amin, M.A. Polymorphisms of base-excision repair genes and the hepatocarcinogenesis. Gene 2018, 675, 62–68. [Google Scholar] [CrossRef]
  134. Wallace, S.S.; Murphy, D.L.; Sweasy, J.B. Base excision repair and cancer. Cancer Lett. 2012, 327, 73–89. [Google Scholar] [CrossRef] [Green Version]
  135. Rozacky, J.; Nemec, A.A.; Sweasy, J.B.; Kidane, D. Gastric cancer associated variant of DNA polymerase beta (Leu22Pro) promotes DNA replication associated double strand breaks. Oncotarget 2015, 6, 24474–24487. [Google Scholar] [CrossRef] [Green Version]
  136. Popanda, O.; Seibold, P.; Nikolov, I.; Oakes, C.C.; Burwinkel, B.; Hausmann, S.; Flesch-Janys, D.; Plass, C.; Chang-Claude, J.; Schmezer, P. Germline variants of base excision repair genes and breast cancer: A polymorphism in DNA polymerase gamma modifies gene expression and breast cancer risk. Int. J. Cancer 2013, 132, 55–62. [Google Scholar] [CrossRef] [Green Version]
  137. Carter, H.; Marty, R.; Hofree, M.; Gross, A.M.; Jensen, J.; Fisch, K.M.; Wu, X.; DeBoever, C.; Van Nostrand, E.L.; Song, Y.; et al. Interaction Landscape of Inherited Polymorphisms with Somatic Events in Cancer. Cancer Discov. 2017, 7, 410–423. [Google Scholar] [CrossRef] [Green Version]
  138. Consortium, I.T.P.-C.A.o.W.G. Pan-cancer analysis of whole genomes. Nature 2020, 578, 82–93. [Google Scholar] [CrossRef] [Green Version]
  139. Goulielmaki, E.; Ioannidou, A.; Tsekrekou, M.; Stratigi, K.; Poutakidou, I.K.; Gkirtzimanaki, K.; Aivaliotis, M.; Evangelou, K.; Topalis, P.; Altmuller, J.; et al. Tissue-infiltrating macrophages mediate an exosome-based metabolic reprogramming upon DNA damage. Nat. Commun. 2020, 11, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Kitamura, T.; Qian, B.Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef] [PubMed]
  141. Pollard, J.W. Macrophages define the invasive microenvironment in breast cancer. J. Leukoc. Biol. 2008, 84, 623–630. [Google Scholar] [CrossRef] [Green Version]
  142. Ohtaki, Y.; Ishii, G.; Nagai, K.; Ashimine, S.; Kuwata, T.; Hishida, T.; Nishimura, M.; Yoshida, J.; Takeyoshi, I.; Ochiai, A. Stromal macrophage expressing CD204 is associated with tumor aggressiveness in lung adenocarcinoma. J. Thorac. Oncol. 2010, 5, 1507–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Soto, L.A.; Rafat, M.; Colomer, M.V.; Giaccia, A.; Graves, E. Tumor-associated macrophages enhance DNA damage repair and improve survival of murine breast cancers after irradiation. Cancer Res. 2016, 76, 1645. [Google Scholar]
  144. Lotze, M.T.; Tracey, K.J. High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nat. Rev. Immunol. 2005, 5, 331–342. [Google Scholar] [CrossRef] [PubMed]
  145. Germano, G.; Frapolli, R.; Belgiovine, C.; Anselmo, A.; Pesce, S.; Liguori, M.; Erba, E.; Uboldi, S.; Zucchetti, M.; Pasqualini, F.; et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013, 23, 249–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Di Mitri, D.; Mirenda, M.; Vasilevska, J.; Calcinotto, A.; Delaleu, N.; Revandkar, A.; Gil, V.; Boysen, G.; Losa, M.; Mosole, S.; et al. Re-education of Tumor-Associated Macrophages by CXCR2 Blockade Drives Senescence and Tumor Inhibition in Advanced Prostate Cancer. Cell Rep. 2019, 28, 2156–2168.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Mantovani, A.; Bottazzi, B.; Colotta, F.; Sozzani, S.; Ruco, L. The origin and function of tumor-associated macrophages. Immunol. Today 1992, 13, 265–270. [Google Scholar] [CrossRef]
