Mechanisms of inflammatory responses to radiation ...

1 downloads 0 Views 798KB Size Report
Mar 5, 2018 - associated with acute dermatitis are erythema, dry desquamation, and moist desquamation that result in epidermal necrosis, fibrinous exudates ...
International Journal of Radiation Biology

ISSN: 0955-3002 (Print) 1362-3095 (Online) Journal homepage: http://www.tandfonline.com/loi/irab20

Mechanisms of inflammatory responses to radiation and normal tissues toxicity; clinical implications Masoud Najafi, Elahe Motevaseli, Alireza Shirazi, Ghazale Graily, Abolhasan Rezaeyan, Farzad Norouzi, Saeed Rezapoor & Hamid Abdollahi To cite this article: Masoud Najafi, Elahe Motevaseli, Alireza Shirazi, Ghazale Graily, Abolhasan Rezaeyan, Farzad Norouzi, Saeed Rezapoor & Hamid Abdollahi (2018): Mechanisms of inflammatory responses to radiation and normal tissues toxicity; clinical implications, International Journal of Radiation Biology To link to this article: https://doi.org/10.1080/09553002.2018.1440092

Accepted author version posted online: 05 Mar 2018.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=irab20

Mechanisms of inflammatory responses to radiation and normal tissues toxicity; clinical implications Masoud Najafi1, Elahe Motevaseli2*, Alireza Shirazi3*, Ghazale Graily3, Abolhasan Rezaeyan4,

ip

t

Farzad Norouzi5, Saeed Rezapoor5, Hamid Abdollahi4

cr

1 Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah

us

University of Medical Science, Kermanshah, Iran

University of Medical Sciences, Tehran, Iran

an

2 Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran

M

3 Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran

d

University of Medical Sciences, Tehran, Iran

pt e

4 Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran

ce

5 Science and Research Branch, Azad University, Tehran, Iran

Ac

6 Department of Radiology, Faculty of Paramedical Sciences, Tehran University of Medical Sciences, Tehran, Iran Corresponding author: E. Motevaseli, [email protected], Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran

Co-Corresponding author: A. Shirazi, [email protected], Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran

t

Abstract

ip

Purpose: Cancer treatment is one of the most challenging diseases in the present era. Among a

cr

few modalities for cancer therapy, radiotherapy plays a pivotal role in more than half of all

us

treatments alone or combined with other cancer treatment modalities. Management of normal

an

tissue toxicity induced by radiation is one of the most important limiting factors for an appropriate radiation treatment course. The evaluation of mechanisms of normal tissue toxicity

M

has shown that immune responses especially inflammatory responses play a key role in both early and late side effects of exposure to ionizing radiation (IR). DNA damage and cell death, as

d

well as damage to some organelles such as mitochondria initiate several signaling pathways that

pt e

result in the response of immune cells. Massive cell damage which is a common phenomenon following exposure to a high dose of IR cause secretion of a lot of inflammatory mediators

ce

including cytokines and chemokines. These mediators initiate different changes in normal tissues

Ac

that may continue for a long time after irradiation. In this study, we reviewed the mechanisms of inflammatory responses to IR that are involved in normal tissue toxicity and considered as the most important limiting factors in radiotherapy. Also, we introduced some agents that have been proposed for management of these responses. Conclusion: The early inflammation during the radiation treatment is often a limiting factor in radiotherapy. In addition to the limiting factors, chronic inflammatory responses may increase

the risk of second primary cancers through continuous free radical production, attenuation of tumor suppressor genes and activation of oncogenes. Moreover, these effects may influence nonirradiated tissues through a mechanism named bystander effect. Keywords: Radiation; Inflammation, Normal Tissue, Clinical Implications, Immune System

t

Introduction

ip

The knowledge of the responses of the immune system to cell death, DNA damage and DNA

cr

damage responses following ionizing radiation (IR) exposure are essential to understanding the

us

underlying normal tissue complications. IR induces single strand and double strand DNA breaks

an

(DSBs). Most single strand breaks can be repaired, while DSBs result in cell death. Exposure to high doses of IR produces massive DNA damage in cells. Accumulation of unrepaired DNA

M

damage can result in the induction of deletion, mutations, chromosome aberrations or cell death. For a typical radiation dose of 2 Gy of gamma or x-ray, about 3000 DNA breaks are produced

pt e

d

per cell exposed. There are two main types of IR induced DNA lesions including direct damages due to direct interaction with radiation and indirect damages due to interaction with IR induced

ce

reactive oxygen species (ROS) produced by chemical changes (Lomax et al. 2013). In addition to ROS production by IR, there are several inflammatory signaling pathways

Ac

activated after radiation exposure which amplify ROS and nitric oxide (NO) production. Inflammatory cytokines and growth factors produced by irradiated cells provoke several signaling cascades that stimulate ROS and NO producing enzymes such as NADPH Oxidase, inducible nitric oxide synthetize (iNOS), and cyclooxygenase-2 (COX-2). These enzymes produce ROS and NO which amplify IR induced DNA damage and cell death (Bours et al. 2000). Moreover, increased superoxide anions production by the electron transfer chains of

mitochondria can amplify intracellular oxidative stress (Zorov et al. 2006). Potent evidences have proposed that during chronic inflammation, free radicals serve as intracellular signals that potentiate the inflammatory response (Filippin et al. 2008). A ROS induced ROS phenomenon after exposure to radiation may continue for a long time leading to appearance of pathological changes in normal tissues (Zorov et al. 2014). Continued ROS formation after exposure to IR

ip

t

may be the origin of radiosensitivity of some cells such as T lymphocytes (Ogawa et al. 2003).

cr

DNA damage and cell death are the main mechanisms involved in triggering of inflammatory responses following exposure to IR. In response to IR, there are tissue-specific responses to

us

DNA damage (Park et al. 2014) and inflammation (Chai Y., Calaf G. M., et al. 2013).

an

Accordingly, we have described the exact mechanisms in each organ and then proposed possible

M

mitigation strategies.

Although the immunologic basis of IR induced normal tissue toxicity is widely studied and

d

taught, there is no comprehensive review in this field. In this present work, we reviewed

pt e

literatures on the underlying mechanisms in which inflammatory responses are involved in

implications.

ce

radiation induced normal tissue toxicity in different organs and summarize some of the clinical

Ac

Radiation cause induced DNA damage and immunologic or anti-immunologic cell death Exposure of cells to radiation leads to oxidative DNA damage, mutation, and several types of cell death. (Rock et al. 2011, Muralidharan and Mandrekar 2013). Mitotic catastrophe, apoptosis, necrosis, necroptosis (secondary necrosis), autophagy, and senescence are various types of cell death induced by IR (Golden and Apetoh 2015b). From immunologic point of view, IR induced cell death can be categorized to immunologic and anti-immunologic cell death. Necrosis and

necroptosis are immunogenic, while apoptosis is anti-immunogenic cell death (Park et al. 2014). Inflammatory or anti-inflammatory responses to IR result in release of different soluble mediators as well as changes in immune cell surface receptors which have been reported to play critical roles in IR induced normal tissues toxicities. For example, some studies have indicated that high levels of some immune factors such as TGF-β have been detected in serum level of the

ip

t

Chernobyl accident survivors of cancer patients undergoing RT (Marozik et al. 2007, Emerit et

cr

al. 1995, Ballardin et al. 2002).

As an interesting issue, secreted products from apoptotic cells determine whether the apoptosis is

us

immunogenic or not. In this light, when cells are exposed to radiation, several pathways can be

an

activated which would lead to cellular stress, production of different species of free radical, DNA damage and finally immunogenic apoptosis (necroptosis) (Opferman 2008). Whilst in another

M

way, normal apoptosis state is mostly non-immunogenic. In regard to necrosis, it is an inherently

d

immunogenic and pathological cell death associated with several inflammatory responses (Green

pt e

et al. 2009). The balance between apoptosis and necrosis play a key role in the immune responses to early and late effects consequences (Golden and Apetoh 2015a).

ce

In regard to cell types, it is widely observed that apoptosis is the most common form of cell

Ac

death in some radiosensitive organs such as bone marrow and peripheral lymphocytes. Studies on acute radiation exposure have shown that depletion of bone marrow stem cells, granulocytes and natural killer (NK) cells are more obvious followed by pelvic and chest bone irradiation, where the bone marrow cells are producing new marrow cells (Iyer and Jhingran 2006) (MacVittie et al. 2015). Also, some studies have identified that the occurrence of apoptosis compared to necrosis is very obvious after exposure radiation doses lower than 1 Gy (Kaur and Asea 2012).

p53 has a crucial role in radiation-induced apoptosis. Apoptosis or programmed cell death dependent on p53 occurs within a few hours after irradiation (Lee et al. 2013). The extrinsic apoptosis pathways stimulated by binding of the cell death surface receptor FAS by FAS ligand. The interaction of FAS by its ligand results in apoptotic cell death, mediated by caspase cascade activation (Watters 1999). The evidence has been shown that intrinsic or extrinsic apoptosis can

ip

t

be independently regulated within the same cell type and the decision of life or death was controlled by the cell (Hotchkiss et al. 2000, Nowsheen and Yang 2012). The mitochondrial

us

cells such as lymphocytes (Enoch and Norbury 1995).

cr

apoptosis pathway is considered as a reason for high intrinsic radiosensitivity of some immune

an

Necrosis has been investigated using different radiation doses, but it is very important after exposure to higher doses of radiation. The most common reason for necrosis in normal cells after

M

irradiation is damages to vessels and hypoxia situation. Vascular damages due to high doses of

d

radiation is a mechanism for nutrition deprivation and hypoxia that amplify radiation induced

pt e

necrosis (Greene-Schloesser et al. 2012). Evidences have shown that combined chemotherapy and radiation therapy increases the incidence of necrosis to manifold that is seen with

ce

radiotherapy alone (Miyaguchi et al. 1997, Keime-Guibert et al. 1998).

Ac

Why IR induced DNA damage and cell death trigger inflammation? Immunogenic VS. tolerogenic cell death Cells dying by normal apoptosis are phagocytosed by macrophages and don’t trigger inflammatory responses. Also, some mechanisms involved in the process of apoptosis cause tolerogenic and anti-inflammatory responses. Interactions of macrophages with apoptotic cells can stimulate macrophages to generate anti-inflammatory cytokines such as IL-10, TGF-β,

platelet-activating factor and prostaglandin E2 (PGE2) which result in suppression of the inflammatory responses. An increased level of TGF-β inhibits the production of proinflammatory cytokines such as IL-1 and TNF-α by activated macrophages. Thus, these changes lead to a loss of immune responses (Chung et al. 2006). In contrast, in some stress conditions like exposure to IR, a higher number of cells may undergo

ip

t

apoptosis, which can overwhelm the phagocytic system. This may result in release of danger

cr

signals from damaged membranes, and organelles such as mitochondria (Rock and Kono 2008; Silva 2010). This may result in release of danger signals from destroying membrane, and

us

organelles such as mitochondria. Danger signals as so called ‘damage-associated molecular

an

patterns (DAMPs)’ are endogenous molecular structures within the cells which are hidden from the immune system. These molecular structures such as uric acid, adenosine triphosphate (ATP),

M

heat shock proteins (HSPs), and high-mobility group box 1 (HMGB1) are liberated after tissue

d

damage like after exposure to radiation (Klune et al. 2008; Krysko et al. 2012; Chacon et al.

pt e

2016). Danger signals released during necrosis, as well as secondary necrosis that occurs following apoptosis trigger pro-inflammatory cytokines production and immune-stimulatory

Ac

Oxidized DNA

ce

response (Poon et al. 2014).

In addition to dying cells, evidences have shown that the DNA damage after irradiation can stimulate the inflammatory responses. The extensive DNA damage results in the release of triggering signals. DAMPs such as HMGB1 and oxidized DNA are considered as a link between DNA damage and immune responses (Gehrke et al. 2013). DNA damage after exposure to radiation can lead to the formation of cell free oxidized DNA. (Veiko 2013).

In addition to nuclear DNA, mitochondrial DNA (mtDNA) has a role in immune system activation. Mitochondrial apoptosis can result in the release of oxidized mtDNA into the cytosol and bound it to NLRP3 inflammasome in the cytosol, which causes activation of the NLRP3 inflammasome (Allam et al. 2014). Also, a part of increased levels of some inflammatory cytokines after exposure to radiation may activate apoptosome and inflammasome during

t

apoptosis (Hogquist et al. 1991; Shimada et al. 2012; Chai J and Shi 2014; Ha et al. 2014). The

ip

inflammasome promotes the maturation and secretion of the inflammatory cytokines IL-1β and

cr

IL-18 (Pétrilli et al. 2007).

us

The oxidized DNA originated from both DNA and mtDNA has been implicated in the secretion

an

of pro-inflammatory cytokines through NLRP3 inflammasome activation (Haneklaus et al. 2013; Abderrazak et al. 2015). On the other hand, generation of ROS via IR itself, stimulates

M

inflammasome activation (Martinon 2010; Sorbara and Girardin 2011; Zhou R et al. 2011). Stoecklein et al. showed that IR induces a dose-dependent augmentation in inflammasome

pt e

d

activation in immune cells, such as macrophages, T and B lymphocytes, dendritic cells and NK cells. They have hypothesized that the inflammasome contributes to both acute and chronic

ce

immune responses due to radiation exposure (Stoecklein et al. 2015).

Ac

Toll-like receptors (TLRs)

The release of intracellular contents like DAMPs can engage in the immune system through some ligands causing inflammation. The pattern recognition receptors (PRRs) that are expressed by Dendritic cells (DCs) recognize some of these DAMPs that stimulate antigen uptake. PRRs including Toll-like receptors (TLRs) identify danger signals, cytokines and chemokines released by the inflammatory cells. DAMPs recognized by TLRs activate the inflammation cascades and transcription of NF-κB and activator proteins (AP-1). Danger signals derived from necrotic cells

compared to apoptosis, have more potent effects on DCs maturation. In addition, secondary necrosis after apoptosis post-irradiation were more effective for DC maturation compared to physiological apoptosis (Piccinini and Midwood 2010; Karki and Igwe 2013). Increased HMGB1, HSPs and oxidized DNA following irradiation usually through TLR2, TLR4, TLR5 and TLR9 can increase expression of NF-κB and pro-inflammatory cytokines (Lyudmila

ip

t

G. Burdelya 2008; Piccinini and Midwood 2010). TLRs are the key link between tissue injury

cr

and cell death, and immune system responses to IR.

us

NF-κB plays a central role in response to danger signals released from necrotic or necroptotic cells. The recognition of DAMPs with TLRs induces NF-κB signaling through the mitogen

an

activated protein kinases (MAPKs), DNA-dependent protein kinases (DNA-PKs) and

M

phosphoinositide 3-kinase (PI3K) pathways (Hellweg 2015). NF-κB controls the expression of many growth factors, apoptosis inhibitors and molecules needed for inflammation, including pro-

d

inflammatory cytokines and chemokines, iNOS, COX-2 and vascular adhesion molecules needed

pt e

for leukocyte recruitment. The cytokines and chemokines generated in irradiated tissues depends on tissue type, because the resident immune cells type and numbers are varying between

ce

different tissues. Therefore, immunomodulatory changes that lead to normal tissue damages and

Ac

carcinogenesis can vary among different tissue types (Pos et al. 2005; Aggarwal et al. 2006).

Fig 1: Mechanisms of inflammation activation following exposure to IR Role of inflammatory responses to IR in carcinogenesis Epidemiological studies have proposed that 25% of all cases of cancer is related to chronic inflammation (Okada 2014). Inflammatory responses to IR can result in DNA damage and

inhibition of DNA repair pathways, damages to non-irradiated tissues through non-targeted effect, increased carcinogenesis risk and some non-cancerous diseases like heart diseases, diabetes, dermatitis and digestive disorders (Kusunoki and Hayashi 2008). In this section, we describe some possible carcinogenic effect of IR through stimulation of inflammation.

t

Activation of Inflammation mediators

ip

Inflammatory responses induced by IR are mediated by several mediators such as NF-κB, iNOS,

cr

COX-2, EGFR, MCP-1, and cytokines such as IL-1, IL-2, IL-6, TNF-α and IFN-γ. An elevated

us

level of inflammatory cytokines in normal tissues is reported in cancer patients undergoing radiotherapy and chemotherapy (Wang XS et al. 2010; Siva et al. 2014). Macrophages and

an

monocytes are the most important immune cells in the initiation and maintenance of

M

inflammation by secreting pro-inflammatory cytokines. Acute inflammation usually occurs due to massive cell death in rapidly dividing cells. This response is the initial response of the body to

d

a high dose radiation and was initiated by release of chemokines and then the increased

pt e

movement of leukocytes from the blood into the irradiated tissues. In this situation, the resident macrophages, neutrophils, lymphocytes and DCs are increased in irradiated tissues during a few

ce

hours after exposure. Depending on location, acute inflammation can cause temporary erythema,

Ac

ulceration, edema and in the lung, pneumonitis (Sprung et al. 2015). High dose radiobiology studies have shown that these doses lead to a continuous inflammation in exposed people. This is associated with a long term increase of macrophage, neutrophil and lymphocyte population in the irradiated tissues. As illustrated in Fig1, the increased activity of these cells leads to continuous production of pro-inflammatory cytokines and chemokines that are associated with prolonged ROS and NO generation. Thus, oxidative stress and cellular damages induced by radiation may continue for a long time. The long term effects of

inflammation can lead to various detrimental effects such as pathological damages and carcinogenesis due to progressive and persistent oxidative stress (Hayashi et al. 2005; Zhao Weiling and Robbins 2009). The effects on DNA repair response

t

DNA repair system plays a key role in tolerance of normal tissues to radiotherapy. On the other

ip

hand, change in DNA repair response, tumor suppressor genes and oncogenes are involved in

cr

both early and late consequences of radiotherapy. However, there is a predictable increase in

us

regulation of DNA repair genes after radiation exposure, evidences have been proposed that chronic inflammation cause suppression of DNA repair response, and mutation in tumor

an

suppressor genes and oncogenes. Thus, chronic inflammation may lead to accumulation of

M

unrepaired DNA. During chronic inflammation, cells may be exposed to high quantities of NO and ROS (Dedon and Tannenbaum 2004). Oxidative and nitrative DNA damage cause mutations

d

of DNA repair genes which are involved in the initiation and promotion of carcinogenesis (Meira

pt e

Lisiane B et al. 2008; Ohnishi et al. 2013; Lin Runhua et al. 2015). For example, decreased XRCC1 expression is associated with acute side effects in breast cancer patients that underwent

ce

radiotherapy (Batar et al. 2016).

Ac

NO plays a key role in suppression of DNA damage responses during the inflammation. NO produced by immune cells including macrophages and neutrophils can inhibit DNA repair and alter the expression of certain genes, which would provide grounds for genomic instability (Jaiswal et al. 2000). There are three forms of nitric oxide sources, including iNOS (inducible NOS), nNOS (neuronal NOS) and eNOS (endothelial NOS). NO at abnormal level is mainly involved in homeostatic processes such as neurotransmission, host defense, and blood pressure regulation (Nathan 1992). The iNOS initially can be found in macrophages, eNOS is membrane-

bound and nNOS can be found in the cytosol. Among the family of nitric oxide synthesis enzymes, iNOS is the main source of NO production during inflammation (Zhao W et al. 2014). It is now understood that in addition to macrophages, several types of cells can express iNOS in the presence of inflammatory cytokines (Bogdan 2015). Studies have confirmed the upregulation of iNOS in irradiated cells too (MacNaughton 1998). NO is produced by iNOS enzyme from L-

ip

t

arginine in inflamed tissues (Rath et al. 2014). NO can react by DNA and leads base oxidation,

cr

deamination, and nitration (Tamir et al. 1996).

Previous studies have reported that mutations in BER genes and defects in MMR repair pathway

us

is associated with chronic inflammation, which can result in incomplete repair of DNA damage,

an

leading to mutation, chromosomal instability and predisposition to several types of epithelial cancer (Kidane et al. 2014). Chien and et al. have suggested that NO, peroxynitrite, and arsenite

M

can inhibit DNA adduct excision in NER pathway (Chien et al. 2004). NO directly affects

d

oxidative DNA damage repair including 8-oxoguanine repair processes. 8-Oxoguanine

pt e

glycosylase (Ogg1) is responsible for the excision of 8-oxoguanine. An inhibition by NO has been investigated for homologue of Ogg1 protein. This is related to nitrosylation of cysteine

ce

residues in the zinc-finger motif of the Ogg1 protein after expose to a NO donor or iNOSinducing cytokines (Wink and Laval 1994; Jaiswal, LaRusso, Nishioka, et al. 2001). Ogg1

Ac

inhibition by NO result in increased accumulation of oxidative DNA lesions (Jaiswal, LaRusso, Shapiro, et al. 2001). The failure to repair 8-oxodG increases mutagenesis and expected to promote cancer initiation and progression. Studies have shown that hOGG1 gene mutations are associated with several human cancers such as lung, kidney and gastric cancer (Chevillard et al. 1998; Shinmura et al. 1998; Rezapoor et al. 2017).

Continuous inhibition of DNA repair by NO may be proposed as a mechanism linking between chronic inflammation and carcinogenesis after exposure to radiation (Kidane et al. 2014). The production of NO by iNOS is a feature of the inflamed gastrointestinal tract and it has been known as risk factors for the esophagus, stomach, liver, pancreas, biliary tract, and bowel carcinogenesis (Kubes and McCafferty 2000; Jaiswal, LaRusso, Gores 2001).

ip

t

Effects on tumor suppressor genes

cr

A relationship between exposure to IR and deletion of tumor suppression genes have been

us

demonstrated (Willey et al. 1993). Molecular genetic analysis have revealed that deletion or mutation of tumor suppressor genes in some chromosomes such as ch8, ch11, ch14 after

an

exposure to IR (Mendonca et al. 1995; Weaver et al. 1997; Mendonca et al. 1998; Mendonca et

M

al. 2004). A variety of studies have proposed that activation of inflammation responses and also anti-inflammatory genes such as TGFB1 reduces the activity of some tumor suppressor genes

d

(Hei T. K. et al. 2011). Although, TGF-β have a suppressive effect on tumor induction, several

pt e

evidences have indicated the clastogenic role of the following exposure to radiation (Krstić et al. 2015). This may continue for a long time after exposure (Randall and Coggle 1996). Abnormal

ce

upregulated TGF-β constantly induces the generation of ROS and NO from reduction/oxidation

Ac

(redox) system enzymes such as NOX4, COX-2, iNOS and also mitochondrial electron transport chain (ETC) (Pazhanisamy et al. 2011; Najafi M. et al. 2014; Masoud Najafi 2018; Yahyapour et al. 2018). Moreover, TGF-β suppresses the activity of antioxidant system enzymes such as superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPx) (Liu R-M and Pravia 2010). Chronic oxidative damage induced by redox system may inactivate tumor suppressor genes and activate oncogenes leading to carcinogenesis (Hussain et al. 2003; Meira Lisiane B. et al. 2008; Lin R. et al. 2015).

There are several reports indicating the upregulation of inflammatory genes and cytokines suppressing p53 activity (Yonish-Rouach et al. 1991; Hudson et al. 1999). NF-κB plays a central role in the regulation of this pathway in cells (Gurova et al. 2005). It was demonstrated that NFκB and p53 negatively regulate the activities of each other. Also, long term up-regulation of NFκB during chronic inflammation inhibiting p53 activity is involved in pathologies associated with

t

genotoxicity such as acute radiation syndrome and loss of normal function of organs (Gudkov

ip

and Komarova 2016). Suppression of NF-κB have been proposed as a strategy for activation of

us

Effects of inflammatory responses on oncogenes

cr

p53 in cancer therapy and amelioration of inflammation (Gudkov et al. 2011).

an

Inflammatory responses to IR have a potent relation to activation of pro-oncogenic pathways.

M

Although, complete mechanisms of radiation induced oncogenes activation are unknown, some studies have proposed a role for inflammatory responses. Based on best knowledge, transcription

d

factors and tyrosine kinases have key roles on oncogenesis following exposure to radiation.

pt e

Signal transducers and activators of transcription members (STATs) are a family of transcription factors that have a potent effect on tumorigenesis. STATs including STAT1, STAT3, and

ce

STAT5 have a key role in cell death and controlling cell-cycle progression and growth arrest

Ac

through cyclin D1 and c-Myc, thus are included among the oncogenes (Calo et al. 2003). STATs which are activated following irradiation in a dose-dependent manner promote cell proliferation (Gao et al. 2014). Blockade of STAT3 using JAK2 inhibitors inhibits tumor proliferation following irradiation (Lau et al. 2015). Thus, blockade of STAT signaling pathways in combination with cancer therapy modalities including radiotherapy may be useful for overcoming tumor radio-resistance (Yu et al. 2009; Sun Y. et al. 2013).

Tyrosine kinases are the other oncogenic signals that serves as a link between inflammatory responses to radiation and cell proliferation. The oncogenes such as EGFR, PDGFR, AKT and RAF have found mutations in different types of human cancers. Abnormal activated tyrosine kinases have also been found among Chernobyl survivors that have been exposed to nuclear disaster (Ricarte-Filho et al. 2013). These kinases are able to stimulate aberrant cell proliferation

ip

t

and malignancy through MAPKs and PI3K pathway (Ciampi et al. 2005). Also, these kinases inhibit apoptosis by activating transcription factors AP1 and NF-κB through suppression of pro-

cr

apoptotic genes including Bax and Bad (Tsatsanis 2000). Hence, aberrant regulation of tyrosine

an

Inflammation in radiation-induced bystander effect

us

kinases may be consistently associated with genetic perturbations and carcinogenesis.

M

The radiation-induced bystander/non-targeted effect is a phenomenon that leads to damage in tissues that have not been directly exposed to IR. Although mechanisms of this phenomenon are

d

not exactly known, studies indicate that the immune system and inflammatory response signals

pt e

play a central role in distant tissue damages after localized irradiation. As noted earlier, exposure to IR causes damage to living cells, and can result in mutation and immunological cell death.

ce

Inflammatory cytokines such as IL-1, IL-6, IL-8, IL-33, TNF-α and TGF-β released by

Ac

macrophages and lymphocytes influence gene expression and epigenetic programming at distant tissues (Liu SZ et al. 2004; Calveley et al. 2005; Koturbash et al. 2008; Fardid et al. 2017). The most important pathways in bystander effect are associated with free radical production induced by inflammatory signaling. Increased COX-2, NADPH Oxidase and iNOS are confirmed in in vitro and in vivo studies (Hei Tom K et al. 2008; Fardid R 2017; Yahyapour, Motevaseli, et al. 2017). An in vitro study by Chai et al. showed that abdominal irradiation enhances COX-2 gene expression in lung by 20-fold and in bronchi by 30-fold. They showed

that COX-2 expression has a key role in pathologic processes characterized by increased local prostaglandins and ROS production. The evidences indicated that the increased COX-2 expression induced by out-of-field effect is regulated in a tissue specific manner (Chai Y., Calaf G. M., et al. 2013; Wang TJC et al. 2015). TGFβ-TGFβR1–COX-2 pathway has key roles in ROS production in distant lung tissues. The expression level of TGβR1 in the lung was

ip

t

significant but not for the liver (Chai Y., Lam R. K., et al. 2013). Moreover, TGFβ can upregulate ROS production in non-irradiated cells through miR-21 and suppression of

cr

superoxide dismutase regulation (Jiang et al. 2014; Xu et al. 2014; Tian et al. 2015). These

us

consequences result in oxidative damage, chromosome aberrations, gene mutations, genomic

an

instability and aneuploidy, as well as epigenetic changes such as DNA hypomethylation, histone modification, alteration in methyltransferases enzymes and RNA-associated silencing

M

(Kovalchuk and Baulch 2008; Mothersill and Seymour 2012; Ghobadi et al. 2017).

d

As mentioned above, immunological consequences induced by radiation at non-targeted tissues

pt e

can result in cellular and molecular changes involved in the carcinogenesis. Fractionated radiotherapy leads to cell death, the release of DAMPs such as oxidized DNA by damaged cells

ce

and upregulation of inflammatory cytokines for several weeks (Ermolaeva and Schumacher 2013; Pateras et al. 2015). At the same time with localized cancer radiotherapy, the non-targeted

Ac

effect induced by radiation cause cellular and molecular damages in distant tissues (Nikitaki et al. 2016). So, this is an important issue for patients that undergo localized radiotherapy, because it may be a threat for second malignancies and decreased survival rate in cancer patients (Najafi M 2017). It is thought that the high incidence of secondary lung cancer in patients that underwent pelvic radiation therapy has a relationship to this phenomenon (Najafi M. et al. 2016).

Inflammatory responses to radiation and pathological damages Radiation pathology is one of the most important radiobiological phenomena among cancer patients undergoing radiation treatment. Histopathologic changes in human cells after exposure to radiation are commonly evident in routine radiation treatments. Pathological changes are the late effects of IR that are seen after exposure to high doses received with organs. The lesions can

ip

t

be divided into the parenchyma or epithelia changes (e.g. atrophy, necrosis, metaplasia), the

cr

stromal elements changes (e.g. fibrosis, fibrinous exudates, necrosis), and the vascular damages (e.g. damage to the endothelial cells, thrombosis and atherosclerosis) (Fajardo 2005). These

us

damages are caused by massive DNA damage and unrepaired damages, cell death and

an

continuous ROS/NO production for a long time after exposure. Continuous elevated levels of inflammatory cytokines, chemokines, growth factors and adhesion molecules lead to several

M

irreversible changes. The most crucial tissues affected by these immune responses and

d

subsequent pathological changes includes the lung, heart, brain, liver, intestine, kidneys, spleen

pt e

and colon. In this section, we present the most important long term pathological changes which include fibrosis, vascular damages and subsequent consequences followed by radiation-activated

Ac

Fibrosis

ce

immune responses.

Radiation can lead to the formation of excess fibrous connection that cause tissue thickness and stiffness. Fibrosis is a late effect of high dose radiation exposure that can affect functions of some irradiated tissues and organs such as the lung, heart, liver, skin, gastrointestinal system, and breast. Radiation-induced fibrosis is highly dependent on multiple factors, which include the radiation dose, irradiated volume and fractionation schedule (Borger et al. 1994).

Fibrosis process is similar to wound healing. Normal wound healing is regulated by the balance between pro-fibrotic cytokines and proteins such as TGF-β versus anti-fibrotic proteins such as IFN-γ. During wound healing, IFN-γ released by T cells downregulate the expression of matrix genes. But, chronic inflammatory reactions induced by a high dose of IR cause long term deposition of extracellular matrix components including collagen (Zhao Weiling and Robbins

ip

t

2009). Then, collagen deposition is amplified, resulting in enhanced formation of myofibroblasts as the main source of extracellular matrix proteins. During normal wound healing,

cr

myofibroblasts undergo apoptosis, but during fibrosis, increased fibrogenic to anti-fibrogenic

us

agents ratio is associated with resistance of this cell to apoptosis and accumulation of collagen in

an

the extracellular matrix (Wynn TA and Ramalingam 2012).

The immune responses that are induced post IR-exposure mediate the radiation-induced fibrosis

M

over a long period of time. Upregulation of pro-inflammatory cytokines (e.g. TNFα, IL1, IL-4,

d

IL6, IL-13), fibrogenic cytokines like TGF-β, chemokines (e.g. MCP-1, MIP-1beta), microRNA-

pt e

21, peroxisome proliferator-activated receptors (PPARs), acute phase proteins (SAP), vascular endothelial growth factor (VEGF), renin-angiotensin-system, and platelet-derived growth factor

ce

(PDGF) in the irradiated tissue are involved in the proliferation of fibroblasts and the synthesis of protein matrix and extracellular matrix metalloproteinases (Wynn T 2008; Yamada et al.

Ac

2013).

There are several studies which claimed that TGF-β1 is responsible for normal tissue remodeling through the initiation, development, and maintenance of fibrosis following irradiation (Minshall et al. 1997; Martin et al. 2000; Wynn TA 2007). The most important signaling pathways for TGF-β1 which stimulates fibrosis after exposure to radiation are TGF-β1-smad2/3 and TGF-β1Rho/ROCK pathways (Bourgier C. et al. 2005; Pohlers et al. 2009). In addition to synthesis of

matrix metalloproteinases and matrixproteins, TGF-β1 suppresses the production of matrix proteases that accelerate accumulation of collagen and fibronectin in extracellular matrix (ECM) (Han et al. 2011; Ding et al. 2013). IL-4 and IL-13 are other cytokines that through IL-4R1 and IL-13R2 stimulate radiation induced fibrosis (Jakubzick et al. 2004; Chung S. I. et al. 2016). Radiation induced fibrosis in the lung can disrupt normal pulmonary actions, including gas

ip

t

exchange and ventilation (Ghafoori et al. 2008). Fibrosis in heart tissue leads to increased

cr

mechanical stiffness of the myocardium, abnormal thickening of the heart valves, destroys normal tissue architecture and disrupts electrical coupling that results in several changes in

us

ventricular properties, and systolic and diastolic dysfunction (Khan and Sheppard 2006; Song

an

and Wang 2015). Pathophysiological changes and ROS formation in the kidney that is associated with increased production of extracellular matrix components and renal hypertrophy may be

M

involved in diabetic nephropathy (Wolf 2004). Fibrosis in the liver can cause cirrhosis, distortion

d

of the hepatic architecture, liver failure, and portal hypertension. Advanced liver fibrosis may

pt e

require liver transplantation (Bataller and Brenner 2005). Inhibition of TGF-β, inflammatory cytokines and oxidative stress can be considered as a strategy

ce

for modulation of fibrosis process. ROS and RNS have a key role in the initiation and

Ac

maintenance of tissue remodeling after radiation treatment. Use of antioxidants and radioprotectors can reduce pathological effects of IR, such as fibrosis and vascular disease. Inhibitors of TGF-β such as Pentoxyfyllin, Relaxin, SB-525334 have been shown to exert important activity against lung fibrosis (Tsoutsou and Koukourakis 2006). Administration of some radioprotectors such as melatonin, amifostine and herbal agents such as curcumin and hesperidin have shown promising results (Erol et al. 2004; Sharma and Haldar 2006; Aghazadeh

et al. 2007; Shirazi et al. 2013), (Arora et al. 2014; Rezaeyan Abolhasan et al. 2016; Rezaeyan A 2016; Ghobadi et al. 2017; Haddadi GH 2017). In addition to clinical side effects, radiation accidents or terrorist radiation exposure are threats for the development of long term pathological changes from months to years after exposure to radiation (Gauter-Fleckenstein et al. 2010). Appropriate mitigators are needed to reduce these

ip

t

effects when administered after the exposure. Some agents such as genistein have shown

cr

promising mitigating effects on pneumonitis and fibrosis even when drug administration was initiated 1 week after exposure. This may be related to the antioxidant and anti-NF-κB activity of

us

genistein (Mahmood et al. 2011).

an

Vascular damage

M

Studies have shown that patients who have undergone radiotherapy for various malignancies such as breast cancer, lung cancer, lymphoma, and head and neck cancers have an increased risk

d

for developing vascular damage (Russell et al. 2009). Understanding of the mechanisms involved

pt e

in radiation-induced vascular damage is very important and can help modify the disease process. Studies have shown that the high radiation sensitivity of the vasculature has been linked to the

ce

endothelial dysfunction (Fajardo and Berthrong 1988; Milliat et al. 2006). Histological analyses

Ac

have shown that lesions in medium and large vessels exhibit features such as lipid accumulation, inflammation, and thrombosis (Weintraub Neal L. et al. 2010). Chronic inflammation and upregulation of matrix metalloproteinases increases intimal thickness and connective tissue content (Raffetto and Khalil 2008). Radiation increases the secretion of pro-inflammatory cytokines and upregulates adhesion molecules in the endothelium that recruits inflammatory cells to sites of vascular injury (Hallahan D et al. 1996; Liu Shinuo et al. 2012). These are associated with induction of continuous oxidative damage in vascular endothelial cells. Evidence

supports the hypothesis that acute and chronic inflammatory responses and subsequent oxidative damage have a central role in late brain vascular injury induced by IR, including cognitive impairment (Robbins MEC and Zhao 2004; Robbins Michael et al. 2012). Interventions designed to reduce chronic inflammatory responses can provide an opportunity to mitigate chronic radiation-induced brain injury, including demyelination and impairment of hippocampal

ip

t

neurogenesis (Wong C Shun and Van der Kogel 2004; Greene-Schloesser and Robbins 2012).

cr

Molecular processes contributing to vascular damage induced by IR includes increased production of pro-inflammatory cytokines, pro-thrombotic factors, adhesion molecules and

us

increased endothelin-1 secretion (Gaugler M 2014). Clinical and experimental studies have

an

shown that the inflammatory responses induced by radiation can accelerate thickening and clogging of cerebral and heart vascular, increasing the risk of vascular stenosis and stroke

M

(Stewart et al. 2010). Atherosclerosis is an inflammatory disease that is associated with

d

upregulation of P-selectin, E-selectin, ICAM-1 and VCAM-1, monocyte recruitment and foam

pt e

cell formation (Montecucco and Mach 2009). Irradiation of carotid arteries with fractionated or high single doses of radiation accelerates the development of atherosclerosis (Hoving et al.

ce

2008). These are associated with rapid upregulation of ICAM-1, VCAM-1, P-selectin and Eselectin gene expression during some hours, and may remain for a long time after exposure to IR

Ac

(Fliss and Menard 1994; Heckmann et al. 1998; Hallahan D. E. and Virudachalam 1999; Mollà et al. 2003). The long term expression of adhesion molecules such as ICAM-1 have a direct link with radiation dose in microvascular endothelium, and it is required for cell infiltration and monocyte recruitment (Hallahan Dennis E. and Virudachalam 1997). Moreover, the sustained upregulation of genes that are involved in remodeling of blood vessels such as plasminogen

activator inhibitor (PAI-1) in irradiated micro-vascular recipient veins can be involved in microvascular occlusion (Halle, Ekström, et al. 2010). NF-κB is a central regulator of inflammation and leukocyte adhesion in endothelial cells (Weintraub Neal L et al. 2010). Martin et al. claimed that the upregulated NF-κB in vascular wall cells, with specific localization to macrophages in the skin of head-and-neck cancer patients who

ip

t

had undergone radiotherapy since 500 weeks ago (Halle, Gabrielsen, et al. 2010). Upregulation

cr

of NF-κB leads to alterations in the production of inflammatory cytokines and chemokines, and leukocyte adhesion molecules. These changes in irradiated vasculature promote adherence of

us

leukocyte and platelet to endothelial cells that result in thrombus formation (Salame et al. 2000).

an

Another important factor in vascular damage following irradiation is the balance between NO

M

and ROS production in vascular endothelium. One of the most important molecular changes involved in endothelial dysfunction is decreased NO secretion and increased generation of ROS

d

(Bauersachs and Widder 2008). A major source of endothelial ROS is NADPH oxidases family,

pt e

including NOX1, NOX2, NOX4 and NOX5. Other sources include COX-2, mitochondria, xanthine oxidase, lipoxygenase and nitric oxide synthase (NOS). Crosstalk between NF-κB,

ce

COX-2, NADPH Oxidases enzymes, iNOS and mitochondria electron transport chains (ETCs)

Ac

are increasingly involved in cellular ROS production (Morgan and Liu 2011; Montezano and Touyz 2012; Yahyapour, Amini, et al. 2017). Studies have shown that IR decreases activity of eNOS in arteries (Qi et al. 1998; Soloviev Anatoly I et al. 2003; Soloviev Anatoly I. et al. 2003). NO has an inhibitory role on vascular muscle proliferation after pathological conditions (Garg and Hassid 1989). Proliferation of vascular muscle cells has a key role in repairing vessel wall following an injury. NO reduces intima formation by some mechanisms such as promoting cell death and some changes on the

expression of remodeling pathway genes, epigenetic mechanisms, and also interaction with ROS (Jeremy et al. 1999; Tsihlis et al. 2011). As NO has a role for prevention of thrombosis formation, the decrease in eNOS activity can cause ischemia and micro-vascular occlusion. Endothelial dysfunction and decrease of NO production after irradiation may cause an imbalance between growth-promoting and growth-inhibiting factors. These changes lead to vascular

ip

t

thickening and occlusion, dilation of the blood vessel lumen, and endothelial cell nuclear enlargement in irradiated tissues (Halle, Ekström, et al. 2010). It has been investigated that

cr

endothelial damage after exposure to radiation leads to a deficit of constitutive eNOS synthesis,

us

which promotes adherence of leukocyte and platelet to endothelial cell and thrombosis

an

development (Gaugler M-H et al. 2005; Gaugler M 2014). Experimental studies have revealed a thickening of intimal and medial vascular layers in irradiated vessels. It is possible that increased

M

secretion of inflammatory cytokines and growth factors, and also suppression of eNOS are

d

involved in these processes (Sugihara et al. 1999).

pt e

The most important concern for vascular damage during radiation treatment is related to ischemia and necrosis of brain white matter and heart cells. Chronic radiation-induced brain

ce

injury is characterized by vascular damage, demyelination, and white matter necrosis. Studies have shown that the irradiation of rat brains with 17.5-25 Gy of X-rays lead to a time and dose-

Ac

dependent vascular/glial changes, reduction in the endothelial cell and vascular density and the hypertrophy of the perivascular astrocytes. The severity of these changes are highly correlated with numbers of necrosis cells (Calvo et al. 1988; Reinhold et al. 1990). After radiation injury in the brain, microglia becomes activated and elevates the production of ROS, and pro-inflammatory cytokines and chemokines such as IL-1β, IL-6, and TNFα, and also MCP-1 and ICAM-1. These factors mediate long term neuro-inflammation and may be

associated with impairment of hippocampal neurogenesis and cognitive function. COX-2 is another gene that is induced in astrocyte and microglial cells and modulates brain inflammation after irradiation (Kyrkanides et al. 2002). Some clinical studies have suggested some changes such as progressive cognitive impairment after brain radiotherapy or chemotherapy among the patient survivors (Giovagnoli and Boiardi

ip

t

1994; Meyers and Brown 2006). These complications resulted in cerebral and spinal cord

cr

radionecrosis, which cause damage to white matter, and is associated with severe vascular lesions such as fibrinoid vascular necrosis, hemorrhage, stenosis and thrombosis (Soussain et al.

us

2009).

an

Several common NF-κB modulators such as melatonin, pravastatin, aspirin, omega-3 fatty acids,

M

and PPARα ligand, can be proposed for inhibition of VCAM-1 and E-select in vascular damage followed by irradiation (Weber et al. 1995; Mishra et al. 2004; Ramanan et al. 2008; Holler et al.

d

2009; Hu et al. 2013). Inhibition of the other pathways involved in vascular oxidative damage,

pt e

such as renin-angiotensin system can be regarded as a target for ameliorating of vascular damage

Ac

2014).

ce

after radiation therapy (Sun Yao 2002; Cohen et al. 2010; Medhora et al. 2012; Mahmood et al.

Inflammatory responses in radiation-induced normal tissue toxicity Hematopoietic system Hematopoietic system injury is one of the most common limiting factors for patients undergoing radiation therapy and is the most important cause of mortality after nuclear or radiological disasters. Moreover, hematopoietic malignancies such as leukemia, known as the most prevalent

cancers which have been reported among irradiated peoples. Evidences have revealed that some mechanisms which are activated after exposure to IR, play a key role in hematopoietic stem cells (HSCs) injury and mortality rate. Primary studies showed that whole body irradiation causes induction of chronic oxidative stress in hematopoietic stem cells. Analyses showed a role for mitochondria and some other ROS/NO producing enzymes in hematopoiesis regulation. The

ip

t

results stated that NADPH oxidase induced ROS production has more important role in HSCs regulation compared to other factors such as mitochondria (Piccoli et al. 2005; Piccoli et al.

cr

2007). The evaluation of different source of ROS production in HSCs including NADPH

us

Oxidases (NOX1-NOX5), cyclooxygenases, lipoxygenases, and mitochondrial electron transport

an

chain-1 (ETC1) showed that NOXs system is primarily responsible for the increased ROS production in HSCs following irradiation. The results indicated that among five different NOXs

M

enzymes, NOX4 significantly upregulated in bone marrow after irradiation. Moreover, results showed that increased NOX4 gene expression was prolonged upto 8 weeks post irradiation. This

pt e

d

indicated that the long term upregulation of NOX4 is a pathway for acute and late effects of IR exposure in the induction of hematopoietic stem cells senescence and residual bone marrow

ce

damages (Wang Y et al. 2010; Pazhanisamy et al. 2011). In addition to ROS, nitric oxide is another mediator for division, survival and mobilization of

Ac

hematopoietic stem and progenitor cells (Aicher et al. 2003; Michurina et al. 2004; Gangoiti et al. 2008). Several experiments have shown that exposure to radiation can elevate the production of NO in irradiated bone marrow cells. The elevated NO production that in itself depends on the presence of inflammatory mediators leads to reduction of numbers of stem cells and their progeny (Punjabi et al. 1994; Aicher et al. 2003). This can cause detrimental effects on the immune responses following exposure to IR (Sato K et al. 2007). Increased DNA damage

associated with suppression of DNA repair pathways was considered as roles of NO in development of genomic instability and increase of cancer risk (Mikhailenko et al. 2013; Mikhailenko and Muzalov 2013; Rezapoor et al. 2017). Several studies have shown that administration of some radioprotectors and mitigators can alleviate

radiation

induced

hematopoietic

toxicity and

improve

the

survival

rate.

ip

t

Supplementation with antioxidants before irradiation results in decreased apoptosis and TGF-β1

cr

mRNA expression in the bone marrow (Wambi et al. 2008). Surprisingly, Brown et al. indicated that antioxidant supplement diet administration starting at 24 hours after whole body irradiation

us

with 8 Gy bring about a better survival when compared to sooner or later antioxidant

an

administration started after the irradiation, and also before irradiation. The results indicated that mitigating of radiation injury is mediated by a reduction in ROS production and suppression of

M

some pro-apoptosis pathways including MEK/ERK in bone marrow cells in response to

d

increased levels of some cytokines and growth factors such as IL-3 and granulocyte-macrophage

pt e

colony-stimulating factor (GM-CSF) (Brown et al. 2010). In another study, Sato et al. showed that the best survival with administration of ascorbic acid after exposure can be obtained at 12

ce

hours after whole body irradiation. This study showed that treatment with ascorbic acid after whole body irradiation with 7.5 Gy can reduce apoptosis in bone marrow cells and damage in

Ac

hematopoietic function. Post-exposure treatment with ascorbic acid, ON 01210.Na (a chlorobenzylsulfone derivative) and antioxidant N-Acetyl-cysteine have shown ability to improve hematological parameters including peripheral white blood cell, platelet counts and GM-CFU counts. Also, supplements with these agents suppressed apoptotic cell death and DNA damage in bone marrow cells, and decreased inflammatory cytokines such as IL-1β, IL-6, IFN-γ

which are involved in ROS and RNS production (Jia et al. 2010; Suman et al. 2012; Sato T et al. 2015). The results of these studies indicated that ROS and RNS production after exposure to irradiation, that in itself is mediated with inflammatory responses or some other unknown pathways play a key role in acute radiation syndromes, genomic instability and maybe non-cancerous diseases.

ip

t

Improvement of survival rate by mitigators and antioxidants open a new window for

cr

management of side effects of radiotherapy and consequences of radiation accidents. Management of immune responses using appropriate supplements can improve lifesaving and

an

us

decrease in chronic inflammation, cancer and non-cancerous diseases.

M

Radiation dermatitis

d

Radiation dermatitis (also known as radiodermatitis or radiation skin burning) is associated with

pt e

epidermal basal cell and endothelial cell damage, commonly occurs following radiotherapy. Radiodermatitis can be divided into acute and chronic dermatitis. The most important changes

ce

associated with acute dermatitis are erythema, dry desquamation, and moist desquamation that result in epidermal necrosis, fibrinous exudates, pain and ulcer. Chronic detrimental changes

Ac

induced by IR may develop during months to years after exposure. These pathological changes commonly included are hypopigmentation or hyperpigmentation, damage to hair follicles and sebaceous glands, persistent telangiectasia and fibrosis. The most serious chronic complication of radiation therapy on the skin is the development of ulceration during months to years after the end of treatment (Hymes et al. 2006; Salvo et al. 2010).

Evidences have remarked that radiation induced dermatitis affects approximately 95 percent of patients undergoing radiotherapy, especially patients with head and neck cancer, breast and lung malignancies (Ryan 2012). The mechanisms involved in radiation dermatitis and strategies for managing its side effects have been indicated in several studies. Studies have shown that inflammatory responses have a central role in dermatitis associated with radiotherapy. Acute

ip

t

radiodermatitis occurs within several hours to weeks after beginning of radiation treatment that is induced by initiation of inflammatory responses in the epidermis and dermis. Acute skin

cr

dermatitis is associated with vascular inflammation and vasodilation as well as swelling and

us

sloughing of epithelial cells (Mancini and Sonis 2014; Arron 2016). Inflammatory responses are

an

induced by structural tissue damage, necrosis and irreversible DSBs in nuclear and mitochondrial DNA. Severe inflammation after irradiation may result in a failure of the skin's barrier function

M

and leads to bacterial infection (Flour 2009). Bacterial infections like Staphylococcus aureus can exacerbate dermatitis through activation of lymphocytes T and subsequent cytokine release (Hill

pt e

d

et al. 2004).

Previous studies have revealed that IL-1, IL-6, TNF-α, TGF-β, IL-8 and eotaxin are the major

ce

cytokines and chemokines that regulate the response of skin cells to IR (Müller and Meineke 2007). Janko et al. showed that IL-1 has an important role in the development of radiodermatitis.

Ac

They showed that a lack of IL-1 or the IL-1 receptor in mice leads to less pathological changes in the skin. This study offered a potential therapeutic targeting of IL-1 for control of the severity of radiation dermatitis (Janko et al. 2012). The production of IL-1 in skin is primarily regulated by monocytes, macrophages, fibroblasts keratinocytes, and numerous other immune mediators. (Liu W et al. 2006; Janko et al. 2012).

In addition to inflammation, fibrosis process induced by TGF-β and other cytokines has an important role in promotion of chronic radiation dermatitis. Inhibition of TGF-β and Smad3 reduce scarring and local inflammatory infiltrate, and also accelerate wound healing process (Ashcroft and Roberts 2000; Flanders et al. 2002; Flanders et al. 2003; Flanders et al. 2008). Although, TGF-β is known as central player in the fibrotic process, other cytokines and growth

ip

t

factors such as IL-1, IL-4, IL-13, TNF-α , platelet-derived growth factor (PDGF), insulin-like growth factor (IGF-1) and connective tissue growth factors (CTGF) are involved in this process

cr

in the skin (Martin et al. 2000).

us

Several studies have been conducted to assess the outcome of different interventions such as

an

administration of antioxidants, radioprotectors, corticosteroids, lotions, creams and antiinflammatory drugs for the prevention and management of radiodermatitis. However, many

M

studies have not shown impressive results for antioxidants compared to anti-inflammatory

d

agents. This may result in more important roles of immune responses than other factors such as

pt e

direct effects of ROS produced by radiation. Halperin et al. in a randomized trial study have reported that there is no discernible benefit for administration of vitamin C to prevent radiation

ce

dermatitis (Halperin et al. 1993). However, the evaluation of the impact of amifostine against acute dermatitis for pelvic cancer patients showed that severity of dermatitis is significantly

Ac

reduced in patients who have received amifostine (Kouvaris JR et al. 2001; Kouvaris J et al. 2002). In another study in a randomized, double-blind, controlled clinical trial radiotherapy, oral administration of curcumin showed that it can reduce the severity of dermatitis in breast cancer patients (Ryan et al. 2013). The effects in of radiation on skin reactions are thought to be caused by different changes such as vasoconstriction, reduced capillary permeability, and inhibition of leukocyte migration.

Inhibition of pro-inflammatory cytokines such as IL-1α/β, TNF-α, IL-6 and MCP-1, as well as TGF-β1 and COX-2 can improve the tolerance of skin to radiotherapy (Chen et al. 2010). Corticosteroids often prescribed in both the prevention and management of radiation dermatitis. Corticosteroids such as mometasonefuroate, emollient and betamethasone creams have shown to be effective in reducing acute radiation dermatitis during radiation therapy (Boström et al. 2001;

ip

t

Ulff et al. 2013). Beetz et al. suggested that corticosteroids through inhibition of IL-6 expression in irradiated human epithelial cells can reduce acute radiation dermatitis (BEETZ and P. KIND

cr

1997). Inhibition of COX-2 by celecoxib can reduce inflammation of the dermis, MCP-1 mRNA

us

expression and skin damages in irradiated skin (Liang et al. 2003; Mohsen Cheki 2018). The

an

targeting of TLR-5 showed a reduction in NF-κB expression and is effective in prevention of dermatitis in head and neck cancer patients that have undergone radiotherapy (Burdelya et al.

M

2012). Evidences have demonstrated that using Aloe Vera gel improves radiation induced dermatitis and skin burning, and accelerates wound healing, especially for breast and head and

pt e

d

neck cancer patients (Williams et al. 1996; Richardson et al. 2005; Atiba et al. 2011). Probably, anti-inflammatory effect of Aloe Vera gel has a key role in alleviation of skin toxicity (Yagi et

ce

al. 2002; Reuter et al. 2008). Mustafa et al. showed that administration of zinc reduces the severity of radiodermatitis and skin damage such as epidermal atrophy, dermal degeneration, and

Ac

hair follicle atrophy in the rats. Moreover, administration of zinc can delay the start of dermatitis (Ertekin et al. 2004).

Role of inflammatory responses to radiation-induced digestive tract injury Digestive system complications are a limiting factor for patients undergoing radiotherapy for thoracic, abdominal, or pelvic malignancies. These complications may limit the completion of radiotherapy and may reduce efficiency of cancer treatment. Moreover, radiation induced acute

gastrointestinal injury is a life threatening sickness in people that have exposed to a heavy dose of IR (Elhammali et al. 2015). Oral, gastric and bowel are the main organs of the digestive system that can be affected by different complications of radiation and chemotherapy (Shadad Abobakr K. et al. 2013). Acute effects may be observed during or shortly after irradiation, while late effects are experienced months to years after irradiation (Lips et al. 2008). Mucositis and

ip

t

xerostomia in patients receiving radiotherapy for head and neck cancers are the most important oral complications. Moreover, gastritis, telangiectasias and bleeding in gastric, and also

cr

mucositis and fibrosis are the most common complications induced by radiation treatment in the

us

intestine. It seems that inflammatory cytokines and some physiological changes are responsible

an

for these acute and late symptoms following exposure to radiation.

M

Oral

Oral complications during and after of radiotherapy of head and neck malignancies primarily are

d

caused by the detrimental effects of IR on the mucosa and salivary glands in the oral cavity and

pt e

oropharynx. These damages can lead to an inflammatory condition in the mucosa and periodontium called mucositis (Vissink, Burlage, et al. 2003). It seems that mucositis and

ce

xerostomia are the most common adverse effects of head and neck cancer radiotherapy (Vissink,

Ac

Jansma, et al. 2003). Mucositis is the inflammation, ulceration and swelling of the mucous membranes lining the digestive system that affects the patients at the pelvis and head and neck after undergoing radiation therapy (Shih et al. 2003; Keefe 2007). The initiation of mucositis is caused by the production of cell transcription factors such as NF-κB, MAPKs, COX-1 and COX2 which upregulate inflammatory cytokines, such as IL-1, IL-2, IL-6, TNF-α, β-actin and matrix metalloproteinases (MMPs) 1 and 3. These processes lead to the destruction of the mucosa (Keskek et al. 2006; Yeoh et al. 2006; Logan et al. 2007; Ong et al. 2010). Mucositis can be

associated with salivary hypo-function, necrosis and subsequent removal of dental pulp, infection, loss of taste and necrosis of the jaw bone. These complications in oral cavity can be due to high turnover rates and radiosensitivity of cells in oral mucosa. Mucositis manifests initially as erythema and may develop with necrosis, ulceration, and bleeding (Springer et al. 2005; Sciubba and Goldenberg 2006; Shiboski et al. 2007).

ip

t

Meirovitz et al. showed a correlation between IL-6 and IL-8 serum levels and severity of oral

cr

mucositis during head and neck cancer treatment (Meirovitz et al. 2010). While a study has proposed that there is no relationship between low-grade oral mucositis induced by

us

chemotherapy and the systemic plasma IL-8 level (Stokman et al. 2006). Also, a study has shown

an

no correlation between IL-1 and TNF-α serum levels and oral mucositis grade (Seyyednejad et al. 2012). Studies have indicated that acute mucositis is caused by the massive death of basal

M

cells, decrease in the number of epithelial cells, and inflammatory responses mediated by NF-κB,

d

pro-inflammatory cytokines, inflammasome and the ceramide pathway (Sonis 2004, 2007).

pt e

Moreover, endotoxins produced by resident bacteria on ulcerated surfaces may amplify the inflammatory responses induced by IR and enhance local injury (Donnelly et al. 2003).

ce

The use of immunomodulatory drugs, cytokines and radioprotectors (e.g. amifostine, glutamine),

Ac

and also antibiotics have been of much interest in the management of radiation-induced mucositis from many years ago. Administration of GM-CSF, IL-11 and TGF-β have provided an effective treatment protocols for preventing oral mucositis in patients with head and neck carcinoma that undergo radiotherapy (Kannan et al. 1997; Rosso et al. 1997; Raber-Durlacher et al. 2013). Also, several clinical trial studies have shown that the use of amifostine can preserve tissue function and can reduce the severity and duration of mucositis and xerostomia (Symonds et al. 1996; Bourhis et al. 2000; Brizel et al. 2000; Savarese et al. 2003). Oritiz et al. showed that

melatonin, through protection of mitochondria against radiation induced oxidative damage and suppression of the NF-κB/NLRP3 inflammasome signaling pathway can prevent development of oral mucositis (Ortiz et al. 2015). The use of low-energy laser as a noninvasive technique is another modality for the acceleration of wound healing, and alleviation of pain and severity of oral mucositis during and after chemo/radiotherapy (Sandoval et al. 2003).

ip

t

Gastric

cr

IR-induced gastritis is a serious complication in radiation therapy and can cause chronic gastric

us

inflammation and bleeding. The initial injury is characterized by acute inflammation of gastric mucosa. Although the pathogenesis of radiation-induced gastritis is not fully understood, it is

an

thought that increased levels of inflammatory cytokines and growth factors are involved in this

M

process. Some evidences have shown that TNFα, IL-1β, IL-21, and cyclooxygenases play a key role in the development of gastritis (Li GQ et al. 2006; Santos et al. 2012; Nishiura et al. 2013).

d

The presence of Helicobacter pylori may be related to gastritis caused by radiation (Abrunhosa-

pt e

Branquinho et al. 2015). Chronic radiation gastritis can cause the multiple telangiectasia and gastric bleeding usually during 2-7 months after treatment It is thought that acute vasculopathy

ce

may be involved in obliteration of endarteritis that leading to mucosal ulceration and bleeding. A

Ac

similar sign is seen in the rectum, intestine, and bladder following pelvic irradiation (Grover and Johnson 1997). The long term duration of these symptoms results in anemia and tumor progression. But taking appropriate drugs can relieve the complications effectively (RodríguezLago et al. 2013). In addition to external radiotherapy, gastrointestinal injury is an important side effect of internal radiation therapy, systemic chemotherapy and chemoradiation therapy for hepatic tumor (Chon et al. 2011). Selective internal radiation therapy (SIRT) by yttrium-90 (Y-90), can provide more

effective treatment and less side effects compared to external beam radiation, while selectively providing local irradiation to the tumor or metastases. However, this protocol may cause gastrointestinal ulcer, cholecystitis, pancreatitis, bone marrow toxicity, and inflammation. Gastrointestinal complications are the most common complications compared to other organs that have been reported (Crowder et al. 2009). Mucosal ulceration caused by Y-90 is seen as

ip

t

necroinflammatory slough associated with granulated tissue that occurs mostly in the gastric antrum, pylorus, and duodenum (Konda et al. 2009; Zimmermann et al. 2009; Omed et al. 2010).

cr

Evidences have revealed that these side effects typically are caused by significant inflammatory

us

responses (Naymagon et al. 2010). Argon plasma coagulation and administration of growth

an

hormone and oral prednisolone (an anti-inflammatory drug) have shown ability to control bleeding caused by radiation-induced gastritis (Shukuwa et al. 2007; Zhang Lan et al. 2012; Yun

M

et al. 2015; Zhang Liang et al. 2015).

d

Radiation Enteritis

pt e

Radiation damage to the intestine occurs following whole body irradiation or radiotherapy of the abdomen and pelvis for gastrointestinal, urological and gynaecological cancers. Irradiation of

ce

these organs may cause acute and long term symptoms such as pain, nausea, diarrhea, weight

Ac

loss, bloating, malabsorption, and rectal bleeding (Stacey and Green 2013; Abdollahi H. 2014; Abdollahi Hamid et al. 2015). These symptoms arise from massive tissue damage and inflammatory responses that reaches its peak at weeks 4 to 5. While the long term symptoms ranges to 30 years after exposure to radiation (Nguyen et al. 2002). Chronic radiation damages to bowel has been reported for 50% of patients receiving pelvic radiotherapy and characterized by inflammation, mucosal cell loss, crypt abscess and swelling of the endothelial lining of arterioles (Theis et al. 2010). Symptoms resulting from intestinal damage are mediated by immune

responses and chronic inflammation that are directly related to the radiation dose. These inflammatory responses may lead to mucositis in the gastrointestinal system (François et al. 2013). Alimentary tract mucositis is one of the most common side effects and dose-limiting factors for almost all patients undergoing radiation therapy and chemotherapy for gastrointestinal malignancies. Development of mucositis in the intestine arises from the cytotoxic effects of

ip

t

radiation on the basal cell of epithelium. These cells are vulnerable to the effects of IR because

cr

of their high mitotic index and cell turnover rate (Theis et al. 2010).

There are some suggestions that persistent oxidative stress is implicated in long term intestinal

us

damages. Continuous ROS production correlates with upregulation of NADPH Oxidase and

an

iNOS, and also increased mitochondria activity in intestine after exposure to radiation (Zhou H et al. 2008; Morgan and Liu 2011). Continuous oxidative damages can cause mutations and

M

genomic instability in genes implicated in gastrointestinal carcinogenesis. This can lead to long-

d

term changes in intestinal stem cells dynamics (Datta et al. 2012).

pt e

Use of anti-inflammatory cytokines and other modifiers such as amino acid supplementation, colony-stimulating factors, surgical and laser therapy have proposed for relief of the side effects

ce

of mucositis (Köstler et al. 2001; Regimbeau et al. 2001; Von Bültzingslöwen et al. 2006;

Ac

Shadad Abobakr K et al. 2013). The short periods of use of growth factor and anti-inflammatory agents such as COX-2 and TNF-α inhibitors, growth factors like TGF-β, IL-1, IL-11, stem cell factor, keratinocyte growth factors, fibroblast growth factor, acidic and basic fibroblast growth factors FGFs, VEGF, and IGF-1 appear to ameliorate intestinal toxicity by reduction of early and late alimentary tract mucosal complications (Okunieff et al. 2005). As well as inhibition of NFκB, COX-2, iNOS and NADPH Oxidase can decrease normal tissue damages and enhance the tumor response to treatment (Erbil et al. 2006; Murakami and Ohigashi 2007; Yi et al. 2014).

A study showed that pre-treatment with ascorbic acid before the whole body irradiation with 13Gy can cause 20% survival compared to 0% survival without ascorbic acid administration. A combination of administration of ascorbic acid pre and post-irradiation leads to 100% survival. Immunohistochemistry analysis showed an increase of TNF-α level in the intestinal tissue and IL-6 in the plasma. This study suggested that TNF-α induced inflammatory responses (but not

ip

t

IL-1β or IL-6) have a key role in radiation-induced intestinal oxidative injury and radiation gastrointestinal syndrome. A combination of administration of ascorbic acid pre and post-

cr

irradiation could significantly reduce TNF-α level and free radical metabolite levels in the small

us

intestine. Authors proposed that the boosting pre-treatment associated with post irradiation

an

treatment by ascorbic acid is necessary to scavenging of ROS generated after irradiation (Ito et al. 2013).

M

Another important side effect of radiotherapy on intestine is fibrosis. TGF-β signaling through the Rho/Rock and Smad3 pathways is involved in the development of sustained fibrogenic

pt e

d

differentiation and small intestinal fibrosis after irradiation (Haydont et al. 2005; Zhu et al. 2011). Bourgier et al. reported that the intestinal cells isolated from radiation enteritis exhibit a

ce

high CTGF level, increased collagen secretory capacity and expression of Rho family. Authors proposed that the inhibition of this signaling pathway can protect intestinal fibrosis against

Ac

radiation exposure (Bourgier C et al. 2005). Proctitis

Proctitis refers to inflammation of the lower parts of the colon including primarily, the sigmoid colon and the rectum. Acute radiation proctitis most commonly occurs in the first few weeks and chronic proctitis begin several months to years after radiation therapy such as external radiotherapy and brachytherapy for pelvic cancers such as prostate, cervical and colon cancers.

Acute radiation proctitis results from direct damage of the epithelium but chronic proctitis occurs because of damage to the blood vessels, especially the small vessels which supply the cells of the sigmoid colon and the rectum. The most important mechanisms involved in the development of chronic radiation proctitis are long term inflammation and free radical production (Do et al. 2011). Indaram et al. indicated that the mucosal levels of IL-2, IL-6, and IL-8 are abnormally

ip

t

high in patients with proctitis that have undergone radiation therapy (Indaram et al. 2000). Although, early changes may not lead to obvious symptoms but If inflammation progresses, it

cr

may evolve to mucosal ulceration, ischemia, ulceration, telangiectasia and bleeding, which can

us

extend decreasing blood cells in patients that underwent radiotherapy (Grover and Johnson

an

1997).

The inhibition of prostaglandin synthesis with administration of anti-inflammatory agents such

M

as mesalamine, prednisone, betamethasone, hydrocortisone, and metronidazole alleviates chronic proctitis (Do et al. 2011). It is thought that free radical production during long term inflammation

pt e

d

is one of the most important mechanisms involved in the development of chronic radiation proctitis (Do et al. 2011). The use of radioprotectors and antioxidants is another modality for

ce

amelioration of long lasting adverse effects of radiation treatment on the lower bowel and rectum. The randomized trials have proposed that pretreatment with amifostine can reduce

Ac

toxicity in the lower bowel and rectum in patients undergoing radiotherapy for pelvic cancers without tumor protection (Liu T et al. 1992; Athanassiou et al. 2003). Moreover, the use of antioxidants agents such as vitamin A, E and C have shown ability to decrease the rate of proctitis symptoms such as hemorrhage, diarrhea and urgency (Kennedy et al. 2001; Patel et al. 2009).

Radiation induced lung injury The lung is a radiosensitive organ, especially for late effects of radiotherapy such as pneumonitis and fibrosis. On the other hand, lung cancer is one of the most common malignancies among cancer patients. Moreover, more than 60% of all patients with lung cancer needs radiation therapy for their disease (Tyldesley et al. 2001). It was estimated that up to 30% of patients with

ip

t

non-small cell lung carcinoma (NSCLC) are needed for management of lung tissue toxicity

cr

(Kong et al. 2006; Schallenkamp et al. 2007). So, a knowledge of the mechanisms of lung injury following radiation treatment is necessary for management of complications that are associated

us

with radiotherapy.

an

Pneumonitis as one of two important detrimental effects of radiation therapy on lung tissue, usually appears during 1 to 6 months after treatment. The most important symptoms of radiation

M

induced pneumonitis consist of chest pain, dyspnea congestion and cough (Wang S et al. 2006).

d

Pneumonitis is an acute reaction of lung to high doses of IR that is mediated by inflammatory

pt e

cells. Macrophages, mast cells and lymphocytes play a key role in this phenomena. These cells secrete inflammatory cytokines such as IL-1, IL-4, IL-6, IL-13 and TNF-α that are responsible

ce

for appearance of lung pneumonitis (Ward PA and Hunninghake 1998; Wirsdörfer and Jendrossek 2016).

Ac

Another important side effect of radiotherapy in the lung is fibrosis that occurs during months to years after radiotherapy. Fibrosis is characterized by deposition of collagen, damage to vascular structure and change in normal function of lung that may threaten the life of patients (Yarnold and Brotons 2010). Targeting of IL-4 and IL-13 is deemed strategic for management of lung fibrosis (Chung Su I. et al. 2016; Groves et al. 2016). Moreover, management of NF-κB, COX-2 and inflammatory cytokines have been suggested for this aim. So, several studies have evaluated

different antioxidants, radioprotectors and mitigators for amelioration of lung toxicity following exposure to radiation. Melatonin has shown a protective effect on inflammatory responses and pathological damage induced by radiation (Tahamtan et al. 2015; M. Najafi 2017; Najafi M et al. 2017). Natural antioxidants have attracted much interests for the management of radiation toxicity in the lung. Some herbal compounds such as hesperidin, curcumin, soy isoflavones and

ip

t

silibinin have been brought to our notice as radioprotective agents for amelioration of oxidative damage and pathological changes after exposure to radiation (Lee JC et al. 2010; Cho et al. 2013;

cr

Hillman et al. 2013; Son et al. 2015; Rezaeyan A. et al. 2016; Haddadi GH 2017).

us

Radiation induced heart diseases

an

Heart disease is one of the most common diseases and one of the main causes of mortality in the

M

general population (Yusuf et al. 2001). The most common types of heart disease are inflammation, fibrosis, atherosclerosis and also diseases related to coronary and carotid arteries

d

(Hansson 2005). There is a large body of data related to A-bomb survivors in Hiroshima and

pt e

Nagasaki which reported significant increases in heart diseases (Chai Y et al. 2012; Chai Y et al. 2013). Moreover, Heart diseases in patients having undergone radiotherapy to treat breast cancer

ce

have had a significant rise. Heart diseases in this population is higher than the general population

Ac

and it is one of the major causes of death during long years after the treatment. The incidence of these diseases was related to the radiation dose received by individuals and other risk factors for heart disease such as blood cholesterol and obesity (Bestor 2000; Bird 2009; Bonasio et al. 2010). The most important heart damages due to radiotherapy have been reported in the form of cardiac fibrosis, coronary and carotid vascular injury, atherosclerosis, heart valves disease and etc., that result in high incidence of heart attacks among people who had been exposed. These

complications lead to an increase in mortality among patients that have undergone radiotherapy (Zaratiegui et al. 2007). Mechanisms of heart disease due to exposure to radiation are not fully understood; although, symptoms are similar to those of other causes of heart diseases. Nevertheless, studies have suggested that immune system cells have a central role in cardiovascular disorders after exposure

ip

t

to IR. After heart irradiation, pericardium is the most sensitive area that is damaged. The main

cr

type of complication seen in this area is fibrosis (Zaratiegui et al. 2007). Fibrosis can interfere with blood supply and increase the probability of ischemia and heart attack (MOSLEMI and

us

MOTEVALIZADEH 2009). The main factors that are involved in cardiovascular fibrosis due to

an

radiation exposure are TGF-β and IL-1β. Increased level of these cytokines, especially TGF-β steadily continues for many years after exposure to acute radiation (Najafi M. et al. 2014). In

M

addition to NO production, these cytokines can increase the production of collagen in the tissues

d

that is the main cause of fibrosis after exposure to radiation (Das and Singal 2004; Bonasio et al.

pt e

2010). Thus, high levels of TGF-β in cancer radiotherapy patients, which typically occur, may cause damages to the coronary arteries function. All of these changes augment the risk of heart

ce

ischemia and stroke during the years after radiotherapy (Kalinich et al. 1989).

Ac

Mast cells are one of the most important immune cells that play an important role in radiationinduced heart diseases. There is a clear evidence that mast cells are involved in some heart problems such as inflammation, atherosclerosis, fibrosis and myocardial degeneration, resulting in tissue remodeling and increased risk of heart failure (Boerma et al. 2005). Abnormal increase in the numbers of these cells have been reported after exposure to thoracic irradiation (Ward WF et al. 1990). Moreover, mast cells hyperplasia upregulates the expression of genes such as endothelin-1 (ET-1). ET-1 is a powerful vasoconstrictor peptide that promotes inflammation and

fibrosis. Long term overexpression of ET-1 causes cardiac hypertrophy leading to many forms of heart disease, including hypertension, valvar disease and heart failure (Frey et al. 2004). Some radioprotectors such as melatonin and hesperidin have been recommended for amelioration of heart damage induced by IR (Gurses et al. 2014; Rezaeyan Abolhasan et al. 2016).

t

Radiation myelopathy

ip

Radiation myelopathy is a detrimental effect of radiation on spinal cord that is characterized by

cr

white matter lesions. Based on clinical observations, myelopathy after exposure to high doses of

us

radiotherapy occurs at two distinct clinical entities. Early delayed injury occurs during 2 to 4 months after exposure and is characterized by extremities upon neck flexion. However, this

an

damage heals after a few months completely (Mul et al. 2012). In contrast to early radiation

M

myelopathy, late myelopathy induced by radiation is typically permanent and irreversible. The symptoms of late myelopathy are very different including reactive gliosis, demyelination and

pt e

diagnosis (Wong C. S. et al. 1994).

d

white matter necrosis that may lead to death of irradiated patients up to 5 years after the date of

Mechanisms of radiation induced myelopathy is not completely recognized. A few studies have

ce

reported that massive necrosis over a long time after radiation plays a key role in this

Ac

phenomena. It is well established that upregulation of genes involved in hypoxia such as HIF1-α and VEGF are implicated in necrosis of white matter. Upregulation of hypoxia-inducible factor 1-alpha (HIF1-α), VEGF, matrix metalloprotease-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1) are associated with disruption of blood spinal cord barrier that lead to necrosis (Tsao et al. 1999; Li YQ et al. 2001; Nordal and Wong 2004; Lee JY et al. 2012).

Targeting factors involved in hypoxia and inflammation are promising strategies for mitigation of radiation induced myelopathy. Suppression of TNF-α and ICAM-1 seem to be able to ameliorate inflammation and permeability in a rat cranial (Yuan et al. 2003). Also, amelioration of prostaglandins production by melatonin have shown promising results (Aghazadeh et al. 2007; Shirazi et al. 2009; Haddadi et al. 2013; Rasoul et al. 2017). In clinical studies, inhibition of

ip

t

VEGF has improved symptoms of myelopathy in radiotherapy patients (Chamberlain et al. 2011;

cr

Psimaras et al. 2016).

us

Conclusion

As stated in this review, inflammation is responsible for several side effects and disorders that

an

are seen in association with radiotherapy or radiation accident. IR causes upregulation and

M

downregulation of several cytokines and immune mediators. Exposure to high doses of IR such as those seen in radiotherapy cause stimulation of inflammatory and anti-inflammatory reactions.

d

Both of these reactions are responsible for most of radiotherapy complications. Although the

pt e

exact mechanisms of some disorders such as diabetes after exposure to radiation remain to be elucidated, it seems that inflammatory reactions are involved. Use of some mitigators such as

ce

herbal agents has borne satisfactory results. However, selecting an appropriate radioprotector

Ac

mitigator for a specified organ depends on the mechanisms involved in radiation response in that organ. Hence, a knowledge of radiation response to IR in different organs is useful for the management of detrimental effects of radiotherapy.

The authors report no conflicts of interest

References

Ac

ce

pt e

d

M

an

us

cr

ip

t

Abderrazak A, Syrovets T, Couchie D, El Hadri K, Friguet B, Simmet T, Rouis M. 2015. NLRP3 inflammasome: from a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox biology. 4:296-307. Abdollahi H. 2014. Probiotic-based protection of normal tissues during radiotherapy. Nutrition (Burbank, Los Angeles County, Calif). 30(4):495-496. eng. Abdollahi H, Atashzar M, Amini M. 2015. The potential use of biogas producing microorganisms in radiation protection. Journal of Medical Hypotheses and Ideas. 9(2):67-71. Abrunhosa-Branquinho A, Barata P, Vitorino E, Oliveira E, Bujor L, Jorge M. 2015. A Case of PostRadiotherapy Gastritis: Radiation Does Not Explain Everything. Case reports in oncology. 8(1):9-14. Aggarwal BB, Shishodia S, Sandur SK, Pandey MK, Sethi G. 2006. Inflammation and cancer: how hot is the link? Biochemical pharmacology. 72(11):1605-1621. Aghazadeh S, Azarnia M, Shirazi A, Mahdavi SR, Zangii BM. 2007. Melatonin as a protective agent in spinal cord damage after gamma irradiation. Reports of Practical Oncology & Radiotherapy. 12(2):95-99. Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C, Technau-Ihling K, Zeiher AM, Dimmeler S. 2003. Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nature medicine. 9(11):1370-1376. Allam R, Lawlor KE, Yu ECW, Mildenhall AL, Moujalled DM, Lewis RS, Ke F, Mason KD, White MJ, Stacey KJ. 2014. Mitochondrial apoptosis is dispensable for NLRP3 inflammasome activation but non‐apoptotic caspase‐8 is required for inflammasome priming. EMBO reports. 15(9):982-990. Effects of an Amifostine analogue on radiation induced lung inflammation and fibrosis. Proceedings of the international conference on radiation biology: frontiers in radiobiology-immunomodulation, countermeasures and therapeutics: abstract book, souvenir and scientific programme; 2014. Arron S. 2016. Anatomy of the Skin and Pathophysiology of Radiation Dermatitis. Skin Care in Radiation Oncology. Springer; p. 9-14. Ashcroft GS, Roberts AB. 2000. Loss of Smad3 modulates wound healing. Cytokine & growth factor reviews. 11(1):125-131. Athanassiou H, Antonadou D, Coliarakis N, Kouveli A, Synodinou M, Paraskevaidis M, Sarris G, Georgakopoulos GR, Panousaki K, Karageorgis P. 2003. Protective effect of amifostine during fractionated radiotherapy in patients with pelvic carcinomas: results of a randomized trial. International Journal of Radiation Oncology* Biology* Physics. 56(4):1154-1160. Atiba A, Nishimura M, Kakinuma S, Hiraoka T, Goryo M, Shimada Y, Ueno H, Uzuka Y. 2011. Aloe vera oral administration accelerates acute radiation-delayed wound healing by stimulating transforming growth factor-β and fibroblast growth factor production. The American Journal of Surgery. 201(6):809818. Bataller R, Brenner DA. 2005. Liver fibrosis. Journal of Clinical Investigation. 115(2):209-218. Batar B, Guven G, Eroz S, Bese NS, Guven M. 2016. Decreased DNA repair gene XRCC1 expression is associated with radiotherapy-induced acute side effects in breast cancer patients. Gene. 582(1):33-37. Bauersachs J, Widder JD. 2008. Endothelial dysfunction in heart failure. Pharmacological reports : PR. 60(1):119-126. eng. BEETZ GM, T. OPPEL, D. VAN BEUNINGEN, RU PETER*, P. KIND A. 1997. Induction of interleukin 6 by ionizing radiation in a human epithelial cell line: control by corticosteroids. International journal of radiation biology. 72(1):33-43. Bestor TH. 2000. The DNA methyltransferases of mammals. Human molecular genetics. 9(16):23952402.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Bird A. 2009. On the track of DNA methylation: an interview with Adrian Bird by Jane Gitschier. PLoS genetics. 5(10):e1000667-e1000667. Boerma M, Wang J, Wondergem J, Joseph J, Qiu X, Kennedy RH, Hauer-Jensen M. 2005. Influence of mast cells on structural and functional manifestations of radiation-induced heart disease. Cancer research. 65(8):3100-3107. eng. Bogdan C. 2015. Nitric oxide synthase in innate and adaptive immunity: an update. Trends in immunology. 36(3):161-178. Bonasio R, Tu S, Reinberg D. 2010. Molecular signals of epigenetic states. Science. 330(6004):612-616. Borger JH, Kemperman H, Smitt HS, Hart A, van Dongen J, Lebesque J, Bartelink H. 1994. Dose and volume effects on fibrosis after breast conservation therapy. International Journal of Radiation Oncology* Biology* Physics. 30(5):1073-1081. Boström Å, Lindman H, Swartling C, Berne B, Bergh J. 2001. Potent corticosteroid cream (mometasone furoate) significantly reduces acute radiation dermatitis: results from a double-blind, randomized study. Radiotherapy and Oncology. 59(3):257-265. Bourgier C, Haydont V, Milliat F, Francois A, Holler V, Lasser P, Bourhis J, Mathe D, Vozenin-Brotons M. 2005. Inhibition of Rho kinase modulates radiation induced fibrogenic phenotype in intestinal smooth muscle cells through alteration of the cytoskeleton and connective tissue growth factor expression. Gut. 54(3):336-343. Bourgier C, Haydont V, Milliat F, François A, Holler V, Lasser P, Bourhis J, Mathé D, Vozenin-Brotons M. 2005. Inhibition of Rho kinase modulates radiation induced fibrogenic phenotype in intestinal smooth muscle cells through alteration of the cytoskeleton and connective tissue growth factor expression. Gut. 54(3):336-343. eng. Bourhis J, De Crevoisier R, Abdulkarim B, Deutsch E, Lusinchi A, Luboinski B, Wibault P, Eschwege F. 2000. A randomized study of very accelerated radiotherapy with and without amifostine in head and neck squamous cell carcinoma. International Journal of Radiation Oncology* Biology* Physics. 46(5):1105-1108. Bours V, Bonizzi G, Bentires-Alj M, Bureau F, Piette J, Lekeux P, Merville M-P. 2000. NF-κB activation in response to toxical and therapeutical agents: role in inflammation and cancer treatment. Toxicology. 153(1):27-38. Brizel DM, Wasserman TH, Henke M, Strnad V, Rudat V, Monnier A, Eschwege F, Zhang J, Russell L, Oster W. 2000. Phase III randomized trial of amifostine as a radioprotector in head and neck cancer. Journal of Clinical Oncology. 18(19):3339-3345. Brown SL, Kolozsvary A, Liu J, Jenrow KA, Ryu S, Kim JH. 2010. Antioxidant diet supplementation starting 24 hours after exposure reduces radiation lethality. Radiation research. 173(4):462-468. Burdelya LG, Gleiberman AS, Toshkov I, Aygun-Sunar S, Bapardekar M, Manderscheid-Kern P, Bellnier D, Krivokrysenko VI, Feinstein E, Gudkov AV. 2012. Toll-like receptor 5 agonist protects mice from dermatitis and oral mucositis caused by local radiation: implications for head-and-neck cancer radiotherapy. International Journal of Radiation Oncology* Biology* Physics. 83(1):228-234. Calo V, Migliavacca M, Bazan V, Macaluso M, Buscemi M, Gebbia N, Russo A. 2003. STAT proteins: from normal control of cellular events to tumorigenesis. J Cell Physiol. 197(2):157-168. eng. Calveley VL, Khan MA, Yeung IW, Vandyk J, Hill RP. 2005. Partial volume rat lung irradiation: temporal fluctuations of in-field and out-of-field DNA damage and inflammatory cytokines following irradiation. International journal of radiation biology. 81(12):887-899. Calvo W, Hopewell J, Reinhold H, Yeung T. 1988. Time-and dose-related changes in the white matter of the rat brain after single doses of X rays. The British journal of radiology. 61(731):1043-1052. Chacon JA, Schutsky K, Powell DJ. 2016. The Impact of Chemotherapy, Radiation and Epigenetic Modifiers in Cancer Cell Expression of Immune Inhibitory and Stimulatory Molecules and Anti-Tumor Efficacy. Vaccines. 4(4):43.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Chai J, Shi Y. 2014. Apoptosome and inflammasome: conserved machineries for caspase activation. National Science Review. 1(1):101-118. Chai Y, Calaf G, Zhou H, Ghandhi S, Elliston C, Wen G, Nohmi T, Amundson S, Hei T. 2012. Radiation induced COX-2 expression and mutagenesis at non-targeted lung tissues of gpt delta transgenic mice. British journal of cancer. 108(1):91-98. Chai Y, Calaf GM, Zhou H, Ghandhi SA, Elliston CD, Wen G, Nohmi T, Amundson SA, Hei TK. 2013. Radiation induced COX-2 expression and mutagenesis at non-targeted lung tissues of gpt delta transgenic mice. British journal of cancer. 108(1):91-98. eng. Chai Y, Lam R, Calaf G, Zhou H, Amundson S, Hei T. 2013. Radiation-induced non-targeted response in vivo: role of the TGFβ-TGFBR1-COX-2 signalling pathway. British journal of cancer. 108(5):1106-1112. Chai Y, Lam RK, Calaf GM, Zhou H, Amundson S, Hei TK. 2013. Radiation-induced non-targeted response in vivo: role of the TGFbeta-TGFBR1-COX-2 signalling pathway. British journal of cancer. 108(5):11061112. eng. Chamberlain MC, Eaton KD, Fink J. 2011. Radiation-induced myelopathy: treatment with bevacizumab. Archives of neurology. 68(12):1608-1609. eng. Chen M-F, Chen W-C, Lai C-H, Hung C-h, Liu K-C, Cheng Y-H. 2010. Predictive factors of radiation-induced skin toxicity in breast cancer patients. BMC cancer. 10(1):508. Chevillard S, Radicella JP, Levalois C, Lebeau J, Poupon M-F, Oudard S, Dutrillaux B, Boiteux S. 1998. Mutations in OGG1, a gene involved in the repair of oxidative DNA damage, are found in human lung and kidney tumours. Oncogene. 16(23):3083-3086. Chien Y-H, Bau D-T, Jan K-Y. 2004. Nitric oxide inhibits DNA-adduct excision in nucleotide excision repair. Free Radical Biology and Medicine. 36(8):1011-1017. Cho YJ, Yi CO, Jeon BT, Jeong YY, Kang GM, Lee JE, Roh GS, Lee JD. 2013. Curcumin Attenuates RadiationInduced Inflammation and Fibrosis in Rat Lungs. The Korean Journal of Physiology & Pharmacology : Official Journal of the Korean Physiological Society and the Korean Society of Pharmacology. 17(4):267274. Chon YE, Seong J, Kim BK, Cha J, Kim SU, Park JY, Ahn SH, Han K-H, Chon CY, Shin SK. 2011. Gastroduodenal complications after concurrent chemoradiation therapy in patients with hepatocellular carcinoma: endoscopic findings and risk factors. International Journal of Radiation Oncology* Biology* Physics. 81(5):1343-1351. Chung EY, Kim SJ, Ma XJ. 2006. Regulation of cytokine production during phagocytosis of apoptotic cells. Cell research. 16(2):154-161. Chung SI, Horton JA, Ramalingam TR, White AO, Chung EJ, Hudak KE, Scroggins BT, Arron JR, Wynn TA, Citrin DE. 2016. IL-13 is a therapeutic target in radiation lung injury. Scientific Reports. 6. eng. Chung SI, Horton JA, Ramalingam TR, White AO, Chung EJ, Hudak KE, Scroggins BT, Arron JR, Wynn TA, Citrin DE. 2016. IL-13 is a therapeutic target in radiation lung injury [Article]. Scientific Reports. 6:39714. Ciampi R, Knauf JA, Kerler R, Gandhi M, Zhu Z, Nikiforova MN, Rabes HM, Fagin JA, Nikiforov YE. 2005. Oncogenic AKAP9-BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J Clin Invest. 115(1):94-101. eng. Cohen EP, Fish BL, Moulder JE. 2010. Mitigation of radiation injuries via suppression of the reninangiotensin system: emphasis on radiation nephropathy. Current drug targets. 11(11):1423-1429. eng. Crowder CD, Grabowski C, Inampudi S, Sielaff T, Sherman CA, Batts KP. 2009. Selective internal radiation therapy-induced extrahepatic injury: an emerging cause of iatrogenic organ damage. The American journal of surgical pathology. 33(7):963-975. Das PM, Singal R. 2004. DNA methylation and cancer. Journal of Clinical Oncology. 22(22):4632-4642. Datta K, Suman S, Kallakury BV, Fornace Jr AJ. 2012. Exposure to heavy ion radiation induces persistent oxidative stress in mouse intestine.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Dedon PC, Tannenbaum SR. 2004. Reactive nitrogen species in the chemical biology of inflammation. Archives of Biochemistry and Biophysics. 423(1):12-22. Ding NH, Li JJ, Sun LQ. 2013. Molecular Mechanisms and Treatment of Radiation-Induced Lung Fibrosis. Current drug targets. 14(11):1347-1356. eng. Do NL, Nagle D, Poylin VY. 2011. Radiation Proctitis: Current Strategies in Management. Gastroenterology Research and Practice. 2011. Donnelly JP, Bellm LA, Epstein JB, Sonis ST, Symonds RP. 2003. Antimicrobial therapy to prevent or treat oral mucositis. The Lancet Infectious Diseases. 3(7):405-412. Elhammali A, Patel M, Weinberg B, Verma V, Liu J, Olsen JR, Gay HA. 2015. Late gastrointestinal tissue effects after hypofractionated radiation therapy of the pancreas. Radiation Oncology. 10(1):186. Erbil Y, Öztezcan S, Olgaç V, Barbaros U, Deveci U, Kirgiz B, Uysal M, Toker GA. 2006. The effect of heme oxygenase-1 induction by glutamine on radiation-induced intestinal damage: the effect of heme oxygenase-1 on radiation enteritis. The American journal of surgery. 191(4):503-509. Ermolaeva M, Schumacher B. 2013. The innate immune system as mediator of systemic DNA damage responses. Communicative & Integrative Biology. 6(6). eng. Erol FS, Topsakal C, Ozveren MF, Kaplan M, Ilhan N, Ozercan IH, Yildiz OG. 2004. Protective effects of melatonin and vitamin E in brain damage due to gamma radiation: an experimental study. Neurosurgical review. 27(1):65-69. eng. Ertekin MV, Tekin SB, Erdoğan F, Karslioğlu I, Gepdiremen A, Sezen O, Balci E, Gündoğdu C. 2004. The effect of zinc sulphate in the prevention of radiation-induced dermatitis. Journal of radiation research. 45(4):543-548. Fajardo LF. 2005. The pathology of ionizing radiation as defined by morphologic patterns. Acta oncologica (Stockholm, Sweden). 44(1):13-22. eng. Fajardo LF, Berthrong M. 1988. Vascular lesions following radiation. Pathology annual. 23 Pt 1:297-330. eng. Fardid R, Najafi M, Salajegheh A, Kazemi E, Rezaeyan A. 2017. Radiation-induced non-targeted effect in vivo: Evaluation of cyclooygenase-2 and endothelin-1 gene expression in rat heart tissues. Journal of cancer research and therapeutics. 13(1):51-55. eng. Fardid R SA, Mosleh-Shirazi MA, Sharifzadeh S, Okhovat MA, Najafi M, Rezaeyan A, Abaszadeh A. 2017. Melatonin ameliorates the production of COX-2, iNOS, and the formation of 8-OHdG in non-targeted lung tissue after pelvic irradiation. Cell J 19(2):324-331. Flanders KC, Ho BM, Arany PR, Stuelten C, Mamura M, Paterniti MO, Sowers A, Mitchell JB, Roberts AB. 2008. Absence of Smad3 induces neutrophil migration after cutaneous irradiation: possible contribution to subsequent radioprotection. The American journal of pathology. 173(1):68-76. Flanders KC, Major CD, Arabshahi A, Aburime EE, Okada MH, Fujii M, Blalock TD, Schultz GS, Sowers A, Anzano MA. 2003. Interference with transforming growth factor-β/Smad3 signaling results in accelerated healing of wounds in previously irradiated skin. The American journal of pathology. 163(6):2247-2257. Flanders KC, Sullivan CD, Fujii M, Sowers A, Anzano MA, Arabshahi A, Major C, Deng C, Russo A, Mitchell JB. 2002. Mice lacking Smad3 are protected against cutaneous injury induced by ionizing radiation. The American journal of pathology. 160(3):1057-1068. Fliss H, Menard M. 1994. Rapid neutrophil accumulation and protein oxidation in irradiated rat lungs. Journal of applied physiology (Bethesda, Md : 1985). 77(6):2727-2733. eng. Flour M. 2009. The pathophysiology of vulnerable skin. World Wide Wounds. 11. François A, Milliat F, Guipaud O, Benderitter M. 2013. Inflammation and immunity in radiation damage to the gut mucosa. BioMed research international. 2013. Frey N, Katus HA, Olson EN, Hill JA. 2004. Hypertrophy of the heart a new therapeutic target? Circulation. 109(13):1580-1589.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Gangoiti P, Granado MH, Arana L, Ouro A, Gómez-Muñoz A. 2008. Involvement of nitric oxide in the promotion of cell survival by ceramide 1-phosphate. FEBS letters. 582(15):2263-2269. Gao L, Li FS, Chen XH, Liu QW, Feng JB, Liu QJ, Su X. 2014. Radiation induces phosphorylation of STAT3 in a dose- and time-dependent manner. Asian Pacific journal of cancer prevention : APJCP. 15(15):61616164. eng. Garg UC, Hassid A. 1989. Nitric oxide-generating vasodilators and 8-bromo-cyclic guanosine monophosphate inhibit mitogenesis and proliferation of cultured rat vascular smooth muscle cells. Journal of Clinical Investigation. 83(5):1774. Gaugler M-H, Vereycken-Holler V, Squiban C, Vandamme M, Vozenin-Brotons M-C, Benderitter M. 2005. Pravastatin limits endothelial activation after irradiation and decreases the resulting inflammatory and thrombotic responses. Radiation research. 163(5):479-487. Gaugler M. 2014. A unifying system: does the vascular endothelium have a role to play in multi-organ failure following radiation exposure? The British journal of radiology. Gauter-Fleckenstein B, Fleckenstein K, Owzar K, Jiang C, Rebouças JS, Batinic-Haberle I, Vujaskovic Z. 2010. Early and late administration of MnTE-2-PyP 5+ in mitigation and treatment of radiation-induced lung damage. Free Radical Biology and Medicine. 48(8):1034-1043. Gehrke N, Mertens C, Zillinger T, Wenzel J, Bald T, Zahn S, Tüting T, Hartmann G, Barchet W. 2013. Oxidative Damage of DNA Confers Resistance to Cytosolic Nuclease TREX1 Degradation and Potentiates STING-Dependent Immune Sensing. Immunity. 39(3):482-495. Ghafoori P, Marks LB, Vujaskovic Z, Kelsey C. 2008. Radiation-Induced Lung Injury-Assessment, Management, and Prevention. Oncology; San Francisco. 22(1):37. Ghobadi A, Shirazi A, Najafi M, Kahkesh MH, Rezapoor S. 2017. Melatonin ameliorates radiation-induced oxidative stress at targeted and nontargeted lung tissue. Journal of medical physics. 42(4):241. Giovagnoli AR, Boiardi A. 1994. Cognitive impairment and quality of life in long-term survivors of malignant brain tumors. Italian journal of neurological sciences. 15(9):481-488. eng. Greene-Schloesser D, Robbins ME. 2012. Radiation-induced cognitive impairment-from bench to bedside. Neuro-oncology. 14(suppl 4):iv37-iv44. Grover N, Johnson A. 1997. Case report: Aminocaproic acid used to control upper gastrointestinal bleeding in radiation gastritis. Digestive diseases and sciences. 42(5):982-984. Groves AM, Johnston CJ, Misra RS, Williams JP, Finkelstein JN. 2016. Effects of IL-4 on pulmonary fibrosis and the accumulation and phenotype of macrophage subpopulations following thoracic irradiation. International journal of radiation biology. 92(12):754-765. Gudkov AV, Gurova KV, Komarova EA. 2011. Inflammation and p53: A Tale of Two Stresses. Genes & Cancer. 2(4):503-516. eng. Gudkov AV, Komarova EA. 2016. p53 and the Carcinogenicity of Chronic Inflammation. Cold Spring Harbor perspectives in medicine. 6(11). eng. Gurova KV, Hill JE, Guo C, Prokvolit A, Burdelya LG, Samoylova E, Khodyakova AV, Ganapathi R, Ganapathi M, Tararova ND et al. 2005. Small molecules that reactivate p53 in renal cell carcinoma reveal a NF-kappaB-dependent mechanism of p53 suppression in tumors. Proc Natl Acad Sci U S A. 102(48):17448-17453. eng. Gurses I, Ozeren M, Serin M, Yucel N, Erkal HS. 2014. Histopathological evaluation of melatonin as a protective agent in heart injury induced by radiation in a rat model. Pathology, research and practice. 210(12):863-871. eng. Ha CT, Li X-H, Fu D, Moroni M, Fisher C, Arnott R, Srinivasan V, Xiao M. 2014. Circulating interleukin-18 as a biomarker of total-body radiation exposure in mice, minipigs, and nonhuman primates (NHP). Haddadi G, Shirazi A, Sepehrizadeh Z, Mahdavi S, Haddadi M. 2013. Radioprotective effect of melatonin on the cervical spinal cord in irradiated rats. Cell J. 2013; 14 (4): 246-253. cell j. 50:1.8-2.0.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Haddadi GH RA, Mosleh-Shirazi MA, Hosseinzadeh M, Fardid R, Najafi M, et al. 2017. Hesperidin as radioprotector against radiation-induced lung damage in rat: A histopathological study. Journal of medical physics. 42:25-32. Hallahan D, Kuchibhotla J, Wyble C. 1996. Cell adhesion molecules mediate radiation-induced leukocyte adhesion to the vascular endothelium. Cancer research. 56(22):5150-5155. Hallahan DE, Virudachalam S. 1997. Intercellular adhesion molecule 1 knockout abrogates radiation induced pulmonary inflammation. Proceedings of the National Academy of Sciences of the United States of America. 94(12):6432-6437. Hallahan DE, Virudachalam S. 1999. Accumulation of P-selectin in the lumen of irradiated blood vessels. Radiation research. 152(1):6-13. eng. Halle M, Ekström M, Farnebo F, Tornvall P. 2010. Endothelial activation with prothrombotic response in irradiated microvascular recipient veins. Journal of plastic, reconstructive & aesthetic surgery. 63(11):1910-1916. Halle M, Gabrielsen A, Paulsson-Berne G, Gahm C, Agardh HE, Farnebo F, Tornvall P. 2010. Sustained Inflammation Due to Nuclear Factor-Kappa B Activation in Irradiated Human Arteries. Journal of the American College of Cardiology. 55(12):1227-1236. Halperin EC, Gaspar L, George S, Darr D, Pinnell S. 1993. A double-blind, randomized, prospective trial to evaluate topical vitamin C solution for the prevention of radiation dermatitis. International Journal of Radiation Oncology* Biology* Physics. 26(3):413-416. Han G, Zhang H, Xie CH, Zhou YF. 2011. Th2-like immune response in radiation-induced lung fibrosis. Oncology reports. 26(2):383-388. eng. Haneklaus M, O’Neill LA, Coll RC. 2013. Modulatory mechanisms controlling the NLRP3 inflammasome in inflammation: recent developments. Current opinion in immunology. 25(1):40-45. Hansson GK. 2005. Inflammation, atherosclerosis, and coronary artery disease. New England Journal of Medicine. 352(16):1685-1695. Hayashi T, Morishita Y, Kubo Y, Kusunoki Y, Hayashi I, Kasagi F, Hakoda M, Kyoizumi S, Nakachi K. 2005. Long-term effects of radiation dose on inflammatory markers in atomic bomb survivors. The American journal of medicine. 118(1):83-86. Haydont V, Mathé D, Bourgier C, Abdelali J, Aigueperse J, Bourhis J, Vozenin-Brotons M-C. 2005. Induction of CTGF by TGF-β1 in normal and radiation enteritis human smooth muscle cells: Smad/Rho balance and therapeutic perspectives. Radiotherapy and oncology. 76(2):219-225. Heckmann M, Douwes K, Peter R, Degitz K. 1998. Vascular Activation of Adhesion Molecule mRNA and Cell Surface Expression by Ionizing Radiation. Experimental Cell Research. 238(1):148-154. Hei TK, Zhao Y, Zhou H, Ivanov V. 2011. Mechanism of Radiation Carcinogenesis: Role of theTGFBI Gene and the Inflammatory Signaling Cascade. Advances in experimental medicine and biology. 720:163-170. eng. Hei TK, Zhou H, Ivanov VN, Hong M, Lieberman HB, Brenner DJ, Amundson SA, Geard CR. 2008. Mechanism of radiation‐induced bystander effects: a unifying model. Journal of Pharmacy and Pharmacology. 60(8):943-950. Hellweg CE. 2015. The nuclear factor κB pathway: a link to the immune system in the radiation response. Cancer letters. Hill A, Hanson M, Bogle MA, Duvic M. 2004. Severe radiation dermatitis is related to Staphylococcus aureus. American journal of clinical oncology. 27(4):361-363. Hillman GG, Singh-Gupta V, Lonardo F, Hoogstra DJ, Abernathy LM, Yunker CK, Rothstein SE, Rakowski J, Sarkar FH, Gadgeel S et al. 2013. Radioprotection of Lung Tissue by Soy Isoflavones. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 8(11):13561364.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Hogquist KA, Nett MA, Unanue ER, Chaplin DD. 1991. Interleukin 1 is processed and released during apoptosis. Proceedings of the National Academy of Sciences. 88(19):8485-8489. Holler V, Buard V, Gaugler M-H, Guipaud O, Baudelin C, Sache A, del R Perez M, Squiban C, Tamarat R, Milliat F. 2009. Pravastatin limits radiation-induced vascular dysfunction in the skin. Journal of Investigative Dermatology. 129(5):1280-1291. Hoving S, Heeneman S, Gijbels MJJ, te Poele JAM, Russell NS, Daemen MJAP, Stewart FA. 2008. SingleDose and Fractionated Irradiation Promote Initiation and Progression of Atherosclerosis and Induce an Inflammatory Plaque Phenotype in ApoE-/- Mice. International Journal of Radiation Oncology Biology Physics. 71(3):848-857. Hu ZP, Fang XL, Fang N, Wang XB, Qian HY, Cao Z, Cheng Y, Wang BN, Wang Y. 2013. Melatonin ameliorates vascular endothelial dysfunction, inflammation, and atherosclerosis by suppressing the TLR4/NF‐κB system in high‐fat‐fed rabbits. Journal of pineal research. 55(4):388-398. Hudson JD, Shoaibi MA, Maestro R, Carnero A, Hannon GJ, Beach DH. 1999. A proinflammatory cytokine inhibits p53 tumor suppressor activity. J Exp Med. 190(10):1375-1382. eng. Hussain SP, Hofseth LJ, Harris CC. 2003. Radical causes of cancer. Nat Rev Cancer. 3(4):276-285. eng. Hymes SR, Strom EA, Fife C. 2006. Radiation dermatitis: clinical presentation, pathophysiology, and treatment 2006. J Am Acad Dermatol. 54(1):28-46. Indaram AV, Visvalingam V, Locke M, Bank S. 2000. Mucosal cytokine production in radiation-induced proctosigmoiditis compared with inflammatory bowel disease. The American journal of gastroenterology. 95(5):1221-1225. Ito Y, Kinoshita M, Yamamoto T, Sato T, Obara T, Saitoh D, Seki S, Takahashi Y. 2013. A Combination of Pre-and Post-Exposure Ascorbic Acid Rescues Mice from Radiation-Induced Lethal Gastrointestinal Damage. International journal of molecular sciences. 14(10):19618-19635. Jaiswal M, LaRusso NF, Burgart LJ, Gores GJ. 2000. Inflammatory cytokines induce DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric oxide-dependent mechanism. Cancer research. 60(1):184-190. Jaiswal M, LaRusso NF, Gores GJ. 2001. Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. American Journal of Physiology-Gastrointestinal and Liver Physiology. 281(3):G626-G634. Jaiswal M, LaRusso NF, Nishioka N, Nakabeppu Y, Gores GJ. 2001. Human Ogg1, a protein involved in the repair of 8-oxoguanine, is inhibited by nitric oxide. Cancer research. 61(17):6388-6393. Jaiswal M, LaRusso NF, Shapiro RA, Billiar TR, Gores GJ. 2001. Nitric oxide–mediated inhibition of DNA repair potentiates oxidative DNA damage in cholangiocytes. Gastroenterology. 120(1):190-199. Jakubzick C, Kunkel SL, Puri RK, Hogaboam CM. 2004. Therapeutic targeting of IL-4- and IL-13-responsive cells in pulmonary fibrosis. Immunologic research. 30(3):339-349. eng. Janko M, Ontiveros F, Fitzgerald TJ, Deng A, DeCicco M, Rock KL. 2012. IL-1 generated subsequent to radiation-induced tissue injury contributes to the pathogenesis of radiodermatitis. Radiat Res. 178(3):166-172. Jeremy JY, Rowe D, Emsley AM, Newby AC. 1999. Nitric oxide and the proliferation of vascular smooth muscle cells. Cardiovascular research. 43(3):580-594. Jia D, Koonce NA, Griffin RJ, Jackson C, Corry PM. 2010. Prevention and mitigation of acute death of mice after abdominal irradiation by the antioxidant N-acetyl-cysteine (NAC). Radiat Res. 173(5):579-589. Jiang Y, Chen X, Tian W, Yin X, Wang J, Yang H. 2014. The role of TGF-β1–miR-21–ROS pathway in bystander responses induced by irradiated non-small-cell lung cancer cells. British journal of cancer. 111(4):772-780. Kalinich JF, Catravas GN, Snyder SL. 1989. The effect of γ radiation on DNA methylation. Radiation research. 117(2):185-197.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Kannan V, Bapsy PP, Anantha N, Doval DC, Vaithianathan H, Banumathy G, Reddy KB, Kumaraswamy SV, Shenoy AM. 1997. Efficacy and safety of granulocyte macrophage-colony stimulating factor (GM-CSF) on the frequency and severity of radiation mucositis in patients with head and neck carcinoma. International Journal of Radiation Oncology* Biology* Physics. 37(5):1005-1010. Karki R, Igwe OJ. 2013. Toll-like receptor 4-mediated nuclear factor kappa B activation is essential for sensing exogenous oxidants to propagate and maintain oxidative/nitrosative cellular stress. PloS one. 8(9). Kaur P, Asea A. 2012. Radiation-induced effects and the immune system in cancer. Frontiers in oncology. 2:191. Keefe DM. 2007. Intestinal mucositis: mechanisms and management. Current opinion in oncology. 19(4):323-327. Kennedy M, Bruninga K, Mutlu EA, Losurdo J, Choudhary S, Keshavarzian A. 2001. Successful and sustained treatment of chronic radiation proctitis with antioxidant vitamins E and C. The American journal of gastroenterology. 96(4):1080-1084. Keskek M, Gocmen E, Kilic M, Gencturk S, Can B, Cengiz M, Okten RM, Koc M. 2006. Increased expression of cyclooxygenase-2 (COX-2) in radiation-induced small bowel injury in rats. J Surg Res. 135(1):76-84. Khan R, Sheppard R. 2006. Fibrosis in heart disease: understanding the role of transforming growth factor-β(1) in cardiomyopathy, valvular disease and arrhythmia. Immunology. 118(1):10-24. Kidane D, Chae WJ, Czochor J, Eckert KA, Glazer PM, Bothwell ALM, Sweasy JB. 2014. Interplay between DNA repair and inflammation, and the link to cancer. Critical reviews in biochemistry and molecular biology. 49(2):116-139. eng. Klune JR, Dhupar R, Cardinal J, Billiar TR, Tsung A. 2008. HMGB1: Endogenous Danger Signaling. Molecular Medicine. 14(7-8):476-484. Konda A, Savin MA, Cappell MS, Duffy MC. 2009. Radiation microsphere–induced GI ulcers after selective internal radiation therapy for hepatic tumors: an underrecognized clinical entity. Gastrointestinal endoscopy. 70(3):561-567. Kong FM, Hayman JA, Griffith KA, Kalemkerian GP, Arenberg D, Lyons S, Turrisi A, Lichter A, Fraass B, Eisbruch A et al. 2006. Final toxicity results of a radiation-dose escalation study in patients with nonsmall-cell lung cancer (NSCLC): predictors for radiation pneumonitis and fibrosis. International journal of radiation oncology, biology, physics. 65(4):1075-1086. eng. Köstler WJ, Hejna M, Wenzel C, Zielinski CC. 2001. Oral mucositis complicating chemotherapy and/or radiotherapy: options for prevention and treatment. CA: a cancer journal for clinicians. 51(5):290-315. Koturbash I, Loree J, Kutanzi K, Koganow C, Pogribny I, Kovalchuk O. 2008. In vivo bystander effect: cranial X-irradiation leads to elevated DNA damage, altered cellular proliferation and apoptosis, and increased p53 levels in shielded spleen. International Journal of Radiation Oncology* Biology* Physics. 70(2):554-562. Kouvaris J, KOULOULIAS V, KOKAKIS J, MATSOPOULOS G, MYRSINI B, VLAHOS L. 2002. The cytoprotective effect of amifostine in acute radiation dermatitis: a retrospective analysis. European Journal of Dermatology. 12(5):458-462. Kouvaris JR, Kouloulias VE, Plataniotis GA, Balafouta EJ, Vlahos LJ. 2001. Dermatitis during radiation for vulvar carcinoma: prevention and treatment with granulocyte‐macrophage colony‐stimulating factor impregnated gauze. Wound Repair and Regeneration. 9(3):187-193. Kovalchuk O, Baulch JE. 2008. Epigenetic changes and nontargeted radiation effects--is there a link? Environmental and molecular mutagenesis. 49(1):16-25. eng. Krstić J, Trivanović D, Mojsilović S, Santibanez JF. 2015. Transforming growth factor-beta and oxidative stress interplay: implications in tumorigenesis and cancer progression. Oxidative medicine and cellular longevity. 2015.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. 2012. Immunogenic cell death and DAMPs in cancer therapy. Nature Reviews Cancer. 12(12):860-875. Kubes P, McCafferty D-M. 2000. Nitric oxide and intestinal inflammation. The American journal of medicine. 109(2):150-158. Kusunoki Y, Hayashi T. 2008. Long-lasting alterations of the immune system by ionizing radiation exposure: implications for disease development among atomic bomb survivors. International journal of radiation biology. 84(1):1-14. Kyrkanides S, Moore AH, Olschowka JA, Daeschner JC, Williams JP, Hansen JT, O’Banion MK. 2002. Cyclooxygenase-2 modulates brain inflammation-related gene expression in central nervous system radiation injury. Molecular Brain Research. 104(2):159-169. Lau J, Ilkhanizadeh S, Wang S, Miroshnikova YA, Salvatierra NA, Wong RA, Schmidt C, Weaver VM, Weiss WA, Persson AI. 2015. STAT3 Blockade Inhibits Radiation-Induced Malignant Progression in Glioma. Cancer research. 75(20):4302-4311. eng. Lee JC, Kinniry PA, Arguiri E, Serota M, Kanterakis S, Chatterjee S, Solomides CC, Javvadi P, Koumenis C, Cengel KA et al. 2010. Dietary curcumin increases antioxidant defenses in lung, ameliorates radiationinduced pulmonary fibrosis, and improves survival in mice. Radiation research. 173(5):590-601. eng. Lee JY, Kim HS, Choi HY, Oh TH, Ju BG, Yune TY. 2012. Valproic acid attenuates blood-spinal cord barrier disruption by inhibiting matrix metalloprotease-9 activity and improves functional recovery after spinal cord injury. Journal of neurochemistry. 121(5):818-829. eng. Li GQ, Xia HH-X, Chen MH, Gu Q, De Wang J, Peng JZ, Chan AO, Cho CH, So HL, Lam SK. 2006. Effects of Cyclooxygenase-1 and-2 Gene Disruption on Helicobacter pylori–Induced Gastric Inflammation. Journal of Infectious Diseases. 193(7):1037-1046. Li YQ, Ballinger JR, Nordal RA, Su ZF, Wong CS. 2001. Hypoxia in radiation-induced blood-spinal cord barrier breakdown. Cancer research. 61(8):3348-3354. eng. Liang L, Hu D, Liu W, Williams JP, Okunieff P, Ding I. 2003. Celecoxib reduces skin damage after radiation: selective reduction of chemokine and receptor mRNA expression in irradiated skin but not in irradiated mammary tumor. American journal of clinical oncology. 26(4):S114-S121. Lin R, Xiao D, Guo Y, Tian D, Yun H, Chen D, Su M. 2015. Chronic inflammation-related DNA damage response: a driving force of gastric cardia carcinogenesis. Oncotarget. 6(5):2856. Lin R, Xiao D, Guo Y, Tian D, Yun H, Chen D, Su M. 2015. Chronic inflammation-related DNA damage response: a driving force of gastric cardia carcinogenesis. Oncotarget. 6(5):2856-2864. eng. Lips IM, Dehnad H, van Gils CH, Boeken Kruger AE, van der Heide UA, van Vulpen M. 2008. High-dose intensity-modulated radiotherapy for prostate cancer using daily fiducial marker-based position verification: acute and late toxicity in 331 patients. Radiation oncology (London, England). 3:15-15. Liu R-M, Pravia KG. 2010. Oxidative stress and glutathione in TGF-β-mediated fibrogenesis. Free Radical Biology and Medicine. 48(1):1-15. Liu S, Jin S, Liu X-D. 2004. Radiation-induced bystander effect in immune response. Biomedical and Environmental Sciences. 17(1):40-46. Liu S, Sammons V, Fairhall J, Reddy R, Tu J, Hong Duong TT, Stoodley M. 2012. Molecular responses of brain endothelial cells to radiation in a mouse model. Journal of Clinical Neuroscience. 19(8):1154-1158. Liu T, Liu Y, He S, Zhang Z, Kligerman M. 1992. Use of radiation with or without WR‐2721 in advanced rectal cancer. Cancer. 69(11):2820-2825. Liu W, Ding I, Chen K, Olschowka J, Xu J, Hu D, Morrow GR, Okunieff P. 2006. Interleukin 1beta (IL1B) signaling is a critical component of radiation-induced skin fibrosis. Radiation research. 165(2):181-191. eng. Logan RM, Stringer AM, Bowen JM, Yeoh AS-J, Gibson RJ, Sonis ST, Keefe DM. 2007. The role of proinflammatory cytokines in cancer treatment-induced alimentary tract mucositis: pathobiology, animal models and cytotoxic drugs. Cancer treatment reviews. 33(5):448-460.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Lomax M, Folkes L, O'Neill P. 2013. Biological consequences of radiation-induced DNA damage: relevance to radiotherapy. Clinical Oncology. 25(10):578-585. Lyudmila G. Burdelya VIK, Thomas C. Tallant, Evguenia Strom, Anatoly S. Gleiberman, Damodar Gupta, et al. . 2008. An Agonist of Toll-Like Receptor 5 Has Radioprotective Activity in Mouse and Primate Models. science. 320(5873):226–230. M. Najafi AS, E. Motevaseli, Gh. Geraily, F. Norouzi, M. Heidari , S. Rezapoor. 2017. The melatonin immunomodulatory actions in radiotherapy. Biophys Rev. 9(2):139–148. MacNaughton W. 1998. Expression, activity and cellular localization of inducible nitric oxide synthase in rat ileum and colon post-irradiation. International journal of radiation biology. 74(2):255-264. Mahmood J, Jelveh S, Calveley V, Zaidi A, Doctrow SR, Hill RP. 2011. Mitigation of Lung Injury after Accidental Exposure to Radiation. Radiat Res. 176(6):770-780. Mahmood J, Jelveh S, Zaidi A, Doctrow SR, Medhora M, Hill RP. 2014. Targeting the Renin-angiotensin system combined with an antioxidant is highly effective in mitigating radiation-induced lung damage. International journal of radiation oncology, biology, physics. 89(4):722-728. eng. Mancini ML, Sonis ST. 2014. Mechanisms of cellular fibrosis associated with cancer regimen-related toxicities. Frontiers in pharmacology. 5:51. Martin M, Lefaix J-L, Delanian S. 2000. TGF-β1 and radiation fibrosis: a master switch and a specific therapeutic target? International Journal of Radiation Oncology* Biology* Physics. 47(2):277-290. Martinon F. 2010. Signaling by ROS drives inflammasome activation. European journal of immunology. 40(3):616-619. Masoud Najafi, Mohsen Cheki, Saeed Rezapoor, Ghazale Geraily, Elahe Motevaseli, Carla Carnovale, Emilio Clementi, Alireza Shirazi. 2018. Metformin: Prevention of genomic instability and cancer: A review. Mutat Res Gen Tox En 827, 1–8. https://doi.org/10.1016/j.mrgentox.2018.01.007 Medhora M, Gao F, Jacobs ER, Moulder JE. 2012. Radiation damage to the lung: mitigation by angiotensin converting enzyme (ACE) inhibitors. Respirology (Carlton, Vic). 17(1):66-71. Meira LB, Bugni JM, Green SL, Lee C-W, Pang B, Borenshtein D, Rickman BH, Rogers AB, Moroski-Erkul CA, McFaline JL. 2008. DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. The Journal of clinical investigation. 118(7):2516-2525. Meira LB, Bugni JM, Green SL, Lee C-W, Pang B, Borenshtein D, Rickman BH, Rogers AB, Moroski-Erkul CA, McFaline JL et al. 2008. DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. The Journal of Clinical Investigation. 118(7):2516-2525. Meirovitz A, Kuten M, Billan S, Abdah-Bortnyak R, Sharon A, Peretz T, Sela M, Schaffer M, Barak V. 2010. Cytokines levels, severity of acute mucositis and the need of PEG tube installation during chemoradiation for head and neck cancer–a prospective pilot study. Radiation oncology (London, England). 5:16. Mendonca MS, Farrington DL, Mayhugh BM, Qin Y, Temples T, Comerford K, Chakrabarti R, Zainabadi K, Redpath JL, Stanbridge EJ et al. 2004. Homozygous deletions within the 11q13 cervical cancer tumorsuppressor locus in radiation-induced, neoplastically transformed human hybrid cells. Genes, chromosomes & cancer. 39(4):277-287. eng. Mendonca MS, Fasching CL, Srivatsan ES, Stanbridge EJ, Redpath JL. 1995. Loss of a putative tumor suppressor locus after gamma-ray-induced neoplastic transformation of HeLa x skin fibroblast human cell hybrids. Radiation research. 143(1):34-44. eng. Mendonca MS, Howard K, Fasching CL, Farrington DL, Desmond LA, Stanbridge EJ, Redpath JL. 1998. Loss of suppressor loci on chromosomes 11 and 14 may be required for radiation-induced neoplastic transformation of HeLa x skin fibroblast human cell hybrids. Radiation research. 149(3):246-255. eng. Meyers CA, Brown PD. 2006. Role and relevance of neurocognitive assessment in clinical trials of patients with CNS tumors. Journal of clinical oncology. 24(8):1305-1309.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Michurina T, Krasnov P, Balazs A, Nakaya N, Vasilieva T, Kuzin B, Khrushchov N, Mulligan RC, Enikolopov G. 2004. Nitric oxide is a regulator of hematopoietic stem cell activity. Molecular Therapy. 10(2):241248. Mikhailenko V, Diomina E, Muzalov I, Gerashchenko B. 2013. Nitric oxide coordinates development of genomic instability in realization of combined effect with ionizing radiation. Experimental oncology. 35(1):58-64. Mikhailenko V, Muzalov I. 2013. Exogenous nitric oxide potentiate DNA damage and alter DNA repair in cells exposed to ionising radiation. Experimental oncology.(35,№ 4):318-324. Milliat F, François A, Isoir M, Deutsch E, Tamarat R, Tarlet G, Atfi A, Validire P, Bourhis J, Sabourin J-C et al. 2006. Influence of Endothelial Cells on Vascular Smooth Muscle Cells Phenotype after Irradiation : Implication in Radiation-Induced Vascular Damages. The American journal of pathology. 169(4):14841495. Minshall EM, Leung DY, Martin RJ, Song YL, Cameron L, Ernst P, Hamid Q. 1997. Eosinophil-associated TGF-β1 mRNA expression and airways fibrosis in bronchial asthma. American journal of respiratory cell and molecular biology. 17(3):326-333. Mishra A, Chaudhary A, Sethi S. 2004. Oxidized omega-3 fatty acids inhibit NF-κB activation via a PPARαdependent pathway. Arteriosclerosis, thrombosis, and vascular biology. 24(9):1621-1627. Mohsen Cheki RY, Bagher Farhood, Abolhassan Rezaeyan, Dheyauldeen shabeeb, Peyman Amini, Saeed Rezapoor, Masoud Najafi. 2018. COX-2 in Radiotherapy; a potential target for radioprotection and radiosensitization Current molecular pharmacology. 11. Mollà M, Gironella M, Miquel R, Tovar V, Engel P, Biete A, Piqué JM, Panés J. 2003. Relative roles of ICAM-1 and VCAM-1 in the pathogenesis of experimental radiation-induced intestinal inflammation. International Journal of Radiation Oncology*Biology*Physics. 57(1):264-273. Atherosclerosis is an inflammatory disease. Seminars in immunopathology; 2009: Springer. Montezano AC, Touyz RM. 2012. Reactive oxygen species and endothelial function–role of nitric oxide synthase uncoupling and Nox family nicotinamide adenine dinucleotide phosphate oxidases. Basic & clinical pharmacology & toxicology. 110(1):87-94. Morgan MJ, Liu Z-g. 2011. Crosstalk of reactive oxygen species and NF-κB signaling. Cell research. 21(1):103-115. MOSLEMI NM, MOTEVALIZADEH AA. 2009. NONCODING RNAS AND CANCER. AVICENNA JOURNAL OF MEDICAL BIOTECHNOLOGY (AJMB). Mothersill C, Seymour C. 2012. Are Epigenetic Mechanisms Involved in Radiation-Induced Bystander Effects? Frontiers in genetics. 3. eng. Mul VE, de Jong JM, Murrer LH, van den Ende PL, Houben RM, Lacko M, Lambin P, Baumert BG. 2012. Lhermitte sign and myelopathy after irradiation of the cervical spinal cord in radiotherapy treatment of head and neck cancer. Strahlentherapie und Onkologie : Organ der Deutschen Rontgengesellschaft [et al]. 188(1):71-76. eng. Müller K, Meineke V. 2007. Radiation-induced alterations in cytokine production by skin cells. Exp Hematol. 35(4):96-104. Murakami A, Ohigashi H. 2007. Targeting NOX, INOS and COX‐2 in inflammatory cells: Chemoprevention using food phytochemicals. International journal of cancer. 121(11):2357-2363. Najafi M, Fardid R, Hadadi G, Fardid M. 2014. The Mechanisms of Radiation-Induced Bystander Effect. Journal of Biomedical Physics & Engineering. 4(4):163-172. Najafi M, Fardid R, Takhshid MA. 2016. Radiation-Induced Oxidative Stress at Out-of-Field. Cell J. 18(3):340-345. eng. Najafi M SA, Rezaeyan A. 2017. Bystander effect and second primary cancers following radiotherapy: What are its significances? Journal of medical physics. 42:55-56.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Najafi M, Shirazi A, Motevaseli E, Rezaeyan A, Salajegheh A, Rezapoor S. 2017. Melatonin as an antiinflammatory agent in radiotherapy. Inflammopharmacology.DOI:10.1007/s10787-10017-10332-10785. Nathan C. 1992. Nitric oxide as a secretory product of mammalian cells. The FASEB journal. 6(12):30513064. Naymagon S, Warner RR, Patel K, Harpaz N, Machac J, Weintraub JL, Kim MK. 2010. Gastroduodenal ulceration associated with radioembolization for the treatment of hepatic tumors: an institutional experience and review of the literature. Digestive diseases and sciences. 55(9):2450-2458. Nguyen NP, Antoine JE, Dutta S, Karlsson U, Sallah S. 2002. Current concepts in radiation enteritis and implications for future clinical trials. Cancer. 95(5):1151-1163. Nikitaki Z, Mavragani IV, Laskaratou DA, Gika V, Moskvin VP, Theofilatos K, Vougas K, Stewart RD, Georgakilas AG. 2016. Systemic mechanisms and effects of ionizing radiation: A new 'old' paradigm of how the bystanders and distant can become the players. Seminars in cancer biology. 37-38:77-95. eng. Nishiura H, Iwamoto S, Kido M, Aoki N, Maruoka R, Ikeda A, Chiba T, Watanabe N. 2013. Interleukin‐21 and tumor necrosis factor‐α are critical for the development of autoimmune gastritis in mice. Journal of gastroenterology and hepatology. 28(6):982-991. Nordal RA, Wong CS. 2004. Intercellular adhesion molecule-1 and blood-spinal cord barrier disruption in central nervous system radiation injury. Journal of neuropathology and experimental neurology. 63(5):474-483. eng. Ogawa Y, Kobayashi T, Nishioka A, Kariya S, Hamasato S, Seguchi H, Yoshida S. 2003. Radiation-induced reactive oxygen species formation prior to oxidative DNA damage in human peripheral T cells. International journal of molecular medicine. 11(2):149-152. Ohnishi S, Ma N, Thanan R, Pinlaor S, Hammam O, Murata M, Kawanishi S. 2013. DNA damage in inflammation-related carcinogenesis and cancer stem cells. Oxidative medicine and cellular longevity. 2013. Okada F. 2014. Inflammation-Related Carcinogenesis: Current Findings in Epidemiological Trends. Yonago Acta Medica. 57(2):65-72. eng. Okunieff P, Cornelison T, Mester M, Liu W, Ding I, Chen Y, Zhang H, Williams JP, Finkelstein J. 2005. Mechanism and modification of gastrointestinal soft tissue response to radiation: role of growth factors. International Journal of Radiation Oncology* Biology* Physics. 62(1):273-278. Omed A, Lawrance JA, Murphy G, Laasch H-U, Wilson G, Illidge T, Tipping J, Zivanovic M, Jeans S. 2010. A retrospective analysis of selective internal radiation therapy (SIRT) with yttrium-90 microspheres in patients with unresectable hepatic malignancies. Clinical radiology. 65(9):720-728. Ong ZY, Gibson RJ, Bowen JM, Stringer AM, Darby JM, Logan RM, Yeoh AS, Keefe DM. 2010. Proinflammatory cytokines play a key role in the development of radiotherapy-induced gastrointestinal mucositis. Radiation oncology (London, England). 5(1):22-29. Ortiz F, Acuña‐Castroviejo D, Doerrier C, Dayoub JC, López LC, Venegas C, García JA, López A, Volt H, Luna‐Sánchez M. 2015. Melatonin blunts the mitochondrial/NLRP3 connection and protects against radiation‐induced oral mucositis. Journal of pineal research. 58(1):34-49. Park B, Yee C, Lee KM. 2014. The Effect of Radiation on the Immune Response to Cancers. International journal of molecular sciences. 15(1):927-943. eng. Patel P, Subhas G, Gupta A, Chang Y-J, Mittal VK, McKendrick A. 2009. Oral vitamin A enhances the effectiveness of formalin 8% in treating chronic hemorrhagic radiation proctopathy. Diseases of the Colon & Rectum. 52(9):1605-1609. Pateras IS, Havaki S, Nikitopoulou X, Vougas K, Townsend PA, Panayiotidis MI, Georgakilas AG, Gorgoulis VG. 2015. The DNA damage response and immune signaling alliance: Is it good or bad? Nature decides when and where. Pharmacology & Therapeutics. 154:36-56. Pazhanisamy SK, Li H, Wang Y, Batinic-Haberle I, Zhou D. 2011. NADPH oxidase inhibition attenuates total body irradiation-induced haematopoietic genomic instability. Mutagenesis. 26(3):431-435. eng.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Pétrilli V, Dostert C, Muruve DA, Tschopp J. 2007. The inflammasome: a danger sensing complex triggering innate immunity. Current opinion in immunology. 19(6):615-622. Piccinini A, Midwood K. 2010. DAMPening inflammation by modulating TLR signalling. Mediators of inflammation. 2010. Piccoli C, D’Aprile A, Ripoli M, Scrima R, Lecce L, Boffoli D, Tabilio A, Capitanio N. 2007. Bone-marrow derived hematopoietic stem/progenitor cells express multiple isoforms of NADPH oxidase and produce constitutively reactive oxygen species. Biochemical and biophysical research communications. 353(4):965-972. Piccoli C, Ria R, Scrima R, Cela O, D'Aprile A, Boffoli D, Falzetti F, Tabilio A, Capitanio N. 2005. Characterization of mitochondrial and extra-mitochondrial oxygen consuming reactions in human hematopoietic stem cells Novel evidence of the occurrence of NAD (P) H oxidase activity. Journal of Biological Chemistry. 280(28):26467-26476. Pohlers D, Brenmoehl J, Löffler I, Müller CK, Leipner C, Schultze-Mosgau S, Stallmach A, Kinne RW, Wolf G. 2009. TGF-β and fibrosis in different organs — molecular pathway imprints. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 1792(8):746-756. Poon IK, Lucas CD, Rossi AG, Ravichandran KS. 2014. Apoptotic cell clearance: basic biology and therapeutic potential. Nature Reviews Immunology. 14(3):166-180. Pos F, Horenblas S, Dom P, Moonen L, Bartelink H. 2005. Organ preservation in invasive bladder cancer: brachytherapy, an alternative to cystectomy and combined modality treatment? International journal of radiation oncology, biology, physics. 61(3):678-686. Eng. Psimaras D, Tafani C, Ducray F, Leclercq D, Feuvret L, Delattre JY, Ricard D. 2016. Bevacizumab in lateonset radiation-induced myelopathy. Neurology. 86(5):454-457. eng. Punjabi C, Laskin J, Hwang S, MacEachern L, Laskin D. 1994. Enhanced production of nitric oxide by bone marrow cells and increased sensitivity to macrophage colony-stimulating factor (CSF) and granulocytemacrophage CSF after benzene treatment of mice. Blood. 83(11):3255-3263. Qi F, Sugihara T, Hattori Y, Yamamoto Y, Kanno M, Abe K. 1998. Functional and morphological damage of endothelium in rabbit ear artery following irradiation with cobalt60. Br J Pharmacol. 123(4):653-660. eng. Raber-Durlacher JE, von Bültzingslöwen I, Logan RM, Bowen J, Al-Azri AR, Everaus H, Gerber E, Gomez JG, Pettersson BG, Soga Y. 2013. Systematic review of cytokines and growth factors for the management of oral mucositis in cancer patients. Supportive Care in Cancer. 21(1):343-355. Raffetto JD, Khalil RA. 2008. Matrix metalloproteinases and their inhibitors in vascular remodeling and vascular disease. Biochemical pharmacology. 75(2):346-359. Ramanan S, Kooshki M, Zhao W, Hsu F-C, Robbins ME. 2008. PPARα ligands inhibit radiation-induced microglial inflammatory responses by negatively regulating NF-κB and AP-1 pathways. Free Radical Biology and Medicine. 45(12):1695-1704. Randall K, Coggle JE. 1996. Long-term expression of transforming growth factor TGF beta 1 in mouse skin after localized beta-irradiation. International journal of radiation biology. 70(3):351-360. eng. Rasoul Y, Peyman A, Saeed R, Abolhasan R, Bagher F, Mohsen C, Hengameh F, Masoud N. 2017. Targeting of Inflammation for Radiation Protection and Mitigation. Current molecular pharmacology. Rath M, Müller I, Kropf P, Closs EI, Munder M. 2014. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Frontiers in immunology. 5:532. Regimbeau J-M, Panis Y, Gouzi J-L, Fagniez P-L, Research FUAfS. 2001. Operative and long term results after surgery for chronic radiation enteritis. The American journal of surgery. 182(3):237-242. Reinhold H, Calvo W, Hopewell J, Van den Berg A. 1990. Development of blood vessel-related radiation damage in the fimbria of the central nervous system. International Journal of Radiation Oncology* Biology* Physics. 18(1):37-42.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Reuter J, Jocher A, Stump J, Grossjohann B, Franke G, Schempp C. 2008. Investigation of the antiinflammatory potential of Aloe vera gel (97.5%) in the ultraviolet erythema test. Skin pharmacology and physiology. 21(2):106-110. Rezaeyan A, Fardid R, Haddadi GH, Takhshid MA, Hosseinzadeh M, Najafi M, Salajegheh A. 2016. Evaluating Radioprotective Effect of Hesperidin on Acute Radiation Damage in the Lung Tissue of Rats. Journal of Biomedical Physics & Engineering. 6(3):165-174. Rezaeyan A FR, Haddadi GH, Takhshid MA, Hosseinzadeh M, Najafi M, et al. 2016. Evaluating radioprotective effect of hesperidin on acute radiation damage in the lung tissue of rats. J Biomed Phys Eng. 6(3):165‑ 174. Rezaeyan A, Haddadi GH, Hosseinzadeh M, Moradi M, Najafi M. 2016. Radioprotective effects of hesperidin on oxidative damages and histopathological changes induced by X-irradiation in rats heart tissue. Journal of medical physics. 41(3):182-191. Rezapoor S, Shirazi A, Abbasi S, Bazzaz JT, Izadi P, Rezaeejam H, Valizadeh M, Soleimani-Mohammadi F, Najafi M. 2017. Modulation of radiation-induced base excision repair pathway gene expression by melatonin. Journal of medical physics. 42(4):245-250. Ricarte-Filho JC, Li S, Garcia-Rendueles ME, Montero-Conde C, Voza F, Knauf JA, Heguy A, Viale A, Bogdanova T, Thomas GA. 2013. Identification of kinase fusion oncogenes in post-Chernobyl radiationinduced thyroid cancers. The Journal of clinical investigation. 123(11):4935-4944. Richardson J, Smith JE, McIntyre M, Thomas R, Pilkington K. 2005. Aloe vera for preventing radiationinduced skin reactions: a systematic literature review. Clinical Oncology. 17(6):478-484. Robbins M, Greene-Schloesser D, Peiffer AM, Shaw E, Chan MD, Wheeler KT. 2012. Radiation-induced brain injury: A review. Frontiers in oncology. 2:73. Robbins M, Zhao W. 2004. Chronic oxidative stress and radiation‐induced late normal tissue injury: a review. International journal of radiation biology. 80(4):251-259. Rock KL, Kono H. 2008. The inflammatory response to cell death. Annual review of pathology. 3:99-126. eng. Rodríguez-Lago I, Carretero C, Herráiz M, Subtil JC, Betés M, Rodríguez-Fraile M, Sola JJ, Bilbao JI, Muñoz-Navas M, Sangro B. 2013. Long-term follow-up study of gastroduodenal lesions after radioembolization of hepatic tumors. World journal of gastroenterology: WJG. 19(19):2935. Rosso M, Blasi G, Gherlone E, Rosso R. 1997. Effect of granulocyte-macrophage colony-stimulating factor on prevention of mucositis in head and neck cancer patients treated with chemo-radiotherapy. Journal of chemotherapy. 9(5):382-385. Russell NS, Hoving S, Heeneman S, Hage JJ, Woerdeman LA, de Bree R, Lohuis PJ, Smeele L, Cleutjens J, Valenkamp A et al. 2009. Novel insights into pathological changes in muscular arteries of radiotherapy patients. Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology. 92(3):477-483. eng. Ryan JL. 2012. Ionizing radiation: the good, the bad, and the ugly. Journal of Investigative Dermatology. 132:985-993. Ryan JL, Heckler CE, Ling M, Katz A, Williams JP, Pentland AP, Morrow GR. 2013. Curcumin for radiation dermatitis: a randomized, double-blind, placebo-controlled clinical trial of thirty breast cancer patients. Radiat Res. 180(1):34-43. Salame MY, Verheye S, Mulkey SP, Chronos NA, King SB, Crocker IR, Robinson KA. 2000. The effect of endovascular irradiation on platelet recruitment at sites of balloon angioplasty in pig coronary arteries. Circulation. 101(10):1087-1090. Salvo N, Barnes E, Van Draanen J, Stacey E, Mitera G, Breen D, Giotis A, Czarnota G, Pang J, De Angelis C. 2010. Prophylaxis and management of acute radiation-induced skin reactions: a systematic review of the literature. Current Oncology. 17(4):94.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Sandoval RL, Koga DH, Buloto LS, Suzuki R, Dib LL. 2003. Management of chemo-and radiotherapy induced oral mucositis with low-energy laser: initial results of AC Camargo Hospital. Journal of applied oral science. 11(4):337-341. Santos J, Ladeira M, Pedrazzoli Jr J, Ribeiro M. 2012. Relationship of IL-1 and TNF-α polymorphisms with Helicobacter pylori in gastric diseases in a Brazilian population. Brazilian Journal of Medical and Biological Research. 45(9):811-817. Sato K, Ozaki K, Oh I, Meguro A, Hatanaka K, Nagai T, Muroi K, Ozawa K. 2007. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood. 109(1):228-234. Sato T, Kinoshita M, Yamamoto T, Ito M, Nishida T, Takeuchi M, Saitoh D, Seki S, Mukai Y. 2015. Treatment of Irradiated Mice with High-Dose Ascorbic Acid Reduced Lethality. PloS one. 10(2). Savarese DM, Savy G, Vahdat L, Wischmeyer PE, Corey B. 2003. Prevention of chemotherapy and radiation toxicity with glutamine. Cancer treatment reviews. 29(6):501-513. Schallenkamp JM, Miller RC, Brinkmann DH, Foote T, Garces YI. 2007. Incidence of radiation pneumonitis after thoracic irradiation: Dose-volume correlates. International journal of radiation oncology, biology, physics. 67(2):410-416. eng. Sciubba JJ, Goldenberg D. 2006. Oral complications of radiotherapy. The lancet oncology. 7(2):175-183. Seyyednejad F, Rezaee A, Haghi S, Goldust M. 2012. Survey of pre-inflammation cytokines levels in radiotherapy-induced-mucositis. Pakistan Journal of Biological Sciences. 15(22):1098. Shadad AK, Sullivan FJ, Martin JD, Egan LJ. 2013. Gastrointestinal radiation injury: prevention and treatment. World journal of gastroenterology: WJG. 19(2):199. Shadad AK, Sullivan FJ, Martin JD, Egan LJ. 2013. Gastrointestinal radiation injury: Symptoms, risk factors and mechanisms. World Journal of Gastroenterology : WJG. 19(2):185-198. Sharma S, Haldar C. 2006. Melatonin prevents X-ray irradiation induced oxidative damagein peripheral blood and spleen of the seasonally breeding rodent, Funambulus pennanti during reproductively active phase. International journal of radiation biology. 82(6):411-419. eng. Shiboski CH, Hodgson TA, Ship JA, Schiødt M. 2007. Management of salivary hypofunction during and after radiotherapy. Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Endodontology. 103:S66. e61-S66. e19. Shih A, Miaskowski C, Dodd MJ, Stotts NA, MacPhail L. 2003. Mechanisms for radiation-induced oral mucositis and the consequences. Cancer nursing. 26(3):222-229. Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM. 2012. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 36(3):401-414. Shinmura K, Kohno T, Kasai H, Koda K, Sugimura H, Yokota J. 1998. Infrequent Mutations of the hOGG1 Gene, That Is Involved in the Excision of 8‐Hydroxyguanine in Damaged DNA, in Human Gastric Cancer. