Next Article in Journal
Establishment and Validation of an Individualized Cell Cycle Process-Related Gene Signature to Predict Cancer-Specific Survival in Patients with Bladder Cancer
Next Article in Special Issue
HuRdling Senescence: HuR Breaks BRAF-Induced Senescence in Melanocytes and Supports Melanoma Growth
Previous Article in Journal
Direct Oral Anticoagulants in Cancer Patients. Time for a Change in Paradigm
Previous Article in Special Issue
Therapy-Induced Senescence: An “Old” Friend Becomes the Enemy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Senescence and Cancer: Role of Nitric Oxide (NO) in SASP

by
Nesrine Mabrouk
1,2,†,
Silvia Ghione
1,2,†,
Véronique Laurens
1,2,
Stéphanie Plenchette
1,2,
Ali Bettaieb
1,2 and
Catherine Paul
1,2,*
1
Laboratory of Immunology and Immunotherapy of Cancers, EPHE, PSL Research University, 75000 Paris, France
2
Laboratory of Immunology and Immunotherapy of Cancers (LIIC), EA7269, University of Burgundy Franche-Comté, 21000 Dijon, France
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2020, 12(5), 1145; https://doi.org/10.3390/cancers12051145
Submission received: 26 March 2020 / Revised: 27 April 2020 / Accepted: 30 April 2020 / Published: 2 May 2020
(This article belongs to the Special Issue Senescence and Cancer)

Abstract

:
Cellular senescence is a cell state involved in both physiological and pathological processes such as age-related diseases and cancer. While the mechanism of senescence is now well known, its role in tumorigenesis still remains very controversial. The positive and negative effects of senescence on tumorigenesis depend largely on the diversity of the senescent phenotypes and, more precisely, on the senescence-associated secretory phenotype (SASP). In this review, we discuss the modulatory effect of nitric oxide (NO) in SASP and the possible benefits of the use of NO donors or iNOS inducers in combination with senotherapy in cancer treatment.

1. Senescence and Cancer

Senescence is an important biological mechanism that plays a crucial role in many physiological processes, such as development and wound healing, and also in various age-related pathologies including cancer [1,2,3]. Indeed, there is a strong relationship between aging and cancer. In both cases, the mechanisms involve an accumulation of cellular damage over time. However, other processes seem to be in opposition, such as hyperproliferation and the increase in cell survival in cancer or the decreased function and fitness in aging [4]. However, many hallmarks of aging are found in cancer, and senescence is one of them [5]. Senescence was first described in 1961 by Hayflick and Moorhead after observing that human diploid cells have a replicative limit in culture [6]. This process, named replicative senescence, is the first form of senescence described and involves the shortening of telomeres [7]. The loss of telomeres is recognized as DNA damage and triggers a DNA-damage response (DDR), involving DNA damage kinases such as ataxia-telangiectasia-mutated protein (ATM), ataxia-telangiectasia and Rad3-related protein (ATR) and checkpoint kinases 1 and 2 (CHK1 and CHK2), which in turn activate cell cycle proteins such as p53 [8]. DDR is also induced by other external damaging factors such as ionizing radiation and chemotherapeutic drugs [9]. Other forms of non-telomeric senescence, called premature senescence, have been described such as stress-induced senescence, which involves reactive oxygen species (ROS), and oncogene-induced senescence (OIS). OIS was firstly described by Serrano et al. (1997) in a model of human fibroblasts expressing an oncogenic form of RASG12V [10]. Since then, more than 50 oncogenes have been described to induce senescence once activated [11]. In most OIS cases, the senescent phenotype is invariably preceded by hyperproliferation, aberrant replication and activation of the DDR [12,13]. PICS, PTEN-loss induced cellular senescence [14] is also a form of premature senescence, occurring independently of DDR or hyper-replication [15]. The loss of two other tumor suppressors, neurofibromin 1 (NF1) and the von Hippel-Lindau factor, has also been described to induce senescence in the same manner as for PICS [9].
The senescent phenotype is often characterized by the modulation of different signaling pathways and a series of cellular events such as the activation of the DDR; cell cycle arrest via the activation of the p53/p21CIP1 and p16INK4A/Retinoblastoma (Rb) pathways; the induction of a senescence-associated secretory phenotype (SASP); resistance against apoptosis; the induction of endoplasmic reticulum stress; and the modulation of metabolism [16]. Since all these biological aspects are not specific to the senescence process, multiple markers should be considered to accurately define the senescent phenotype. The most commonly used assay is based on the histochemical detection of Senescence-Associated beta-galactosidase (SA-β-gal) [17]. Other canonical senescence markers include senescence inducers such as CDK inhibitors (p16, p21, p15 and p27) and the phosphorylated forms of p53 and Rb, which are proteins related to senescence-associated heterochromatin foci (SAHF) and the expression of extracellular factors associated with SASP [9]. In vitro morphological changes are also a common feature of senescence. Indeed, senescent cells are often characterized by structural cell changes such as enlarged morphology, the modification of plasma membrane composition, the accumulation of lysosomes and mitochondria, and nuclear changes [16].
Cellular senescence was primarily described as a tumor suppressor mechanism [14,18,19,20]. Indeed, first of all, senescence can inhibit the growth of cancer cells and then induce SASP, which in turn induces the recruitment of immune cells. However, the aging of tumor microenvironment and genetic and epigenetic mutations induced by tumor progression cooperate to bypass senescence [4]. Moreover, the long-term implications of senescent cells—and more particularly, the paracrine effect of SASP—potentially favor tumor development [21,22,23,24]. Recently, in a very comprehensive review, Faget et al. (2019) highlighted the existence of different SASPs in immunosupportive or immunosuppressive scenarios [25]. To counteract the pro-tumorigenic effects of SASP, new therapies have also been developed to dampen SASP’s proinflammatory effects (senomorphic) or to specifically clear senescent cells (senolytics) [26]. These senotherapies selectively kill senescent cells or stimulate their elimination by the immune system [27]. These approaches include chemical inhibitors of the nuclear factor-kappa B (NF-κB), Janus kinase (JAK) and mammalian target of rapamycin (mTOR) pathways and free radical scavengers [28,29,30].
Nitric oxide (NO) might be included in these therapies because a lot of pathways are regulated by post-translational modifications—such as cysteine S-nitrosylation (Cys-NO), tyrosine nitration (Tyr-NO) or even metal nitrosylation [31]—and because NO is also strongly implicated in immunomodulation [32]. Here, we describe the involvement of NO in senescence via (1) the modulation of SASP pathways, (2) the regulation of apoptosis via death receptor pathway modulation, (3) immunomodulation and (4) the modulation of the senolytic effects. Altogether, these data pave the way towards the development of new therapeutic strategies based on NO.

2. SASP in Cancer: A Double-Edged Sword

It is now well established that senescent cells are able to secrete a pool of molecules that define the SASP. This condition mediates the paracrine activity of senescent cells, which results in tissue microenvironment modulation [25]. Many SASP components are actually identified, such as growth factors, cytokines, chemokines, proteases and extracellular matrix (ECM) components [26,33]. SASP soluble factors include, among others, interleukin 6 (IL-6), IL1α, IL-10, IL-13, IL-15, chemokines such as CC motif ligand 2 (CCL2), CCL5, CCL8 and C-X-C motif 1 (CXCL1), CXCL2, CXCL5, CXCL12, granulocytes macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor 1 (CSF1), and cytokines such as interferon γ (IFNγ), tumor necrosis factor α (TNFα), insulin-like growth factors (IGFs) and transforming growth factor β (TGFβ) [33,34]. In addition to soluble factors, senescent cells can secrete proteases, such as matrix metalloproteinases (MMPs) and serine proteases, and regulators of plasminogen activation pathways, which induce ECM modifications. In fact, it is well established that ECM changes (e.g., rigidity loss) increase the metastatic properties of cancer cells and provide optimal conditions for migration. These changes in the ECM are associated with poor prognosis in cancer patients [35]. For this reason, these molecules are involved in carcinogenesis and cell migration. MMP family members involved in SASP are stromelysin-1 and -2 (also known as MMP-3 and -10) and collagenase 1 (MMP-1), while plasminogen activators include urokinase (uPA), tissue-type plasminogen activators (tPA), uPA receptor (uPAR) and inhibitors of these serine proteases (PAI-1 and 2) [24,36]. The SASP factors are summarized in Table 1.
The SASP molecules can modulate stromal and immune cells, with a different role in cancer development. In fact, SASP is considered as a double-edged sword because it can lead to either anti-tumorigenic or a pro-tumorigenic effects in a context-dependent manner. Indeed, several SASP factors induce the clearance of senescent pre-neoplastic cells, preserving tissue homeostasis. During the early stage of senescence transformation, these cells are recognized and eliminated by the immune system recruited by SASP, to prevent the malignant transformation [37]. For example, a high level of TGFβ secreted by macrophages in the tumor microenvironment maintains cellular senescence and decreases tumor growth in aggressive B cell lymphoma [38]. Alessio et al. (2019) recently showed that the induction of acute SASP (A-SASP) in mesenchymal stem cells (MSCs) decreases immortalized prostate cancer cell PNT2 proliferation in vitro [39].
Nevertheless, long-term exposure to SASP factors or the aging of the tumor microenvironment increases inflammation and tumor progression [4,25]. Therefore, senescent cells can also contribute to tumorigenesis via SASP [40]. The accumulation of cytoplasmic DNA in senescent cells is able to activate the GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, causing chronic inflammation. cGAS binds to dsDNA, and this condition causes conformational changes in its catalytic center, inducing the catalysis of cyclic GMP-AMP (cGAMP). This second messenger stimulates STING, an endoplasmic reticulum-located protein, which induces the activation and nuclear translocation of interferon-regulatory factor 3 (IRF3) and NF-κB. This causes the expression of, for example, type I IFN, a crucial player in inflammation [41]. STING activation is associated with inflammatory diseases, and the link between inflammation and cancer is well recognized [42]. For example, several studies reported the importance of the STING signaling pathway in 7,12dimethylbenz[α]anthracene (DMBA)-induced carcinogenesis. This compound can drive the development of skin tumors by DNA damage, which induces STING-dependent pro-inflammatory cytokine production [43,44]. Furthermore, a recent study has shown that STING-deficient mice are resistant to DMBA-induced skin tumors [45]. Another study showed that STING activation induces obesity-associated hepatocellular carcinoma (HCC) in mice [46]. The over-activation of the cGAS-STING signaling pathway was also described in lung cancer [47].
Although chemotherapy is beneficial to treat cancer, these compounds can promote SASP secretion via increased DNA damage, leading to inflammation and subsequent cancer progression [48]. It is actually known that several anti-cancer drugs, such as docetaxel, bleomycin, cyclophosphamide, doxorubicin, vincristine, etoposide, 5-fluorouracil (5-FU), cisplatin and also ionizing radiation can mediate tumor-induced senescence [34,49]. This mechanism is principally mediated by DNA damage and inflammation in the tumor microenvironment. In this context, Tato-Costa et al. (2016) showed that the SASP induced by 5-fluorouracil (5-FU) and doxorubicin causes epithelial-to-mesenchymal transition (EMT) in vitro and in clinical samples from patients with rectal cancer [49]. Another important factor included in SASP is NO [50]. Senescent cells are able to secrete NO, via nitric oxide synthase (NOS). In the SASP microenvironment, fibroblasts are major NO producers, enhancing cancer cell proliferation. It is well known that NO is involved in monocyte differentiation, and these cells, under certain conditions, can induce cancer progression [51,52]. Moreover, NO is also involved in the modulation of a large number of signaling pathways [53].

3. Role of NO in the Induction of the SASP

According to previous reports, several mechanisms are involved in the regulation of SASP factors. Considering the large number of signaling molecules involved in SASP secretion, the present review is focused on the most crucial ones. The transcription factor NF-κB plays an important role in SASP. It can be activated by several SASP inducers such as toll like receptor 2 (TLR2), TNFα, reactive oxygen species (ROS) and genotoxic agents [54,55]. The activation of NF-κB involves IκB phosphorylation by IκB kinase (IKK), which leads to its degradation by the proteasome, enabling the active NF-κB transcription factor (p65/p50) to translocate into the nucleus and thereby induce the expression of target genes such as those which code for IL-6, IL-8 and CXCL1 [56,57,58]. Other senescence activators such as CXCR2 ligands IL-8 and GRO/Gro1 are also upregulated in cells undergoing senescence following the activation of mitogen-activated kinases [59]. Recently, Loo et al. (2017) reported that the expression of cyclooxygenase 2 (COX-2), a rate limiting enzyme involved in prostaglandin biosynthesis, is increased in senescent hepatic stellate cells and triggers the overproduction of the SASP prostaglandin E2 (PGE2) via PGE2 receptor EP4, limiting anti-tumor immunity in obesity-associated HCC [60].
The identification of signals that can promote senescence in a given tumor type may provide new therapeutic targets for cancer. Several drugs (see below) modulate some of these signals, including NO.
NO, a highly reactive free radical, has pleotropic functions in multiple biological processes, such as neurotransmission, vasodilatation and macrophage-mediated immunity. NO is synthetized from L-arginine by NOS in the presence of oxygen. Endothelial NOS (eNOS) and neuronal NOS (nNOS), two NOSs whose activities are dependent on calcium levels, produce low amounts of NO, while the third NOS is inducible (iNOS) and independent of calcium levels. iNOS is activated by IL-1β, Toll Like 4 receptor agonists (TLR4), interferon γ (IFNγ) and oxidative stress and produces higher amounts of NO [61]. As a very unstable molecule, NO reacts mainly with the superoxide anion to generate peroxynitrite, a compound with high oxidative potential [62].
NO is also released from pharmacological agents. There are direct NO donors—such as sodium nitroprusside, molsidomine or diethylamino-NONOate—and donors that require metabolism such as the classic nitrovasodilators and organic nitrate and nitrite esters—including nitroglycerin (also called glyceryl trinitrate or GTN), isosorbide dinitrate, isosorbide 5-mononitrate and nicorandil—that have been used in the treatment of cardiovascular diseases [63,64,65].
In cancer, NO plays a role in promoting as well as inhibiting tumors [62,66,67]. This dichotomy seems to be related to its concentration, its location and its targets [68]. A few clinical studies have shown that NO donors could mediate anti-tumor activities used alone or in combination with standard therapies [69,70,71]. Indeed, in the first phase II clinical trial, the use of nitroglycerin combined with vinorelbine and cisplatin improved the overall response and time to disease progression in patients with non-small-cell lung cancer (NSCLC) [69]. In a second phase II study, the addition of nitroglycerin to cisplatin and vinorelbine and concurrent radiotherapy in patients with advanced NSCLC had an acceptable toxicity profile and the possibility of adding nitroglycerin to chemotherapy and radiotherapy was supported [70]. In another phase II study conducted in patients with prostate cancer, nitroglycerin increased the prostate-specific antigen (PSA) doubling time and the safety of the drug was confirmed [71].
NO-induced survival or cell death seems to be related to its biochemical action on proteins by causing post-translational modifications [31]. S-nitrosylation, the transfer of NO to a free-SH group of a specific cysteine residue, is now considered to be essential for regulating the function of many proteins and signaling pathways including those involved in cancer regulation [72]. It modulates protein structure, function, expression, location or interaction with other protein partners [73]. The ambivalent nature of NO in cancer is dictated by the impact of S-nitrosylation on proteins involved in signaling pathways that trigger both survival and cell death. Thus, NO and its derivatives, such as peroxynitrites, are able to cause direct or indirect DNA damage. Direct damage includes DNA base deamination, adduct formation and single strand breaks in the DNA. Indirect damage is due to the interactions of NO reactive species with other molecules such as amines, thiols or lipids [74]. Furthermore, NO and its derivatives modulate SASP factors. Recently, the NO donors sodium nitroprusside dihydrate (SNP) and diethylenetriamine/nitric oxide adduct (DETA/NO) have been reported to cause DNA double-strand breaks (DSBs), initiating cellular senescence programs in numerous cell lines of different origins (cervical and lung cancers, fibroblastic cell lines). This effect is associated with the activation of the protein kinase ATM, an upstream activator of the DDR; the activation of NF-κB; and an increase in SASP factors like IL-6 and IL-8 [75]. NO can also regulate other pathways involved in the secretion of SASP factors such as NF-κB and the mitogen activated protein kinase c-Jun N-terminal kinase (JNK) pathways [76]. Indeed, Reynaert et al. (2004) have reported that NO triggers the S-nitrosylation of the inhibitor of NF-κB kinase subunit β (IKKβ, a regulator of the classical NF-κB pathway activation) at cysteine residue 179 and consequently results in NF-κB inhibition [77]. In addition, NO can S-nitrosylate both the p50 and p65 NF-κB subunits at cysteine residues 62 and 38, respectively, reducing their DNA binding and inhibiting target gene transcription [78,79]. Moreover, the tyrosine nitration of IκBα at tyrosine 181 by endogenous NO promotes NF-κB signaling through the dissociation of IκBα from NF-κB [80]. The endogenous production of NO or NO donors such S-nitro-N-acetyl-penicillamine (SNAP) can suppress JNK activation via S-nitrosylation at cysteine 116 [81]. However, the relationship of NO-induced senescence with its ability to target NF-κB and JNK has never been reported so far (Figure 1).
NO also targets some cytokines that belong to SASP factors. For instance, the NO-aspirin derivative NCX-4016 (100 mM) inhibits the release of numerous cytokines (IL-1β, IL-18, IL-8, IL-12, IFNγ and TNFα) in monocytes isolated from human PBMCs of healthy donors after challenge with the bacterial endotoxin, lipopolysaccharides (LPS) (1 μg/mL). This effect is due to the inhibition of IL-1β Converting Enzyme, also named caspase-1, required for intracellular processing/maturation of IL-1β and IL-18 [82]. These data may suggest that in the context of senescence, the NO-mediated inhibition of these pro-inflammatory cytokines could alleviate cancer development, invasion, and metastasis [83]. Furthermore, NO also targets another SASP factor, the signal transducer and activator of transcription 3 (STAT3), an important target in cancer therapy and a key kinase involved in the IL-6 signaling pathway. Indeed, STAT3 is regulated by the S-nitrosylation of the cysteine residue at position 259, which inhibits STAT3 phosphorylation, its downstream activation and affects IL-6-mediated cell proliferation [84]. As mentioned before, COX2 expression is significantly increased in senescent cells. The overexpression of PGE2 (the major COX-2 product), which functions as a key SASP factor in the tumor microenvironment, suppresses the anti-tumor immunity and progression [60]. A previous study has established that NO can modulate COX2 activity. Indeed, Kim et al. (2005) showed that iNOS specifically binds to the enzyme COX2, induces its S-nitrosylation and enhances its catalytic activity [85]. NO can also modulate immune cell recruitment through its action on chemokines, another group of SASP factors. Indeed, Giustizieri et al. (2002) have reported that the NO donor, S-nitrosoglutathione (GSNO), diminished, in a dose-dependent manner, both the mRNA and protein levels of CCL5 (RANTES) and CXCL1 (GROα) in keratinocytes cultured from healthy or psoriatic patients [86]. These results were confirmed in another cellular model by Kashiwagi et al. (2002,) which showed that CCL5 and CXCL1 are up-regulated in renal cortex of rats chronically treated with an NO synthase inhibitor, and induced the recruitment of monocytes/macrophages [87] (Figure 1).

4. NO Involvement in Expression/Activation of Death Receptors and Death Ligands

The tumor necrosis factor (TNF) ligand family members TNFα, Fas ligand (FasL) and Tumor-necrosis-factor related apoptosis ligand (TRAIL) are major immunoregulatory cytokines of the tumor microenvironment, also found to be important SASP components (Table 1) [24]. These cytokines exert paradoxical functions, either by sustaining tumor growth and chemoresistance or by killing tumor cells, in the tumor microenvironment in a context-dependent manner [88]. One anti-tumor strategy would consist of switching the roles of TNFα, FasL and TRAIL from their pro-apoptotic functions to favor tumor cell death. A growing number of studies indicate that NO can regulate the signaling pathways driven by TNFα, FasL and TRAIL at many different levels [89,90]. Several studies have indicated that NO may exert dual effects in cancer, that can lead, on one hand, to enhanced tumor growth and progression (genotoxicity, apoptosis resistance, angiogenesis, invasion and metastasis) and on the other hand, to tumoricidal effects (cytostatic and/or cytotoxic effects on tumor cells) [91,92]. The biological impact of NO relies on various aspects (e.g., the NO concentration, tumor redox microenvironment and duration of NO exposure). Thus, NO donor-based therapies are currently under investigation to further sustain the tumoricidal effect of NO.
Several reports have demonstrated the role of NO-mediated sensitization of cancer cells to apoptosis in many ways [88]. Thus, NO-based therapies could represent a new potential strategy to reduce the threshold of cancer cell resistance. Various NO donors are under investigation to understand the molecular mechanisms that underly their modes of action. It is becoming even more evident that the post-translational modifications of selective proteins by NO exert an important regulatory control, either positive or negative, over various signaling pathways engaged by TNF ligands and their receptors [88,93]. The cellular response at least relies on the relative threshold of NO production (either endogenous or from NO-releasing drugs), cellular context and severity of oxidative stress. If the biological outcome of the NO-induced S-nitrosylation of Fas [94], DR4 [95] and TNFR1 [96] results in cell death induction, the molecular mechanisms that control this process have not been fully unraveled yet. Importantly, the NO donor GTN can mediate the S-nitrosylation of Fas at cysteine 304, which consequently leads to its aggregation into lipid rafts to stimulate downstream signaling and cancer cell death [94]. In agreement, the S-nitrosylation of the transcriptional repressor Ying Yang 1 (YY1) inhibits its DNA-binding capacity to the silencer region of Fas promoter and then up-regulates FAS gene expression and consequently sensitizes cells to apoptosis [97,98].
The S-nitrosylation of TNFR1 by the NO donor NONOate has also been described in hepatoma cells; however, the exact targeted cysteine residue is unknown [96]. As previously described, the classical NF-κB pathway activated by the TNFα/TNFR1 system is controlled by the S-nitrosylation and nitration of specific target proteins at different levels of the molecular pathway. More recently, the S-nitrosylation of cIAP1 (a positive regulator of the NF-κB signaling pathway), induced by the NO donor GTN (particularly at cysteine 571), appeared as a critical cornerstone for switching the cancer cell fate from TNFα/TNFR1-mediated cell survival (through the activation of the classical NF-κB cascade) to TNFα/TNFR1-mediated cell death [90].
To date, the regulatory role of NO in the TRAIL/TRAIL receptors (TRAILR) system is less documented. Only DR4 was reported to undergo S-nitrosylation and, furthermore, by a specific NO donor (Nitrosylcobalamin), particularly at cysteine 336, and consequently foster cancer cell apoptosis [95]. Accordingly, NO disrupts the transcriptional repressor activity of YY1 not only on FAS but also on DR5, up-regulates its expression and sensitizes cancer cells to TRAIL-induced apoptosis [99].
Several crucial factors of apoptosis involved in the TNF signaling pathways, such as caspases, Bcl-2 family proteins or FLIP, can undergo post-translational modifications by NO that would impact cell fate [88,93]. Although SASP components include NO, the S-nitrosylation of Fas, DR4 and TNFR1 was demonstrated exclusively via the NO released by NO donors.
Interestingly, beside their dichotomous responses in cancer, TNF ligands, particularly TNFα and FasL, can lead to the process of senescence. Indeed, cancer cells’ senescent phenotype can arise in response to TNFα/TNF receptor 1 (TNFR1) through the activation of the p16INK4A/Rb pathway [100]. The senescent phenotype can also occur in response to FasL/Fas in a context-dependent manner, particularly in microsatellite instability-high type colon tumors. Mechanistically, Fas-induced senescence was caused by a chronic DNA damage response via caspase-activated DNAse resulting in p53 activation and p21 expression [101].
Whether the SASP could modulate the TNF ligand systems via NO, either in an autocrine or paracrine manner, remains to be demonstrated.

5. Role of NO in the SASP-Immunomodulatory Effect

Because senescent cells remain viable and exhibit the SASP phenotype with a wide spectrum of diverse physiological functions, their existence in the tumor mass can have an ambivalent impact, from tumor regression to promotion. SASP cytokines act on the recruitment and activation states of immune cells. They can cause the tumor infiltration of immunosuppressive cells like macrophages and myeloid-derived suppressor cells (MDSCs), thus promoting tumor growth. Conversely, they can also induce the tumor infiltration of natural killer cells (NK) and effector T lymphocytes and thus have anti-tumor properties [102].
In recent years, NO has emerged as an important immunomodulatory agent in the tumor microenvironment [32,103]. In situ, different sources of endogenous NO can be considered. Indeed, NO can be produced by various types of cell expressing iNOS and eNOS. Moreover, NO, as well as ROS, can also derive from senescent cells as non-macromolecular components of the SASP [24] (Table 1). It has been demonstrated that the tumor microenvironment has all the conditions for iNOS expression and NO production, which is important for the maintenance and progression of an aggressive tumor phenotype in breast cancer [104]. iNOS has an immunosuppressive role within the tumor microenvironment via its actions on MDSCs as well as via the loss of the effector function of cytotoxic T lymphocytes (CTLs). Furthermore, two studies have shown that two NO-releasing drugs—NO-aspirin and NO-aspirin derivative (AT38)—induced, in numerous types of cancer, a feedback inhibition of iNOS in MDSCs [105,106]. Such effects result in the decreased MDSC-induced nitration of T-cell receptors, a massive infiltration of the tumor by T-cells and an enhanced efficacy of DNA cancer vaccination [105,106]. Indeed, NO from iNOS-expressing tumor cells disturbs the polarization and directional secretion of cytotoxic granules in the immune synapse of tumor infiltrating lymphocytes (TILs) [107]. Nitration by iNOS-derived NO is also involved in the polarization of tumor-associated macrophages. NO leads to the suppression of the M1 macrophage signature gene activation and induces a pro-tumorigenic environment [108].
Among the SASP chemokine data for nitration, CCL2 and CXCL12 have been reported. These chemokines interact with the G-protein coupled receptors CCR2 and CXCR4, respectively, and bind to glycosaminoglycans (GAGs) present on the surface of endothelial cells and in the extracellular matrix. CCL2, also known as monocyte chemoattractant protein-1 (MCP-1), greatly contributes to the recruitment of monocytes, memory T cells and dendritic cells into sites of inflammation and tumors. It has been shown that peroxynitrite-treated CCL2 lost its ability to recruit CD8+ T cells, but the recruitment of myeloid-derived suppressor cells was unaltered [106]. Nitrated CCL2 has a reduced affinity to its receptor CCR2, which may explain its failure to induce the chemotaxis of CD8+ T cells expressing low levels of the CCR2 but the retention of its ability to induce the migration of myeloid cells expressing high levels of CCR2 [109]. CXCL12, also known as stroma cell-derived factor-1 (SDF-1), represents the single natural ligand for the chemokine receptor CXCR4 and induces the activation and migration of most leucocytes. The nitration of tyrosine 7 in CXCL12 hampers the chemokine’s ability to induce lymphocyte chemotaxis (Figure 1). This nitration of CXCL12 does not affect its ability to bind to the CXCR4 receptor but does impair its ability to signal through this receptor. If nitrated CXCL12 binds to GAGs with a similar affinity as wild type CXCL12, nitrated CCL2 has a reduced ability to bind GAGs compared to wild type CCL2, and therefore could limit further leukocyte chemotaxis. When nitration reduces receptor activation capacity without affecting receptor affinity, this influences the receptor’s signaling in situations where many chemokines can bind the same receptor [110].
NO from MDSCs has been described to suppress T lymphocyte proliferation via the suppression of STAT5 phosphorylation [111]. However, as we have underlined above, the immunosuppressive effect of MDSCs can be constrained by NO-releasing drugs [105,106]. NO is also involved in STAT3 phosphorylation through src homology protein tyrosine phosphatase 2 (SHP2) nitration, known as a negative regulator of STAT3 phosphorylation [112]. This study was conducted in a mouse model of ulcerative colitis, and the impact of NO on STAT3 phosphorylation was investigated in LPS-activated macrophages but not in a tumor context or in T lymphocytes in whose activation STAT3 plays a key role.
SASP is also involved in immune resistance mechanisms, since high levels of IFNγ drive the expression of the programmed death ligand 1 (PD-L1), one of the targets of immune checkpoint inhibitor therapy (ICT). In tumors, high expression of the transcription factor YY1 modulates PD-L1 expression. Treatment with NO donors results in the inhibition of PD-L1 expression via the S-nitrosylation of YY1 [113]. By a nitroproteomic approach, authors uncover a potential mechanism for the ICT where a key protein for T cell activation is nitrated and inactivated by MDSCs. Indeed, lymphocyte-specific protein tyrosine kinase (LCK), an initiating tyrosine kinase in the T cell receptor signaling cascade, is nitrated at tyrosine 394 by MDSCs [114]. After ICT, high expression of iNOS is observed in intratumoral myeloid cells and is dependent on IFNγ as evidenced using high dimensional profiling [115]. Thus, IFNγ drives the polarization of newly recruited monocytes to become iNOS-positive macrophages. This remodeling by ICT is also correlated with an increase in activated T cells and a decrease in Tregs. Therefore, combining ICT with reactive nitrogen species reducing agents could represent treatment strategies for ICT-resistant cancers.
Growing literature suggests that the induction of senescence in the immune compartment is also a mechanism used by the immune system to regulate the immune response. Human Tregs induce senescence in responder T cells by the regulation of STAT1/STAT3 signaling [116]. Moreover, the autologous infusion of tumor antigen specific CD4+ Th1 lymphocyte can promote senescence in pancreatic tumor cells by releasing SASP factors such as IFNγ and TNFα. Such cytokine-induced senescence strictly requires STAT1 and TNFR1 signaling pathways, themselves affected by S-nitrosylation [100].
Since NO is present in the tumor microenvironment associated with senescent cells, it seems important to precisely determine the nitration or S-nitrosylation state of molecules detected in SASP or involved in SASP signaling.
Altogether, several studies put forward arguments to show the immunosuppressive effect of NO. In most cases, these arguments are based on the immunosuppressive effect of endogenous NO produced, in small amounts, by NOSs expressed in immune cells, particularly in MDSCs. On the other hand, NO donors generating high amounts of NO can reduce this immunosuppression. Such effects may be due to the ability of these donors to induce a feedback inhibition of iNOS, a key mechanism of MDSC-mediated immunosuppression. However, few clinical trials have tested the anti-tumor potential of the agonist TLR4, known to induce the production of iNOS and NO [117,118].

6. Senolytic Drugs and NO

As discussed in the “senescence and cancer” part, senescence can play an anti- or a pro-tumorigenic role in cancer. In fact, senescent cells correspond to cells that have irreversibly lost their capability to divide but that are very resistant to apoptotic stimuli [119]. We can distinguish six Senescent-Cell Anti-Apoptotic Pathways (SCAPs): BCL-2/BCL-XL, the PI3K/AKT/ceramide metabolic network, MDM2/p53/p21/serpin elements, Ephirins/dependence receptors/tyrosine kinases, the hypoxia inducible factor (HIF-1α) pathway, and the heat shock protein 90 (HSP-90)-dependent pathway [27]. It seems as though the inhibition of these different pathways could induce apoptosis preferentially in senescent cells, which thus could delay age-associated pathologies [119]. Molecules used to induce senescent cell removal are called senolytics. To this group belong dasatinib and quercetin, BCL-2 family inhibitors, Forkhead box O 4 (FOXO4) inhibitors and others such as nicotinamide riboside, danazol, fisetin, piperlongumine and heat shock protein 90 (HSP90) inhibitors [34]. Thus far, many researchers view senolytics as a second wave of adjuvant tumor therapy, following chemotherapy or radiotherapy [120]. At this stage, links also exist between NO and senolytic drugs as described below.

6.1. Dasatinib/Quercetin

Dasatinib is a second-generation tyrosine kinase inhibitor that targets several kinases including breakpoint cluster region–protein Abelson (Bcr-Abl), c-Kit, and platelet-derived growth factor receptor A (PDGFRA) and B but also Src kinase family members. It is commonly used in the treatment of imatinib-resistant chronic myeloid leukemia patients [121,122]. Quercetin, instead, is a polyphenol compound that can be found in food like nuts, teas, vegetables and herbs and which is highlighted for its cytotoxic effects in several types of cancer without harming healthy cells [123]. Many modifications can be induced by these two senolytic drugs of NO levels. Indeed, dasatinib can induce an increase in plasma NO in pulmonary artery and smooth muscle cells [124]. Additionally, Hu et al. (2018) recently found that Src inhibition by dasatinib increases iNOS, a pro-inflammatory macrophage marker, in both intestinal and bone marrow-derived macrophages [125]. Conversely, Cruz et al. (2016) reported that dasatinib is able to reduce lung inflammation and fibrosis by promoting the polarization of macrophages from the M1 to M2 phenotype. This effect is due to the ability of dasatinib to reduce iNOS expression specifically in silicotic macrophages [126]. Concerning quercetin, all studies demonstrated that this senolytic drug induces a downregulation of iNOS expression, thus inhibiting nitric oxide production in vivo and in vitro (Table 2) [127,128,129,130,131,132,133].

6.2. Bcl-2 Inhibitors

The Bcl2 family is composed of a set of proteins that play an important role in promoting (Bax, Bak, Bid and Bim) but also in inhibiting (Bcl-2, Bcl-xL, and Bcl-W) apoptosis. Thus, the overexpression of anti-apoptotic members such as Bcl-2 or the downregulation or mutation of pro-apoptotic ones may be the cause of acquired resistance to apoptosis and cancer development [136]. Targeting apoptotic inhibitors represents, then, an efficient and promising cancer treatment. Post-translational modifications induced by NO on Bcl-2 have been reported. Indeed, Wright et al. (2016) revealed that Bcl-2 can be S-nitrosylated with NO donor DPTA-NONOate, thus inducing resistance to autophagy initiation in malignantly transformed lung cells [137].

6.3. Hsp90 Inhibitors

Hsp90 is a chaperone protein that assists the regulation, the folding and the stabilization/degradation of multiple proteins involved in multiple signaling pathways [138]. Proteins involved in tumor growth (EGFR, HER2, BRAF, Akt, etc.) can also be stabilized by this chaperone protein, leading to cancer development [139]. Hsp90 inhibitors are thus highlighted as anti-cancer drugs. In 1998, Garcia-Cardena et al. reported that Hsp90 can interact with eNOS and enhance its activity to induce NO production [140]. This activation seems to be induced by Hsp90 and Akt [134]. Indeed, it has been demonstrated that the Hsp90-dependent phosphorylation of eNOS at serine 1177 (human eNOS) or 1179 (bovine eNOS) is a key post-translational modification in the initiation of eNOS activation and NO synthesis [141]. The inhibition of Hsp90 by Geldanamycin [134] or taxotere [135] induced a marked reduction in eNOS activity (Table 2). eNOS can also be responsible for post-translational modifications of Hsp90 and further inhibiting eNOS activation. Martínez-Ruiz et al. (2005) were the first to determine that human Hsp90 can be S-nitrosylated at cysteine 597 in endothelial cells, which inhibits its ATPase activity in vitro [142] and in vivo [143].
In order to improve anti-cancer treatments, new combination therapies could be tested to successively induce the therapy-induced senescence (TIS) and the subsequent elimination of senescent cells by the immune system (senotherapy). Indeed, Xue et al. (2007) have shown in a p53 rescue experiment that innate immune cells such as macrophages, neutrophils and NK cells are involved in senescent cell removal [144]. Adaptive immune cells such as the Th1 subclass of CD4+ cells are also involved in immune-mediated clearance in a model of OIS in murine hepatocytes [145].
More recently, the combination of senescence-inducing chemotherapy (oxaliplatin or cisplatin) with a TLR4 agonist, the lipid A OM-174, has demonstrated strong anti-tumor efficacy in a model of advanced colon carcinoma [146]. The anti-tumor efficacy of this Lipid A has previously been demonstrated in several models of tumors [147,148,149] and was dependent on cytokine secretion (IFNγ, IL-1β and TNFα), iNOS activation and tumor-associated neutrophil (TAN) reprogramming into anti-tumorigenic N1 [149,150]. Moreover, the safety and tolerance at biologically active concentrations of the lipid A compound have been demonstrated in a phase I clinical trial in patients with cancers of different origins [117]. However, in our animal model with advanced colon carcinoma, lipid A—when used alone (as well as with platinum drugs)—failed to induce tumor regression, while pretreatment with oxaliplatin or cisplatin followed by lipid A injections induced a large regression of colorectal tumors [146]. The anti-tumor effect of this combination is correlated with a sequential induction of (1) cancer cell senescence and the recruitment of pro-tumorigenic TAN (N2), induced by platinum derivative drugs; and (2) apoptosis and N1 TAN recruitment triggered by lipid A.

7. Conclusions

In the fight against cancer, the search for new therapeutic combinations remains a major challenge. Senescence and, more particularly, SASP are major targets of these treatments. Indeed, many treatments currently used in anti-cancer therapies induce senescence, which is called therapy-induced senescence (TIS). These treatments include docetaxel, bleomycin, cyclophosphamide, doxorubicin, vincristine, etoposide and oxaliplatin [48,146] but also ionizing radiation [34]. However, senescence and, more particularly, SASP have an early anti-tumorigenic effect whereas in the long term, they will have a pro-tumorigenic role via their autocrine and paracrine actions [151]. In order to avoid the negative side effects of SASP after TIS, a new type of therapy called senotherapy has emerged to either counteract these SASP effects or to specifically eliminate senescent cells. The first strategy consists of counteracting the pro-tumorigenic effects of SASP. This can be achieved with the use of telomerase inhibitors, the therapeutic modulation of the cell cycle, p53 and myc targeting, the use of immunotherapy that targets MDSCs, and the reprogramming of SASP [34]. In this review, we highlighted NO as an innovative approach to modulate SASP effects. Even though NO has been described to induce the senescence of tumor cells [75], it also mainly modulates the expression of SASP factors, as well as other senomorphic agents [34], by regulating numerous signaling pathways such as NF-κB and STAT3 [84,93]. The second therapeutic strategy consists of inducing senescence and subsequently clearing the senescent cells either specifically, by inducing their apoptosis, or by activating the immune system. The use of NO donors as senotherapy seems to be a promising solution since they are described to potentiate the apoptosis induced by death ligands such as FasL, TNFα and TRAIL. In addition, several clinical trials have shown the advantage of using an NO donor such as GTN in combination with TIS agents such as vincristine, cisplatin and radiotherapy in the treatment of non-small cell lung cancer [69,71]. NO, through its immunomodulatory potential, can also be used to eliminate senescent cells. Thus, the combination of TIS agents such as oxaliplatin and cisplatin with an immunomodulatory agent such as lipid A has significant anti-tumor efficacy in an advanced cancer model [146]. Overall, the recent clinical trials that involved either TLR4 agonists [117,118] or NO donors [69,70,71] suggest that these therapies, used alone or in combination with TIS, might be beneficial to treat cancer. These results prompt us to reconsider the commonly accepted pro-tumorigenic effect of NO based on its production by immunosuppressive MDSCs. Further studies will be needed to better understand the pathophysiological conditions that determine whether NO will tip the balance to tumor progression or cancer cure.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge the financial support from Région Bourgogne, La Ligue Contre le Cancer (Conférence de coordination Interrégionale Est) and Qatar Foundation, but also acknowledge Olivier Burgy for manuscript editing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Davaapil, H.; Brockes, J.P.; Yun, M.H. Conserved and novel functions of programmed cellular senescence during vertebrate development. Development 2017, 144, 106–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Lee, J.S. Cellular senescence, aging, and age-related disease: Special issue of BMB Reports in 2019. BMB Rep. 2019, 52, 1–2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Cerella, C.; Grandjenette, C.; Dicato, M.; Diederich, M. Roles of Apoptosis and Cellular Senescence in Cancer and Aging. Curr. Drug Targets 2016, 17, 405–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Fane, M.; Weeraratna, A.T. How the ageing microenvironment influences tumour progression. Nat. Rev. Cancer 2020, 20, 89–106. [Google Scholar] [CrossRef] [PubMed]
  5. Aunan, J.R.; Cho, W.C.; Søreide, K. The Biology of Aging and Cancer: A Brief Overview of Shared and Divergent Molecular Hallmarks. Aging Dis. 2017, 8, 628–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
  7. Harley, C.B.; Futcher, A.B.; Greider, C.W. Telomeres shorten during ageing of human fibroblasts. Nature 1990, 345, 458–460. [Google Scholar] [CrossRef]
  8. Campisi, J.; d’Adda di Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef]
  9. Muñoz-Espín, D.; Serrano, M. Cellular senescence: From physiology to pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496. [Google Scholar] [CrossRef]
  10. Serrano, M.; Lin, A.W.; McCurrach, M.E.; Beach, D.; Lowe, S.W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 1997, 88, 593–602. [Google Scholar] [CrossRef] [Green Version]
  11. Gorgoulis, V.G.; Halazonetis, T.D. Oncogene-induced senescence: The bright and dark side of the response. Curr. Opin. Cell Biol. 2010, 22, 816–827. [Google Scholar] [CrossRef] [PubMed]
  12. Di Micco, R.; Fumagalli, M.; Cicalese, A.; Piccinin, S.; Gasparini, P.; Luise, C.; Schurra, C.; Garre, M.; Nuciforo, P.G.; Bensimon, A.; et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 2006, 444, 638–642. [Google Scholar] [CrossRef] [PubMed]
  13. Bartkova, J.; Rezaei, N.; Liontos, M.; Karakaidos, P.; Kletsas, D.; Issaeva, N.; Vassiliou, L.V.; Kolettas, E.; Niforou, K.; Zoumpourlis, V.C.; et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 2006, 444, 633–637. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, Z.; Trotman, L.C.; Shaffer, D.; Lin, H.K.; Dotan, Z.A.; Niki, M.; Koutcher, J.A.; Scher, H.I.; Ludwig, T.; Gerald, W.; et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 2005, 436, 725–730. [Google Scholar] [CrossRef] [Green Version]
  15. Alimonti, A.; Nardella, C.; Chen, Z.; Clohessy, J.G.; Carracedo, A.; Trotman, L.C.; Cheng, K.; Varmeh, S.; Kozma, S.C.; Thomas, G.; et al. A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis. J. Clin. Investig. 2010, 120, 681–693. [Google Scholar] [CrossRef]
  16. Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of Cellular Senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef]
  17. Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereira-Smith, O. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef] [Green Version]
  18. Michaloglou, C.; Vredeveld, L.C.; Soengas, M.S.; Denoyelle, C.; Kuilman, T.; van der Horst, C.M.; Majoor, D.M.; Shay, J.W.; Mooi, W.J.; Peeper, D.S. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 2005, 436, 720–724. [Google Scholar] [CrossRef]
  19. Braig, M.; Lee, S.; Loddenkemper, C.; Rudolph, C.; Peters, A.H.; Schlegelberger, B.; Stein, H.; Dörken, B.; Jenuwein, T.; Schmitt, C.A. Oncogene-induced senescence as an initial barrier in lymphoma development. Nature 2005, 436, 660–665. [Google Scholar] [CrossRef]
  20. Collado, M.; Gil, J.; Efeyan, A.; Guerra, C.; Schuhmacher, A.J.; Barradas, M.; Benguría, A.; Zaballos, A.; Flores, J.M.; Barbacid, M.; et al. Tumour biology: Senescence in premalignant tumours. Nature 2005, 436, 642. [Google Scholar] [CrossRef]
  21. Krtolica, A.; Parrinello, S.; Lockett, S.; Desprez, P.Y.; Campisi, J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proc. Natl. Acad. Sci. USA 2001, 98, 12072–12077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Milanovic, M.; Fan, D.N.Y.; Belenki, D.; Däbritz, J.H.M.; Zhao, Z.; Yu, Y.; Dörr, J.R.; Dimitrova, L.; Lenze, D.; Monteiro Barbosa, I.A.; et al. Senescence-associated reprogramming promotes cancer stemness. Nature 2018, 553, 96–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.W.; Lasitschka, F.; Andrulis, M.; et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 2013, 15, 978–990. [Google Scholar] [CrossRef] [PubMed]
  24. Coppé, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking senescence: Context-dependent effects of SASP in cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef] [PubMed]
  26. Sun, Y.; Coppé, J.P.; Lam, E.W. Cellular Senescence: The Sought or the Unwanted? Trends Mol. Med. 2018, 24, 871–885. [Google Scholar] [CrossRef] [PubMed]
  27. Sikora, E.; Bielak-Zmijewska, A.; Mosieniak, G. Targeting normal and cancer senescent cells as a strategy of senotherapy. Ageing Res. Rev. 2019, 55, 100941. [Google Scholar] [CrossRef]
  28. Laberge, R.M.; Sun, Y.; Orjalo, A.V.; Patil, C.K.; Freund, A.; Zhou, L.; Curran, S.C.; Davalos, A.R.; Wilson-Edell, K.A.; Liu, S.; et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol. 2015, 17, 1049–1061. [Google Scholar] [CrossRef]
  29. Herranz, N.; Gallage, S.; Mellone, M.; Wuestefeld, T.; Klotz, S.; Hanley, C.J.; Raguz, S.; Acosta, J.C.; Innes, A.J.; Banito, A.; et al. Erratum: mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat. Cell Biol. 2015, 17, 1370. [Google Scholar] [CrossRef]
  30. Fuhrmann-Stroissnigg, H.; Ling, Y.Y.; Zhao, J.; McGowan, S.J.; Zhu, Y.; Brooks, R.W.; Grassi, D.; Gregg, S.Q.; Stripay, J.L.; Dorronsoro, A.; et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat. Commun. 2017, 8, 422. [Google Scholar] [CrossRef]
  31. Leon, L.; Jeannin, J.F.; Bettaieb, A. Post-translational modifications induced by nitric oxide (NO): Implication in cancer cells apoptosis. Nitric Oxide 2008, 19, 77–83. [Google Scholar] [CrossRef] [PubMed]
  32. Bogdan, C. Nitric oxide synthase in innate and adaptive immunity: An update. Trends Immunol. 2015, 36, 161–178. [Google Scholar] [CrossRef] [PubMed]
  33. Tchkonia, T.; Zhu, Y.; van Deursen, J.; Campisi, J.; Kirkland, J.L. Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J. Clin. Investig. 2013, 123, 966–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Calcinotto, A.; Kohli, J.; Zagato, E.; Pellegrini, L.; Demaria, M.; Alimonti, A. Cellular Senescence: Aging, Cancer, and Injury. Physiol. Rev. 2019, 99, 1047–1078. [Google Scholar] [CrossRef]
  35. Kaur, A.; Ecker, B.L.; Douglass, S.M.; Kugel, C.H.; Webster, M.R.; Almeida, F.V.; Somasundaram, R.; Hayden, J.; Ban, E.; Ahmadzadeh, H.; et al. Remodeling of the Collagen Matrix in Aging Skin Promotes Melanoma Metastasis and Affects Immune Cell Motility. Cancer Discov. 2019, 9, 64–81. [Google Scholar] [CrossRef] [Green Version]
  36. Lopes-Paciencia, S.; Saint-Germain, E.; Rowell, M.C.; Ruiz, A.F.; Kalegari, P.; Ferbeyre, G. The senescence-associated secretory phenotype and its regulation. Cytokine 2019, 117, 15–22. [Google Scholar] [CrossRef]
  37. Zeng, S.; Shen, W.H.; Liu, L. Senescence and Cancer. Cancer Transl. Med. 2018, 4, 70–74. [Google Scholar] [CrossRef]
  38. Reimann, M.; Lee, S.; Loddenkemper, C.; Dörr, J.R.; Tabor, V.; Aichele, P.; Stein, H.; Dörken, B.; Jenuwein, T.; Schmitt, C.A. Tumor stroma-derived TGF-beta limits myc-driven lymphomagenesis via Suv39h1-dependent senescence. Cancer Cell 2010, 17, 262–272. [Google Scholar] [CrossRef] [Green Version]
  39. Alessio, N.; Aprile, D.; Squillaro, T.; Di Bernardo, G.; Finicelli, M.; Melone, M.A.; Peluso, G.; Galderisi, U. The senescence-associated secretory phenotype (SASP) from mesenchymal stromal cells impairs growth of immortalized prostate cells but has no effect on metastatic prostatic cancer cells. Aging (Albany N.Y.) 2019, 11, 5817–5828. [Google Scholar] [CrossRef]
  40. Loo, T.M.; Miyata, K.; Tanaka, Y.; Takahashi, A. Cellular senescence and senescence-associated secretory phenotype via the cGAS-STING signaling pathway in cancer. Cancer Sci. 2020, 111, 304–311. [Google Scholar] [CrossRef] [Green Version]
  41. Glück, S.; Ablasser, A. Innate immunosensing of DNA in cellular senescence. Curr. Opin. Immunol. 2019, 56, 31–36. [Google Scholar] [CrossRef] [PubMed]
  42. Barber, G.N. STING: Infection, inflammation and cancer. Nat. Rev. Immunol. 2015, 15, 760–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Salcedo, R.; Cataisson, C.; Hasan, U.; Yuspa, S.H.; Trinchieri, G. MyD88 and its divergent toll in carcinogenesis. Trends Immunol. 2013, 34, 379–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Swann, J.B.; Vesely, M.D.; Silva, A.; Sharkey, J.; Akira, S.; Schreiber, R.D.; Smyth, M.J. Demonstration of inflammation-induced cancer and cancer immunoediting during primary tumorigenesis. Proc. Natl. Acad. Sci. USA 2008, 105, 652–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ahn, J.; Xia, T.; Konno, H.; Konno, K.; Ruiz, P.; Barber, G.N. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 2014, 5, 5166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Takahashi, A.; Loo, T.M.; Okada, R.; Kamachi, F.; Watanabe, Y.; Wakita, M.; Watanabe, S.; Kawamoto, S.; Miyata, K.; Barber, G.N.; et al. Downregulation of cytoplasmic DNases is implicated in cytoplasmic DNA accumulation and SASP in senescent cells. Nat. Commun. 2018, 9, 1249. [Google Scholar] [CrossRef] [PubMed]
  47. Kitajima, S.; Ivanova, E.; Guo, S.; Yoshida, R.; Campisi, M.; Sundararaman, S.K.; Tange, S.; Mitsuishi, Y.; Thai, T.C.; Masuda, S.; et al. Suppression of STING Associated with LKB1 Loss in KRAS-Driven Lung Cancer. Cancer Discov. 2019, 9, 34–45. [Google Scholar] [CrossRef] [Green Version]
  48. Ewald, J.A.; Desotelle, J.A.; Wilding, G.; Jarrard, D.F. Therapy-induced senescence in cancer. J. Natl. Cancer Inst. 2010, 102, 1536–1546. [Google Scholar] [CrossRef] [Green Version]
  49. Tato-Costa, J.; Casimiro, S.; Pacheco, T.; Pires, R.; Fernandes, A.; Alho, I.; Pereira, P.; Costa, P.; Castelo, H.B.; Ferreira, J.; et al. Therapy-Induced Cellular Senescence Induces Epithelial-to-Mesenchymal Transition and Increases Invasiveness in Rectal Cancer. Clin. Colorectal. Cancer 2016, 15, 170–178.e3. [Google Scholar] [CrossRef]
  50. Sato, I.; Morita, I.; Kaji, K.; Ikeda, M.; Nagao, M.; Murota, S. Reduction of nitric oxide producing activity associated with in vitro aging in cultured human umbilical vein endothelial cell. Biochem. Biophys. Res. Commun. 1993, 195, 1070–1076. [Google Scholar] [CrossRef]
  51. MacMicking, J.; Xie, Q.W.; Nathan, C. Nitric oxide and macrophage function. Annu. Rev. Immunol. 1997, 15, 323–350. [Google Scholar] [CrossRef] [PubMed]
  52. Cassetta, L.; Pollard, J.W. Targeting macrophages: Therapeutic approaches in cancer. Nat. Rev. Drug Discov. 2018, 17, 887–904. [Google Scholar] [CrossRef] [PubMed]
  53. Bettaieb, A.; Plenchette, S.; Paul, C.; Laurens, V.; Romagny, S.; Jeannin, J.F. S-nitrosylationof cancer cells. In Nitric Oxide and Cancer: Pathogenesis and Therapy; Springer International Publishing: Cham, Switzerland, 2015; pp. 97–109. [Google Scholar]
  54. Hari, P.; Millar, F.R.; Tarrats, N.; Birch, J.; Quintanilla, A.; Rink, C.J.; Fernández-Duran, I.; Muir, M.; Finch, A.J.; Brunton, V.G.; et al. The innate immune sensor Toll-like receptor 2 controls the senescence-associated secretory phenotype. Sci. Adv. 2019, 5, eaaw0254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Salminen, A.; Kauppinen, A.; Kaarniranta, K. Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal. 2012, 24, 835–845. [Google Scholar] [CrossRef] [Green Version]
  56. Wajapeyee, N.; Serra, R.W.; Zhu, X.; Mahalingam, M.; Green, M.R. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell 2008, 132, 363–374. [Google Scholar] [CrossRef] [Green Version]
  57. Kuilman, T.; Michaloglou, C.; Vredeveld, L.C.; Douma, S.; van Doorn, R.; Desmet, C.J.; Aarden, L.A.; Mooi, W.J.; Peeper, D.S. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 2008, 133, 1019–1031. [Google Scholar] [CrossRef] [Green Version]
  58. Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef] [Green Version]
  59. Odagiri, N.; Matsubara, T.; Higuchi, M.; Takada, S.; Urushima, H.; Sato-Matsubara, M.; Teranishi, Y.; Yoshizato, K.; Kawada, N.; Ikeda, K. Involvement of ERK1/2 activation in the gene expression of senescence-associated secretory factors in human hepatic stellate cells. Mol. Cell Biochem. 2019, 455, 7–19. [Google Scholar] [CrossRef]
  60. Loo, T.M.; Kamachi, F.; Watanabe, Y.; Yoshimoto, S.; Kanda, H.; Arai, Y.; Nakajima-Takagi, Y.; Iwama, A.; Koga, T.; Sugimoto, Y.; et al. Gut Microbiota Promotes Obesity-Associated Liver Cancer through PGE. Cancer Discov. 2017, 7, 522–538. [Google Scholar] [CrossRef] [Green Version]
  61. Mishra, D.; Patel, V.; Banerjee, D. Nitric Oxide and S-Nitrosylation in Cancers: Emphasis on Breast Cancer. Breast. Cancer (Auckl.) 2020, 14, 1178223419882688. [Google Scholar] [CrossRef]
  62. Kamm, A.; Przychodzen, P.; Kuban-Jankowska, A.; Jacewicz, D.; Dabrowska, A.M.; Nussberger, S.; Wozniak, M.; Gorska-Ponikowska, M. Nitric oxide and its derivatives in the cancer battlefield. Nitric Oxide 2019, 93, 102–114. [Google Scholar] [CrossRef] [PubMed]
  63. Gruetter, C.A.; Barry, B.K.; McNamara, D.B.; Gruetter, D.Y.; Kadowitz, P.J.; Ignarro, L. Relaxation of bovine coronary artery and activation of coronary arterial guanylate cyclase by nitric oxide, nitroprusside and a carcinogenic nitrosoamine. J. Cyclic Nucleotide Res. 1979, 5, 211–224. [Google Scholar] [PubMed]
  64. Gruetter, C.A.; Barry, B.K.; McNamara, D.B.; Kadowitz, P.J.; Ignarro, L.J. Coronary arterial relaxation and guanylate cyclase activation by cigarette smoke, N’-nitrosonornicotine and nitric oxide. J. Pharmacol. Exp. Ther. 1980, 214, 9–15. [Google Scholar] [PubMed]
  65. Gruetter, C.A.; Gruetter, D.Y.; Lyon, J.E.; Kadowitz, P.J.; Ignarro, L.J. Relationship between cyclic guanosine 3’:5’-monophosphate formation and relaxation of coronary arterial smooth muscle by glyceryl trinitrate, nitroprusside, nitrite and nitric oxide: Effects of methylene blue and methemoglobin. J. Pharmacol. Exp. Ther. 1981, 219, 181–186. [Google Scholar]
  66. Jeannin, J.F.; Leon, L.; Cortier, M.; Sassi, N.; Paul, C.; Bettaieb, A. Nitric oxide-induced resistance or sensitization to death in tumor cells. Nitric Oxide 2008, 19, 158–163. [Google Scholar] [CrossRef]
  67. Bonavida, B.; Baritaki, S. Dual role of NO donors in the reversal of tumor cell resistance and EMT: Downregulation of the NF-κB/Snail/YY1/RKIP circuitry. Nitric Oxide 2011, 24, 1–7. [Google Scholar] [CrossRef]
  68. Ridnour, L.A.; Thomas, D.D.; Donzelli, S.; Espey, M.G.; Roberts, D.D.; Wink, D.A.; Isenberg, J.S. The biphasic nature of nitric oxide responses in tumor biology. Antioxid. Redox Signal. 2006, 8, 1329–1337. [Google Scholar] [CrossRef]
  69. Yasuda, H.; Yamaya, M.; Nakayama, K.; Sasaki, T.; Ebihara, S.; Kanda, A.; Asada, M.; Inoue, D.; Suzuki, T.; Okazaki, T.; et al. Randomized phase II trial comparing nitroglycerin plus vinorelbine and cisplatin with vinorelbine and cisplatin alone in previously untreated stage IIIB/IV non-small-cell lung cancer. J. Clin. Oncol. 2006, 24, 688–694. [Google Scholar] [CrossRef]
  70. Siemens, D.R.; Heaton, J.P.; Adams, M.A.; Kawakami, J.; Graham, C.H. Phase II study of nitric oxide donor for men with increasing prostate-specific antigen level after surgery or radiotherapy for prostate cancer. Urology 2009, 74, 878–883. [Google Scholar] [CrossRef]
  71. Arrieta, O.; Blake, M.; de la Mata-Moya, M.D.; Corona, F.; Turcott, J.; Orta, D.; Alexander-Alatorre, J.; Gallardo-Rincón, D. Phase II study. Concurrent chemotherapy and radiotherapy with nitroglycerin in locally advanced non-small cell lung cancer. Radiother. Oncol. 2014, 111, 311–315. [Google Scholar] [CrossRef]
  72. Stamler, J.S.; Lamas, S.; Fang, F.C. Nitrosylation. the prototypic redox-based signaling mechanism. Cell 2001, 106, 675–683. [Google Scholar] [CrossRef] [Green Version]
  73. Hess, D.T.; Stamler, J.S. Regulation by S-nitrosylation of protein post-translational modification. J. Biol. Chem. 2012, 287, 4411–4418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Felley-Bosco, E. Role of nitric oxide in genotoxicity: Implication for carcinogenesis. Cancer Metastasis Rev. 1998, 17, 25–37. [Google Scholar] [CrossRef] [PubMed]
  75. Bagheri, M.; Nair, R.R.; Singh, K.K.; Saini, D.K. ATM-ROS-iNOS axis regulates nitric oxide mediated cellular senescence. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 177–190. [Google Scholar] [CrossRef] [PubMed]
  76. Picco, V.; Pagès, G. Linking JNK Activity to the DNA Damage Response. Genes Cancer 2013, 4, 360–368. [Google Scholar] [CrossRef] [Green Version]
  77. Reynaert, N.L.; Ckless, K.; Korn, S.H.; Vos, N.; Guala, A.S.; Wouters, E.F.; van der Vliet, A.; Janssen-Heininger, Y.M. Nitric oxide represses inhibitory kappaB kinase through S-nitrosylation. Proc. Natl. Acad. Sci. USA 2004, 101, 8945–8950. [Google Scholar] [CrossRef] [Green Version]
  78. Kelleher, Z.T.; Matsumoto, A.; Stamler, J.S.; Marshall, H.E. NOS2 regulation of NF-kappaB by S-nitrosylation of p65. J. Biol. Chem. 2007, 282, 30667–30672. [Google Scholar] [CrossRef] [Green Version]
  79. Marshall, H.E.; Stamler, J.S. Inhibition of NF-kappa B by S-nitrosylation. Biochemistry 2001, 40, 1688–1693. [Google Scholar] [CrossRef]
  80. Yakovlev, V.A.; Barani, I.J.; Rabender, C.S.; Black, S.M.; Leach, J.K.; Graves, P.R.; Kellogg, G.E.; Mikkelsen, R.B. Tyrosine nitration of IkappaBalpha: A novel mechanism for NF-kappaB activation. Biochemistry 2007, 46, 11671–11683. [Google Scholar] [CrossRef] [Green Version]
  81. Park, H.S.; Park, E.; Kim, M.S.; Ahn, K.; Kim, I.Y.; Choi, E.J. Selenite inhibits the c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) through a thiol redox mechanism. J. Biol. Chem. 2000, 275, 2527–2531. [Google Scholar] [CrossRef] [Green Version]
  82. Fiorucci, S.; Santucci, L.; Cirino, G.; Mencarelli, A.; Familiari, L.; Soldato, P.D.; Morelli, A. IL-1 beta converting enzyme is a target for nitric oxide-releasing aspirin: New insights in the antiinflammatory mechanism of nitric oxide-releasing nonsteroidal antiinflammatory drugs. J. Immunol. 2000, 165, 5245–5254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Wang, S.; Song, Y.; Liu, D. EAI045: The fourth-generation EGFR inhibitor overcoming T790M and C797S resistance. Cancer Lett. 2017, 385, 51–54. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, J.; Won, J.S.; Singh, A.K.; Sharma, A.K.; Singh, I. STAT3 regulation by S-nitrosylation: Implication for inflammatory disease. Antioxid. Redox Signal. 2014, 20, 2514–2527. [Google Scholar] [CrossRef] [PubMed]
  85. Kim, S.F.; Huri, D.A.; Snyder, S.H. Inducible nitric oxide synthase binds, S-nitrosylates, and activates cyclooxygenase-2. Science 2005, 310, 1966–1970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Giustizieri, M.L.; Albanesi, C.; Scarponi, C.; De Pità, O.; Girolomoni, G. Nitric oxide donors suppress chemokine production by keratinocytes in vitro and in vivo. Am. J. Pathol. 2002, 161, 1409–1418. [Google Scholar] [CrossRef] [Green Version]
  87. Kashiwagi, M.; Masutani, K.; Shinozaki, M.; Hirakata, H. MCP-1 and RANTES are expressed in renal cortex of rats chronically treated with nitric oxide synthase inhibitor. Involvement in macrophage and monocyte recruitment. Nephron 2002, 92, 165–173. [Google Scholar] [CrossRef]
  88. Plenchette, S.; Romagny, S.; Laurens, V.; Bettaieb, A. NO and cancer: Itinerary of a double agent. Med. Sci. (Paris) 2016, 32, 625–633. [Google Scholar] [CrossRef] [Green Version]
  89. Plenchette, S. Role Of S-Nitrosylation In The Extrinsic Apoptotic Signalling Pathway In Cancer. Redox Biol. 2015, 5, 415. [Google Scholar] [CrossRef] [Green Version]
  90. Romagny, S.; Bouaouiche, S.; Lucchi, G.; Ducoroy, P.; Bertoldo, J.B.; Terenzi, H.; Bettaieb, A.; Plenchette, S. S-Nitrosylation of cIAP1 Switches Cancer Cell Fate from TNFα/TNFR1-Mediated Cell Survival to Cell Death. Cancer Res. 2018, 78, 1948–1957. [Google Scholar] [CrossRef] [Green Version]
  91. Choudhari, S.K.; Chaudhary, M.; Bagde, S.; Gadbail, A.R.; Joshi, V. Nitric oxide and cancer: A review. World J. Surg. Oncol. 2013, 11, 118. [Google Scholar] [CrossRef] [Green Version]
  92. Cheng, H.; Wang, L.; Mollica, M.; Re, A.T.; Wu, S.; Zuo, L. Nitric oxide in cancer metastasis. Cancer Lett. 2014, 353, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Plenchette, S.; Romagny, S.; Laurens, V.; Bettaieb, A. S-Nitrosylation in TNF superfamily signaling pathway: Implication in cancer. Redox Biol. 2015, 6, 507–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Leon-Bollotte, L.; Subramaniam, S.; Cauvard, O.; Plenchette-Colas, S.; Paul, C.; Godard, C.; Martinez-Ruiz, A.; Legembre, P.; Jeannin, J.F.; Bettaieb, A. S-nitrosylation of the death receptor fas promotes fas ligand-mediated apoptosis in cancer cells. Gastroenterology 2011, 140, 2009–2018. [Google Scholar] [CrossRef] [PubMed]
  95. Tang, Z.; Bauer, J.A.; Morrison, B.; Lindner, D.J. Nitrosylcobalamin promotes cell death via S nitrosylation of Apo2L/TRAIL receptor DR4. Mol. Cell Biol. 2006, 26, 5588–5594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rodríguez-Hernández, A.; Navarro-Villarán, E.; González, R.; Pereira, S.; Soriano-De Castro, L.B.; Sarrias-Giménez, A.; Barrera-Pulido, L.; Álamo-Martínez, J.M.; Serrablo-Requejo, A.; Blanco-Fernández, G.; et al. Regulation of cell death receptor S-nitrosylation and apoptotic signaling by Sorafenib in hepatoblastoma cells. Redox Biol. 2015, 6, 174–182. [Google Scholar] [CrossRef] [Green Version]
  97. Hongo, F.; Garban, H.; Huerta-Yepez, S.; Vega, M.; Jazirehi, A.R.; Mizutani, Y.; Miki, T.; Bonavida, B. Inhibition of the transcription factor Yin Yang 1 activity by S-nitrosation. Biochem. Biophys. Res. Commun. 2005, 336, 692–701. [Google Scholar] [CrossRef]
  98. Garbán, H.J.; Bonavida, B. Nitric oxide inhibits the transcription repressor Yin-Yang 1 binding activity at the silencer region of the Fas promoter: A pivotal role for nitric oxide in the up-regulation of Fas gene expression in human tumor cells. J. Immunol. 2001, 167, 75–81. [Google Scholar] [CrossRef] [Green Version]
  99. Huerta-Yepez, S.; Vega, M.; Escoto-Chavez, S.E.; Murdock, B.; Sakai, T.; Baritaki, S.; Bonavida, B. Nitric oxide sensitizes tumor cells to TRAIL-induced apoptosis via inhibition of the DR5 transcription repressor Yin Yang 1. Nitric Oxide 2009, 20, 39–52. [Google Scholar] [CrossRef]
  100. Braumüller, H.; Wieder, T.; Brenner, E.; Aßmann, S.; Hahn, M.; Alkhaled, M.; Schilbach, K.; Essmann, F.; Kneilling, M.; Griessinger, C.; et al. T-helper-1-cell cytokines drive cancer into senescence. Nature 2013, 494, 361–365. [Google Scholar] [CrossRef] [Green Version]
  101. Raats, D.A.; Frenkel, N.; van Schelven, S.J.; Rinkes, I.H.; Laoukili, J.; Kranenburg, O. CD95 ligand induces senescence in mismatch repair-deficient human colon cancer via chronic caspase-mediated induction of DNA damage. Cell Death Dis. 2017, 8, e2669. [Google Scholar] [CrossRef] [Green Version]
  102. Burton, D.G.A.; Stolzing, A. Cellular senescence: Immunosurveillance and future immunotherapy. Ageing Res. Rev. 2018, 43, 17–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. PeÑarando, J.; Aranda, E.; RodrÍguez-Ariza, A. Immunomodulatory roles of nitric oxide in cancer: Tumor microenvironment says “NO” to antitumor immune response. Transl Res. 2019, 210, 99–108. [Google Scholar] [CrossRef] [PubMed]
  104. Heinecke, J.L.; Ridnour, L.A.; Cheng, R.Y.; Switzer, C.H.; Lizardo, M.M.; Khanna, C.; Glynn, S.A.; Hussain, S.P.; Young, H.A.; Ambs, S.; et al. Tumor microenvironment-based feed-forward regulation of NOS2 in breast cancer progression. Proc. Natl. Acad. Sci. USA 2014, 111, 6323–6328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. De Santo, C.; Serafini, P.; Marigo, I.; Dolcetti, L.; Bolla, M.; Del Soldato, P.; Melani, C.; Guiducci, C.; Colombo, M.P.; Iezzi, M.; et al. Nitroaspirin corrects immune dysfunction in tumor-bearing hosts and promotes tumor eradication by cancer vaccination. Proc. Natl. Acad. Sci. USA 2005, 102, 4185–4190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Molon, B.; Ugel, S.; Del Pozzo, F.; Soldani, C.; Zilio, S.; Avella, D.; De Palma, A.; Mauri, P.; Monegal, A.; Rescigno, M.; et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011, 208, 1949–1962. [Google Scholar] [CrossRef] [PubMed]
  107. Bronte, V.; Kasic, T.; Gri, G.; Gallana, K.; Borsellino, G.; Marigo, I.; Battistini, L.; Iafrate, M.; Prayer-Galetti, T.; Pagano, F.; et al. Boosting antitumor responses of T lymphocytes infiltrating human prostate cancers. J. Exp. Med. 2005, 201, 1257–1268. [Google Scholar] [CrossRef]
  108. Lu, G.; Zhang, R.; Geng, S.; Peng, L.; Jayaraman, P.; Chen, C.; Xu, F.; Yang, J.; Li, Q.; Zheng, H.; et al. Myeloid cell-derived inducible nitric oxide synthase suppresses M1 macrophage polarization. Nat. Commun. 2015, 6, 6676. [Google Scholar] [CrossRef]
  109. Thompson, S.; Martínez-Burgo, B.; Sepuru, K.M.; Rajarathnam, K.; Kirby, J.A.; Sheerin, N.S.; Ali, S. Regulation of Chemokine Function: The Roles of GAG-Binding and Post-Translational Nitration. Int. J. Mol. Sci. 2017, 18, 1692. [Google Scholar] [CrossRef]
  110. Janssens, R.; Mortier, A.; Boff, D.; Vanheule, V.; Gouwy, M.; Franck, C.; Larsen, O.; Rosenkilde, M.M.; Van Damme, J.; Amaral, F.A.; et al. Natural nitration of CXCL12 reduces its signaling capacity and chemotactic activity in vitro and abrogates intra-articular lymphocyte recruitment in vivo. Oncotarget 2016, 7, 62439–62459. [Google Scholar] [CrossRef] [Green Version]
  111. Sato, K.; Ozaki, K.; Oh, I.; Meguro, A.; Hatanaka, K.; Nagai, T.; Muroi, K.; Ozawa, K. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 2007, 109, 228–234. [Google Scholar] [CrossRef]
  112. Tun, X.; Yasukawa, K.; Yamada, K.I. Nitric Oxide Is Involved in Activation of Toll-Like Receptor 4 Signaling through Tyrosine Nitration of Src Homology Protein Tyrosine Phosphatase 2 in Murine Dextran Sulfate-Induced Colitis. Biol. Pharm. Bull. 2018, 41, 1843–1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Hays, E.; Bonavida, B. Nitric Oxide-Mediated Enhancement and Reversal of Resistance of Anticancer Therapies. Antioxidants 2019, 8, 407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Feng, S.; Cheng, X.; Zhang, L.; Lu, X.; Chaudhary, S.; Teng, R.; Frederickson, C.; Champion, M.M.; Zhao, R.; Cheng, L.; et al. Myeloid-derived suppressor cells inhibit T cell activation through nitrating LCK in mouse cancers. Proc. Natl. Acad. Sci. USA 2018, 115, 10094–10099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Gubin, M.M.; Esaulova, E.; Ward, J.P.; Malkova, O.N.; Runci, D.; Wong, P.; Noguchi, T.; Arthur, C.D.; Meng, W.; Alspach, E.; et al. High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy. Cell 2018, 175, 1014–1030.e1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Liu, X.; Mo, W.; Ye, J.; Li, L.; Zhang, Y.; Hsueh, E.C.; Hoft, D.F.; Peng, G. Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition. Nat. Commun. 2018, 9, 249. [Google Scholar] [CrossRef]
  117. Isambert, N.; Fumoleau, P.; Paul, C.; Ferrand, C.; Zanetta, S.; Bauer, J.; Ragot, K.; Lizard, G.; Jeannin, J.F.; Bardou, M. Phase I study of OM-174, a lipid A analogue, with assessment of immunological response, in patients with refractory solid tumors. BMC Cancer 2013, 13, 172. [Google Scholar] [CrossRef]
  118. Galluzzi, L.; Vacchelli, E.; Eggermont, A.; Fridman, W.H.; Galon, J.; Sautès-Fridman, C.; Tartour, E.; Zitvogel, L.; Kroemer, G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012, 1, 699–716. [Google Scholar] [CrossRef] [Green Version]
  119. Kang, C. Senolytics and Senostatics: A Two-Pronged Approach to Target Cellular Senescence for Delaying Aging and Age-Related Diseases. Mol. Cells 2019, 42, 821–827. [Google Scholar] [CrossRef]
  120. Short, S.; Fielder, E.; Miwa, S.; von Zglinicki, T. Senolytics and senostatics as adjuvant tumour therapy. EBioMedicine 2019, 41, 683–692. [Google Scholar] [CrossRef] [Green Version]
  121. Kannaiyan, R.; Mahadevan, D. A comprehensive review of protein kinase inhibitors for cancer therapy. Expert Rev. Anticancer. Ther. 2018, 18, 1249–1270. [Google Scholar] [CrossRef]
  122. Ghione, S.; Mabrouk, N.; Paul, C.; Bettaieb, A.; Plenchette, S. Protein kinase inhibitor-based cancer therapies: Considering the potential of nitric oxide (NO) to improve cancer treatment. Biochem. Pharmacol. 2020. [Google Scholar] [CrossRef] [PubMed]
  123. Shafabakhsh, R.; Asemi, Z. Quercetin: A natural compound for ovarian cancer treatment. J. Ovarian Res. 2019, 12, 55. [Google Scholar] [CrossRef] [PubMed]
  124. Baumgart, B.; Guha, M.; Hennan, J.; Li, J.; Woicke, J.; Simic, D.; Graziano, M.; Wallis, N.; Sanderson, T.; Bunch, R.T. In vitro and in vivo evaluation of dasatinib and imatinib on physiological parameters of pulmonary arterial hypertension. Cancer Chemother. Pharmacol. 2017, 79, 711–723. [Google Scholar] [CrossRef] [PubMed]
  125. Hu, X.; Wang, H.; Han, C.; Cao, X. Src promotes anti-inflammatory (M2) macrophage generation via the IL-4/STAT6 pathway. Cytokine 2018, 111, 209–215. [Google Scholar] [CrossRef] [PubMed]
  126. Cruz, F.F.; Horta, L.F.; Maia, L.e.A.; Lopes-Pacheco, M.; da Silva, A.B.; Morales, M.M.; Gonçalves-de-Albuquerque, C.F.; Takiya, C.M.; de Castro-Faria-Neto, H.C.; Rocco, P.R. Dasatinib Reduces Lung Inflammation and Fibrosis in Acute Experimental Silicosis. PLoS ONE 2016, 11, e0147005. [Google Scholar] [CrossRef]
  127. Singh, S.P.; Konwar, B.K. Molecular docking studies of quercetin and its analogues against human inducible nitric oxide synthase. Springerplus 2012, 1, 69. [Google Scholar] [CrossRef] [Green Version]
  128. García-Mediavilla, V.; Crespo, I.; Collado, P.S.; Esteller, A.; Sánchez-Campos, S.; Tuñón, M.J.; González-Gallego, J. The anti-inflammatory flavones quercetin and kaempferol cause inhibition of inducible nitric oxide synthase, cyclooxygenase-2 and reactive C-protein, and down-regulation of the nuclear factor kappaB pathway in Chang Liver cells. Eur. J. Pharmacol. 2007, 557, 221–229. [Google Scholar] [CrossRef]
  129. Ma, J.Q.; Li, Z.; Xie, W.R.; Liu, C.M.; Liu, S.S. Quercetin protects mouse liver against CCl₄-induced inflammation by the TLR2/4 and MAPK/NF-κB pathway. Int. Immunopharmacol. 2015, 28, 531–539. [Google Scholar] [CrossRef]
  130. Martínez-Flórez, S.; Gutiérrez-Fernández, B.; Sánchez-Campos, S.; González-Gallego, J.; Tuñón, M.J. Quercetin attenuates nuclear factor-kappaB activation and nitric oxide production in interleukin-1beta-activated rat hepatocytes. J. Nutr. 2005, 135, 1359–1365. [Google Scholar] [CrossRef] [Green Version]
  131. Shen, S.C.; Lee, W.R.; Lin, H.Y.; Huang, H.C.; Ko, C.H.; Yang, L.L.; Chen, Y.C. In vitro and in vivo inhibitory activities of rutin, wogonin, and quercetin on lipopolysaccharide-induced nitric oxide and prostaglandin E(2) production. Eur. J. Pharmacol. 2002, 446, 187–194. [Google Scholar] [CrossRef]
  132. Morales, A.I.; Vicente-Sánchez, C.; Jerkic, M.; Santiago, J.M.; Sánchez-González, P.D.; Pérez-Barriocanal, F.; López-Novoa, J.M. Effect of quercetin on metallothionein, nitric oxide synthases and cyclooxygenase-2 expression on experimental chronic cadmium nephrotoxicity in rats. Toxicol. Appl. Pharmacol. 2006, 210, 128–135. [Google Scholar] [CrossRef] [PubMed]
  133. Banerjee, T.; Van der Vliet, A.; Ziboh, V.A. Downregulation of COX-2 and iNOS by amentoflavone and quercetin in A549 human lung adenocarcinoma cell line. Prostaglandins Leukot Essent Fatty Acids 2002, 66, 485–492. [Google Scholar] [CrossRef] [PubMed]
  134. Takahashi, S.; Mendelsohn, M.E. Synergistic activation of endothelial nitric-oxide synthase (eNOS) by HSP90 and Akt: Calcium-independent eNOS activation involves formation of an HSP90-Akt-CaM-bound eNOS complex. J. Biol. Chem. 2003, 278, 30821–30827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Murtagh, J.; Lu, H.; Schwartz, E.L. Taxotere-induced inhibition of human endothelial cell migration is a result of heat shock protein 90 degradation. Cancer Res. 2006, 66, 8192–8199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Hassan, M.; Watari, H.; AbuAlmaaty, A.; Ohba, Y.; Sakuragi, N. Apoptosis and molecular targeting therapy in cancer. Biomed. Res. Int. 2014, 2014, 150845. [Google Scholar] [CrossRef] [Green Version]
  137. Wright, C.; Iyer, A.K.; Kulkarni, Y.; Azad, N. S-Nitrosylation of Bcl-2 Negatively Affects Autophagy in Lung Epithelial Cells. J. Cell Biochem. 2016, 117, 521–532. [Google Scholar] [CrossRef] [Green Version]
  138. Scroggins, B.T.; Neckers, L. Just say NO: Nitric oxide regulation of Hsp90. EMBO Rep. 2009, 10, 1093–1094. [Google Scholar] [CrossRef] [Green Version]
  139. Pillai, R.N.; Ramalingam, S.S. Heat shock protein 90 inhibitors in non-small-cell lung cancer. Curr. Opin. Oncol. 2014, 26, 159–164. [Google Scholar] [CrossRef]
  140. García-Cardeña, G.; Fan, R.; Shah, V.; Sorrentino, R.; Cirino, G.; Papapetropoulos, A.; Sessa, W.C. Dynamic activation of endothelial nitric oxide synthase by Hsp90. Nature 1998, 392, 821–824. [Google Scholar] [CrossRef]
  141. Natarajan, M.; Konopinski, R.; Krishnan, M.; Roman, L.; Bera, A.; Hongying, Z.; Habib, S.L.; Mohan, S. Inhibitor-κB kinase attenuates Hsp90-dependent endothelial nitric oxide synthase function in vascular endothelial cells. Am. J. Physiol. Cell Physiol. 2015, 308, C673–C683. [Google Scholar] [CrossRef] [Green Version]
  142. Martínez-Ruiz, A.; Lamas, S. Detection and identification of S-nitrosylated proteins in endothelial cells. Methods Enzymol. 2005, 396, 131–139. [Google Scholar] [CrossRef] [PubMed]
  143. Retzlaff, M.; Stahl, M.; Eberl, H.C.; Lagleder, S.; Beck, J.; Kessler, H.; Buchner, J. Hsp90 is regulated by a switch point in the C-terminal domain. EMBO Rep. 2009, 10, 1147–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Xue, W.; Zender, L.; Miething, C.; Dickins, R.A.; Hernando, E.; Krizhanovsky, V.; Cordon-Cardo, C.; Lowe, S.W. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 2007, 445, 656–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Kang, T.W.; Yevsa, T.; Woller, N.; Hoenicke, L.; Wuestefeld, T.; Dauch, D.; Hohmeyer, A.; Gereke, M.; Rudalska, R.; Potapova, A.; et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 2011, 479, 547–551. [Google Scholar] [CrossRef]
  146. Seignez, C.; Martin, A.; Rollet, C.E.; Racoeur, C.; Scagliarini, A.; Jeannin, J.F.; Bettaieb, A.; Paul, C. Senescence of tumor cells induced by oxaliplatin increases the efficiency of a lipid A immunotherapy via the recruitment of neutrophils. Oncotarget 2014, 5, 11442–11451. [Google Scholar] [CrossRef] [Green Version]
  147. Onier, N.; Hilpert, S.; Reveneau, S.; Arnould, L.; Saint-Giorgio, V.; Exbrayat, J.M.; Jeannin, J.F. Expression of inducible nitric oxide synthase in tumors in relation with their regression induced by lipid A in rats. Int. J. Cancer 1999, 81, 755–760. [Google Scholar] [CrossRef]
  148. Gautier, T.; Paul, C.; Deckert, V.; Desrumaux, C.; Klein, A.; Labbé, J.; Le Guern, N.; Athias, A.; Monier, S.; Hammann, A.; et al. Innate immune response triggered by triacyl lipid A is dependent on phospholipid transfer protein (PLTP) gene expression. FASEB J. 2010, 24, 3544–3554. [Google Scholar] [CrossRef]
  149. Lamrani, M.; Sassi, N.; Paul, C.; Yousfi, N.; Boucher, J.L.; Gauthier, N.; Labbé, J.; Seignez, C.; Racoeur, C.; Athias, A.; et al. TLR4/IFNγ pathways induce tumor regression via NOS II-dependent NO and ROS production in murine breast cancer models. Oncoimmunology 2016, 5, e1123369. [Google Scholar] [CrossRef] [Green Version]
  150. Martin, A.; Seignez, C.; Racoeur, C.; Isambert, N.; Mabrouk, N.; Scagliarini, A.; Reveneau, S.; Arnould, L.; Bettaieb, A.; Jeannin, J.F.; et al. Tumor-derived granzyme B-expressing neutrophils acquire antitumor potential after lipid A treatment. Oncotarget 2018, 9, 28364–28378. [Google Scholar] [CrossRef] [Green Version]
  151. Eggert, T.; Wolter, K.; Ji, J.; Ma, C.; Yevsa, T.; Klotz, S.; Medina-Echeverz, J.; Longerich, T.; Forgues, M.; Reisinger, F.; et al. Distinct Functions of Senescence-Associated Immune Responses in Liver Tumor Surveillance and Tumor Progression. Cancer Cell 2016, 30, 533–547. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Impact of nitric oxide (NO) in senescence-mediated signaling pathways. NO plays important roles in different senescence-associated secretory phenotype (SASP) signaling pathways. Exogenous and endogenous NO can cause DNA damage that can lead to protein kinase ataxia-telangiectasia-mutated protein (ATM) and Chk2 activation, or NF-κB activation and interleukin 6 (IL-6)/IL-8 gene transcription. All these changes can induce the initiation of cellular senescence programs. NO is also able to induce direct modifications in NF-κB, either by S-nitrosylating both p60/p65 NF-κB subunits and leading to SASP gene transcription inhibition, or by S-nitrosylating the NF-κB kinase inhibitor subunit α (IKKα), which results in NF-κB inhibition. S-nitrosylation can also affect the c-Jun N-terminal kinase (JNK) signaling pathway by leading to the inhibition of its activation. Moreover, cyclooxygenase 2 (COX2), a prostaglandin E2 (PGE2) secretion inducer, is also an NO target. The S-nitrosylation of COX2 stimulates its activity and induces anti-tumor immunity suppression. Another group of SASP factors altered by NO are chemokines such as CCL2, CCL5, CXCL1 and CXCL12, which can transcriptionally and translationally be decreased in response to NO donors, thus reducing immune cell recruitment.
Figure 1. Impact of nitric oxide (NO) in senescence-mediated signaling pathways. NO plays important roles in different senescence-associated secretory phenotype (SASP) signaling pathways. Exogenous and endogenous NO can cause DNA damage that can lead to protein kinase ataxia-telangiectasia-mutated protein (ATM) and Chk2 activation, or NF-κB activation and interleukin 6 (IL-6)/IL-8 gene transcription. All these changes can induce the initiation of cellular senescence programs. NO is also able to induce direct modifications in NF-κB, either by S-nitrosylating both p60/p65 NF-κB subunits and leading to SASP gene transcription inhibition, or by S-nitrosylating the NF-κB kinase inhibitor subunit α (IKKα), which results in NF-κB inhibition. S-nitrosylation can also affect the c-Jun N-terminal kinase (JNK) signaling pathway by leading to the inhibition of its activation. Moreover, cyclooxygenase 2 (COX2), a prostaglandin E2 (PGE2) secretion inducer, is also an NO target. The S-nitrosylation of COX2 stimulates its activity and induces anti-tumor immunity suppression. Another group of SASP factors altered by NO are chemokines such as CCL2, CCL5, CXCL1 and CXCL12, which can transcriptionally and translationally be decreased in response to NO donors, thus reducing immune cell recruitment.
Cancers 12 01145 g001
Table 1. The senescence-associated secretory phenotype (SASP) factors (based on [24,34]).
Table 1. The senescence-associated secretory phenotype (SASP) factors (based on [24,34]).
Interleukins and Other Inflammatory MoleculesIL-1; IL-1β; IL-6; IL-7; IL-8; IL-13; IL-15; TGFβ; GM-CSF; G-CSF; CSF-1; IFN-γ; BLC; MIF
Chemokines and Growth Factors/RegulatorsCXCL1; CXCL2; CXCL5; CXCL12; CCL2; CCL5; CCL8; CCL13; MIP-1α; MIP-3α; HCC-4; eotaxin/eotaxin-3; TECK; ENA-78; Amphiregulin; epiregulin; heregulin; EGF; bFGF; HGF; KGF (FGF7); VEGF; angiogenin; SCF
Receptors and LigandsICAM-1/3; OPG; TNFα; sTNFRI; sTNFRII; TRAIL-R3; Fas; uPAR; SGP130; EGF-R
Proteases and Extracellular Matrix ProteinsMMP-1/3/10/12/13/14; TIMP-1/2; PAI-1/2; tPA; uPA; cathepsin B
Non-Protein Molecules and Insoluble FactorsNitric oxide; ROS; PGE2; fibronectin; collagens; laminin
Table 2. Senolytic-induced nitric oxide (NO) modulations.
Table 2. Senolytic-induced nitric oxide (NO) modulations.
Senolytic DrugsNO ChangesModelReferences
DasatinibIncreased NOPulmonary artery endothelial cells and smooth muscle cells[124]
Increase iNOS expresionIntestinal and bone morrow-derived macrophages[125]
Decrease iNOS expresionSilicotic macrophages[126]
QuercetinInhibition of iNOS [127]
Inhibition of mRNA iNOSHuman hepatocyte-derived cell line[128]
Inhibition of NO production and iNOS expressionLivers of CCl4-treated mice[129]
In vitro (rat hepatocyte)[130]
In vitro and in vivo (RAW 264.7 macrophages, primary peritoneal macrophages and Balb/c mice)[131]
Chronic cadmium nephrotoxicity in rats[132]
Lung adenocarcinoma cell lines[133]
Hsp90 inhibitorReduction of NO productionEndothelial cells[134]
Blocked VEGF-induced increase in eNOS activityEndothelial cells[135]

Share and Cite

MDPI and ACS Style

Mabrouk, N.; Ghione, S.; Laurens, V.; Plenchette, S.; Bettaieb, A.; Paul, C. Senescence and Cancer: Role of Nitric Oxide (NO) in SASP. Cancers 2020, 12, 1145. https://doi.org/10.3390/cancers12051145

AMA Style

Mabrouk N, Ghione S, Laurens V, Plenchette S, Bettaieb A, Paul C. Senescence and Cancer: Role of Nitric Oxide (NO) in SASP. Cancers. 2020; 12(5):1145. https://doi.org/10.3390/cancers12051145

Chicago/Turabian Style

Mabrouk, Nesrine, Silvia Ghione, Véronique Laurens, Stéphanie Plenchette, Ali Bettaieb, and Catherine Paul. 2020. "Senescence and Cancer: Role of Nitric Oxide (NO) in SASP" Cancers 12, no. 5: 1145. https://doi.org/10.3390/cancers12051145

APA Style

Mabrouk, N., Ghione, S., Laurens, V., Plenchette, S., Bettaieb, A., & Paul, C. (2020). Senescence and Cancer: Role of Nitric Oxide (NO) in SASP. Cancers, 12(5), 1145. https://doi.org/10.3390/cancers12051145

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