  148. Sun, Y.; Nelson, P.S. Molecular pathways: Involving microenvironment damage responses in cancer therapy resistance. Clin. Cancer Res. 2012, 18, 4019–4025. [Google Scholar] [CrossRef] [Green Version]
  149. Wang, L.A.; Yang, B.; Tang, T.; Yang, Y.; Zhang, D.; Xiao, H.; Xu, J.; Wang, L.; Lin, L.; Jiang, J. Correlation of APE1 with VEGFA and CD163(+) macrophage infiltration in bladder cancer and their prognostic significance. Oncol. Lett. 2020, 20, 2881–2887. [Google Scholar] [CrossRef]
  150. Mehta, A.K.; Cheney, E.M.; Hartl, C.A.; Pantelidou, C.; Oliwa, M.; Castrillon, J.A.; Lin, J.R.; Hurst, K.E.; de Oliveira Taveira, M.; Johnson, N.T.; et al. Targeting immunosuppressive macrophages overcomes PARP inhibitor resistance in BRCA1-associated triple-negative breast cancer. Nat. Cancer 2021, 2, 66–82. [Google Scholar] [CrossRef] [PubMed]
  151. Mouchemore, K.A.; Pixley, F.J. CSF-1 signaling in macrophages: Pleiotrophy through phosphotyrosine-based signaling pathways. Crit. Rev. Clin. Lab. Sci. 2012, 49, 49–61. [Google Scholar] [CrossRef] [PubMed]
  152. Lin, E.Y.; Gouon-Evans, V.; Nguyen, A.V.; Pollard, J.W. The macrophage growth factor CSF-1 in mammary gland development and tumor progression. J. Mammary Gland. Biol. Neoplasia 2002, 7, 147–162. [Google Scholar] [CrossRef]
  153. Azzam, G.; Wang, X.; Bell, D.; Murphy, M.E. CSF1 is a novel p53 target gene whose protein product functions in a feed-forward manner to suppress apoptosis and enhance p53-mediated growth arrest. PLoS ONE 2013, 8, e74297. [Google Scholar] [CrossRef]
  154. DeNardo, D.G.; Brennan, D.J.; Rexhepaj, E.; Ruffell, B.; Shiao, S.L.; Madden, S.F.; Gallagher, W.M.; Wadhwani, N.; Keil, S.D.; Junaid, S.A.; et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011, 1, 54–67. [Google Scholar] [CrossRef] [Green Version]
  155. Pedersen, M.B.; Danielsen, A.V.; Hamilton-Dutoit, S.J.; Bendix, K.; Norgaard, P.; Moller, M.B.; Steiniche, T.; d’Amore, F. High intratumoral macrophage content is an adverse prognostic feature in anaplastic large cell lymphoma. Histopathology 2014, 65, 490–500. [Google Scholar] [CrossRef] [PubMed]
  156. Nguewa, P.A.; Fuertes, M.A.; Valladares, B.; Alonso, C.; Perez, J.M. Poly(ADP-ribose) polymerases: Homology, structural domains and functions. Novel therapeutical applications. Prog. Biophys. Mol. Biol. 2005, 88, 143–172. [Google Scholar] [CrossRef] [PubMed]
  157. Chen, A. PARP inhibitors: Its role in treatment of cancer. Chin. J. Cancer 2011, 30, 463–471. [Google Scholar] [CrossRef]
  158. Ronson, G.E.; Piberger, A.L.; Higgs, M.R.; Olsen, A.L.; Stewart, G.S.; McHugh, P.J.; Petermann, E.; Lakin, N.D. PARP1 and PARP2 stabilise replication forks at base excision repair intermediates through Fbh1-dependent Rad51 regulation. Nat. Commun. 2018, 9, 746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. El-Khamisy, S.F.; Masutani, M.; Suzuki, H.; Caldecott, K.W. A requirement for PARP-1 for the assembly or stability of XRCC1 nuclear foci at sites of oxidative DNA damage. Nucleic Acids Res. 2003, 31, 5526–5533. [Google Scholar] [CrossRef] [Green Version]
  160. Sobczak, M.; Zyma, M.; Robaszkiewicz, A. The Role of PARP1 in Monocyte and Macrophage Commitment and Specification: Future Perspectives and Limitations for the Treatment of Monocyte and Macrophage Relevant Diseases with PARP Inhibitors. Cells 2020, 9, 2040. [Google Scholar] [CrossRef] [PubMed]
  161. Tokarz, P.; Płoszaj, T.; Regdon, Z.; Virag, L.; Robaszkiewicz, A. PARP1-LSD1 functional interplay controls transcription of SOD2 that protects human pro-inflammatory macrophages from death under an oxidative condition. Free Radic. Biol. Med. 2019, 131, 218–224. [Google Scholar] [CrossRef]
  162. Lee, E.K.; Konstantinopoulos, P.A. PARP inhibition and immune modulation: Scientific rationale and perspectives for the treatment of gynecologic cancers. Ther. Adv. Med. Oncol. 2020, 12, 1758835920944116. [Google Scholar] [CrossRef]
  163. Scaffidi, P.; Misteli, T.; Bianchi, M.E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002, 418, 191–195. [Google Scholar] [CrossRef] [PubMed]
  164. Wang, H.; Bloom, O.; Zhang, M.; Vishnubhakat, J.M.; Ombrellino, M.; Che, J.; Frazier, A.; Yang, H.; Ivanova, S.; Borovikova, L.; et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999, 285, 248–251. [Google Scholar] [CrossRef] [PubMed]
  165. Muller, S.; Scaffidi, P.; Degryse, B.; Bonaldi, T.; Ronfani, L.; Agresti, A.; Beltrame, M.; Bianchi, M.E. New EMBO members’ review: The double life of HMGB1 chromatin protein: Architectural factor and extracellular signal. EMBO J. 2001, 20, 4337–4340. [Google Scholar] [CrossRef]
  166. Falciola, L.; Spada, F.; Calogero, S.; Langst, G.; Voit, R.; Grummt, I.; Bianchi, M.E. High mobility group 1 protein is not stably associated with the chromosomes of somatic cells. J. Cell Biol. 1997, 137, 19–26. [Google Scholar] [CrossRef] [Green Version]
  167. Bianchi, M.E. HMGB1 loves company. J. Leukoc. Biol. 2009, 86, 573–576. [Google Scholar] [CrossRef]
  168. Gorgulho, C.M.; Romagnoli, G.G.; Bharthi, R.; Lotze, M.T. Johnny on the Spot-Chronic Inflammation Is Driven by HMGB1. Front. Immunol. 2019, 10, 1561. [Google Scholar] [CrossRef]
  169. Rojas, A.; Delgado-Lopez, F.; Perez-Castro, R.; Gonzalez, I.; Romero, J.; Rojas, I.; Araya, P.; Anazco, C.; Morales, E.; Llanos, J. HMGB1 enhances the protumoral activities of M2 macrophages by a RAGE-dependent mechanism. Tumor Biol. 2016, 37, 3321–3329. [Google Scholar] [CrossRef] [PubMed]
  170. Liu, T.; Xiang, A.; Peng, T.; Doran, A.C.; Tracey, K.J.; Barnes, B.J.; Tabas, I.; Son, M.; Diamond, B. HMGB1-C1q complexes regulate macrophage function by switching between leukotriene and specialized proresolving mediator biosynthesis. Proc. Natl. Acad. Sci. USA 2019, 116, 23254–23263. [Google Scholar] [CrossRef] [PubMed]
  171. Chabanon, R.M.; Muirhead, G.; Krastev, D.B.; Adam, J.; Morel, D.; Garrido, M.; Lamb, A.; Henon, C.; Dorvault, N.; Rouanne, M.; et al. PARP inhibition enhances tumor cell-intrinsic immunity in ERCC1-deficient non-small cell lung cancer. J. Clin. Investig. 2019, 129, 1211–1228. [Google Scholar] [CrossRef]
  172. Ding, L.; Kim, H.J.; Wang, Q.; Kearns, M.; Jiang, T.; Ohlson, C.E.; Li, B.B.; Xie, S.; Liu, J.F.; Stover, E.H.; et al. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep. 2018, 25, 2972–2980.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Dunphy, G.; Flannery, S.M.; Almine, J.F.; Connolly, D.J.; Paulus, C.; Jonsson, K.L.; Jakobsen, M.R.; Nevels, M.M.; Bowie, A.G.; Unterholzner, L. Non-canonical Activation of the DNA Sensing Adaptor STING by ATM and IFI16 Mediates NF-kappaB Signaling after Nuclear DNA Damage. Mol. Cell 2018, 71, 745–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Almine, J.F.; O’Hare, C.A.; Dunphy, G.; Haga, I.R.; Naik, R.J.; Atrih, A.; Connolly, D.J.; Taylor, J.; Kelsall, I.R.; Bowie, A.G.; et al. IFI16 and cGAS cooperate in the activation of STING during DNA sensing in human keratinocytes. Nat. Commun. 2017, 8, 14392. [Google Scholar] [CrossRef]
  175. Jonsson, K.L.; Laustsen, A.; Krapp, C.; Skipper, K.A.; Thavachelvam, K.; Hotter, D.; Egedal, J.H.; Kjolby, M.; Mohammadi, P.; Prabakaran, T.; et al. IFI16 is required for DNA sensing in human macrophages by promoting production and function of cGAMP. Nat. Commun. 2017, 8, 14391. [Google Scholar] [CrossRef]
  176. Xanthoudakis, S.; Curran, T. Identification and characterization of Ref-1, a nuclear protein that facilitates AP-1 DNA-binding activity. EMBO J. 1992, 11, 653–665. [Google Scholar] [CrossRef] [PubMed]
  177. Demple, B.; Herman, T.; Chen, D.S. Cloning and expression of APE, the cDNA encoding the major human apurinic endonuclease: Definition of a family of DNA repair enzymes. Proc. Natl. Acad. Sci. USA 1991, 88, 11450–11454. [Google Scholar] [CrossRef] [Green Version]
  178. Tell, G.; Quadrifoglio, F.; Tiribelli, C.; Kelley, M.R. The many functions of APE1/Ref-1: Not only a DNA repair enzyme. Antioxid. Redox Signal. 2009, 11, 601–620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Woo, J.; Park, H.; Sung, S.H.; Moon, B.I.; Suh, H.; Lim, W. Prognostic value of human apurinic/apyrimidinic endonuclease 1 (APE1) expression in breast cancer. PLoS ONE 2014, 9, e99528. [Google Scholar] [CrossRef]
  180. Yoo, D.G.; Song, Y.J.; Cho, E.J.; Lee, S.K.; Park, J.B.; Yu, J.H.; Lim, S.P.; Kim, J.M.; Jeon, B.H. Alteration of APE1/ref-1 expression in non-small cell lung cancer: The implications of impaired extracellular superoxide dismutase and catalase antioxidant systems. Lung Cancer 2008, 60, 277–284. [Google Scholar] [CrossRef] [PubMed]
  181. Yuan, C.L.; He, F.; Ye, J.Z.; Wu, H.N.; Zhang, J.Y.; Liu, Z.H.; Li, Y.Q.; Luo, X.L.; Lin, Y.; Liang, R. APE1 overexpression is associated with poor survival in patients with solid tumors: A meta-analysis. Oncotarget 2017, 8, 59720–59728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Jedinak, A.; Dudhgaonkar, S.; Kelley, M.R.; Sliva, D. Apurinic/Apyrimidinic endonuclease 1 regulates inflammatory response in macrophages. Anticancer Res. 2011, 31, 379–385. [Google Scholar] [PubMed]
  183. Qing, Y.; Li, Q.; Ren, T.; Xia, W.; Peng, Y.; Liu, G.L.; Luo, H.; Yang, Y.X.; Dai, X.Y.; Zhou, S.F.; et al. Upregulation of PD-L1 and APE1 is associated with tumorigenesis and poor prognosis of gastric cancer. Drug Des. Dev. Ther. 2015, 9, 901–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Balkwill, F.; Charles, K.A.; Mantovani, A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 2005, 7, 211–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Lawrence, T.; Willoughby, D.A.; Gilroy, D.W. Anti-inflammatory lipid mediators and insights into the resolution of inflammation. Nat. Rev. Immunol. 2002, 2, 787–795. [Google Scholar] [CrossRef]
  186. Kaplanski, G.; Marin, V.; Montero-Julian, F.; Mantovani, A.; Farnarier, C. IL-6: A regulator of the transition from neutrophil to monocyte recruitment during inflammation. Trends Immunol. 2003, 24, 25–29. [Google Scholar] [CrossRef]
  187. Bosca, L.; Zeini, M.; Traves, P.G.; Hortelano, S. Nitric oxide and cell viability in inflammatory cells: A role for NO in macrophage function and fate. Toxicology 2005, 208, 249–258. [Google Scholar] [CrossRef]
  188. Aguilera-Aguirre, L.; Bacsi, A.; Radak, Z.; Hazra, T.K.; Mitra, S.; Sur, S.; Brasier, A.R.; Ba, X.; Boldogh, I. Innate inflammation induced by the 8-oxoguanine DNA glycosylase-1-KRAS-NF-kappaB pathway. J. Immunol. 2014, 193, 4643–4653. [Google Scholar] [CrossRef] [Green Version]
  189. Arneth, B. Systemic Lupus Erythematosus and DNA Degradation and Elimination Defects. Front Immunol. 2019, 10, 1697. [Google Scholar] [CrossRef] [Green Version]
  190. Seo, Y.; Kinsella, T.J. Essential role of DNA base excision repair on survival in an acidic tumor microenvironment. Cancer Res. 2009, 69, 7285–7293. [Google Scholar] [CrossRef] [Green Version]
  191. Xia, T.; Konno, H.; Ahn, J.; Barber, G.N. Deregulation of STING Signaling in Colorectal Carcinoma Constrains DNA Damage Responses and Correlates With Tumorigenesis. Cell Rep. 2016, 14, 282–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Tan, Y.S.; Sansanaphongpricha, K.; Xie, Y.; Donnelly, C.R.; Luo, X.; Heath, B.R.; Zhao, X.; Bellile, E.; Hu, H.; Chen, H.; et al. Mitigating SOX2-potentiated Immune Escape of Head and Neck Squamous Cell Carcinoma with a STING-inducing Nanosatellite Vaccine. Clin. Cancer Res. 2018, 24, 4242–4255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Kim, C.; Wang, X.D.; Yu, Y. PARP1 inhibitors trigger innate immunity via PARP1 trapping-induced DNA damage response. eLife 2020, 9, e60637. [Google Scholar] [CrossRef]
  194. Amouzegar, A.; Chelvanambi, M.; Filderman, J.N.; Storkus, W.J.; Luke, J.J. STING Agonists as Cancer Therapeutics. Cancers 2021, 13, 2695. [Google Scholar] [CrossRef]
  195. Wang, C.; Guan, Y.; Lv, M.; Zhang, R.; Guo, Z.; Wei, X.; Du, X.; Yang, J.; Li, T.; Wan, Y.; et al. Manganese Increases the Sensitivity of the cGAS-STING Pathway for Double-Stranded DNA and Is Required for the Host Defense against DNA Viruses. Immunity 2018, 48, 675–687.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Zhao, Z.; Ma, Z.; Wang, B.; Guan, Y.; Su, X.D.; Jiang, Z. Mn(2+) Directly Activates cGAS and Structural Analysis Suggests Mn(2+) Induces a Noncanonical Catalytic Synthesis of 2′3′-cGAMP. Cell Rep. 2020, 32, 108053. [Google Scholar] [CrossRef]
  197. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Borresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [Green Version]
  198. Donigan, K.A.; Hile, S.E.; Eckert, K.A.; Sweasy, J.B. The human gastric cancer-associated DNA polymerase beta variant D160N is a mutator that induces cellular transformation. DNA Repair 2012, 11, 381–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Rivera, B.; Castellsague, E.; Bah, I.; van Kempen, L.C.; Foulkes, W.D. Biallelic NTHL1 Mutations in a Woman with Multiple Primary Tumors. N. Engl. J. Med. 2015, 373, 1985–1986. [Google Scholar] [CrossRef] [PubMed]
  200. Donigan, K.A.; Sun, K.W.; Nemec, A.A.; Murphy, D.L.; Cong, X.; Northrup, V.; Zelterman, D.; Sweasy, J.B. Human POLB gene is mutated in high percentage of colorectal tumors. J. Biol. Chem. 2012, 287, 23830–23839. [Google Scholar] [CrossRef] [Green Version]
  201. Weber, J.S.; Kudchadkar, R.R.; Yu, B.; Gallenstein, D.; Horak, C.E.; Inzunza, H.D.; Zhao, X.; Martinez, A.J.; Wang, W.; Gibney, G.; et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J. Clin. Oncol. 2013, 31, 4311–4318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef] [PubMed]
  203. Sato, H.; Niimi, A.; Yasuhara, T.; Permata, T.B.M.; Hagiwara, Y.; Isono, M.; Nuryadi, E.; Sekine, R.; Oike, T.; Kakoti, S.; et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat. Commun. 2017, 8, 1751. [Google Scholar] [CrossRef] [PubMed]
  204. Tian, W.; Shan, B.; Zhang, Y.; Ren, Y.; Liang, S.; Zhao, J.; Zhao, Z.; Wang, G.; Zhao, X.; Peng, D.; et al. Association between DNA damage repair gene somatic mutations and immune-related gene expression in ovarian cancer. Cancer Med. 2020, 9, 2190–2200. [Google Scholar] [CrossRef] [Green Version]
  205. Permata, T.B.M.; Hagiwara, Y.; Sato, H.; Yasuhara, T.; Oike, T.; Gondhowiardjo, S.; Held, K.D.; Nakano, T.; Shibata, A. Base excision repair regulates PD-L1 expression in cancer cells. Oncogene 2019, 38, 4452–4466. [Google Scholar] [CrossRef] [PubMed]
  206. Colebatch, A.; Hitchins, M.; Williams, R.; Meagher, A.; Hawkins, N.J.; Ward, R.L. The role of MYH and microsatellite instability in the development of sporadic colorectal cancer. Br. J. Cancer 2006, 95, 1239–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Lizotte, P.H.; Baird, J.R.; Stevens, C.A.; Lauer, P.; Green, W.R.; Brockstedt, D.G.; Fiering, S.N. Attenuated Listeria monocytogenes reprograms M2-polarized tumor-associated macrophages in ovarian cancer leading to iNOS-mediated tumor cell lysis. Oncoimmunology 2014, 3, e28926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Base excision repair capacity in macrophage. Monocytes are key players in the immune system and contribute as a source for dendritic cells and macrophages. Monocytes differentiate into M1 and M2 macrophages. DNA repair capacity restored in M1 and M2 macrophage. Monocytes are deficient in main base excision repair genes, including POLB, XRCC1, and ligase III and non-homologous end joining repair genes, including DNA-PK.
Figure 1. Base excision repair capacity in macrophage. Monocytes are key players in the immune system and contribute as a source for dendritic cells and macrophages. Monocytes differentiate into M1 and M2 macrophages. DNA repair capacity restored in M1 and M2 macrophage. Monocytes are deficient in main base excision repair genes, including POLB, XRCC1, and ligase III and non-homologous end joining repair genes, including DNA-PK.
Biomedicines 10 00557 g001
Figure 2. Aberrant BER induces DNA-sensor-mediated inflammatory signaling. The cGAS-STING signaling pathway in BER deficient tumor cells (A) and immune cells (B). In both cases, tumor-associated mutation of BER genes or germline variant associated with impaired BER function in cancer cells and/or macrophage may contribute to accumulation of unrepaired endogenous oxidative DNA damage product that may leak to cytosol and stimulate cGAS and produce second messenger 2′,3′-cGAMP. After 2′,3′-cGAMP binds to STING protein and then phosphorylate TBK1 that leads to IRF3 activation and/or NF-κB to facilitate their translocation into the nucleus and drives the expression of inflammatory genes, including Type I interferon. In contrast, macrophages with high DNA repair capacity (without pathogenic variant of BER gens) suppress innate immune signaling mediated by cGAS/STING. (C) Schematic representation of impaired BER function will likely leads to accumulation of oxidative stress related DNA damage and may trigger the release of cytosolic DNA and cGAS/STING dependent inflammatory response.
Figure 2. Aberrant BER induces DNA-sensor-mediated inflammatory signaling. The cGAS-STING signaling pathway in BER deficient tumor cells (A) and immune cells (B). In both cases, tumor-associated mutation of BER genes or germline variant associated with impaired BER function in cancer cells and/or macrophage may contribute to accumulation of unrepaired endogenous oxidative DNA damage product that may leak to cytosol and stimulate cGAS and produce second messenger 2′,3′-cGAMP. After 2′,3′-cGAMP binds to STING protein and then phosphorylate TBK1 that leads to IRF3 activation and/or NF-κB to facilitate their translocation into the nucleus and drives the expression of inflammatory genes, including Type I interferon. In contrast, macrophages with high DNA repair capacity (without pathogenic variant of BER gens) suppress innate immune signaling mediated by cGAS/STING. (C) Schematic representation of impaired BER function will likely leads to accumulation of oxidative stress related DNA damage and may trigger the release of cytosolic DNA and cGAS/STING dependent inflammatory response.
Biomedicines 10 00557 g002
Figure 3. BER gene landscape and its impacts on macrophage infiltration in tumor microenvironment. (A) Heatmap of overexpression of BER genes from 40 types of cancer and 17 different BER repair genes expression in tumor cells versus macrophage infiltration. (B) Five representative genes (NEIL2, OGG1, APEX1, POLB, DNA LIG3) selected to demonstrate the correlation of overexpression of those repair genes and M2 infiltration in tumor microenvironment. Rho represents correlation coefficient. All data were extracted from TCGA data set and analyzed using TIMER 2.0 using CIBERSORT.
Figure 3. BER gene landscape and its impacts on macrophage infiltration in tumor microenvironment. (A) Heatmap of overexpression of BER genes from 40 types of cancer and 17 different BER repair genes expression in tumor cells versus macrophage infiltration. (B) Five representative genes (NEIL2, OGG1, APEX1, POLB, DNA LIG3) selected to demonstrate the correlation of overexpression of those repair genes and M2 infiltration in tumor microenvironment. Rho represents correlation coefficient. All data were extracted from TCGA data set and analyzed using TIMER 2.0 using CIBERSORT.
Biomedicines 10 00557 g003aBiomedicines 10 00557 g003b
Figure 4. BER-based therapeutic intervention in immune cells (macrophage) modulates inflammatory signaling. Macrophages harbor pathogenic germline variant of BER genes treated with PARP1 inhibitor (Olaparib) will likely accumulate unrepaired products such as PARP1-DNA complex and generate cytosolic DNA to stimulate cGAS/STING pathways and enhance type interferon response (represent by blue color). In contrast, macrophages harbor overexpression of BER genes (e.g., APE1) targeted by APE1 inhibitor (E3330) suppresses NF-kB mediated inflammatory response (represented by purple color).
Figure 4. BER-based therapeutic intervention in immune cells (macrophage) modulates inflammatory signaling. Macrophages harbor pathogenic germline variant of BER genes treated with PARP1 inhibitor (Olaparib) will likely accumulate unrepaired products such as PARP1-DNA complex and generate cytosolic DNA to stimulate cGAS/STING pathways and enhance type interferon response (represent by blue color). In contrast, macrophages harbor overexpression of BER genes (e.g., APE1) targeted by APE1 inhibitor (E3330) suppresses NF-kB mediated inflammatory response (represented by purple color).
Biomedicines 10 00557 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhao, S.; Habib, S.L.; Senejani, A.G.; Sebastian, M.; Kidane, D. Role of Base Excision Repair in Innate Immune Cells and Its Relevance for Cancer Therapy. Biomedicines 2022, 10, 557. https://doi.org/10.3390/biomedicines10030557

AMA Style

Zhao S, Habib SL, Senejani AG, Sebastian M, Kidane D. Role of Base Excision Repair in Innate Immune Cells and Its Relevance for Cancer Therapy. Biomedicines. 2022; 10(3):557. https://doi.org/10.3390/biomedicines10030557

Chicago/Turabian Style

Zhao, Shengyuan, Samy L. Habib, Alireza G. Senejani, Manu Sebastian, and Dawit Kidane. 2022. "Role of Base Excision Repair in Innate Immune Cells and Its Relevance for Cancer Therapy" Biomedicines 10, no. 3: 557. https://doi.org/10.3390/biomedicines10030557

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop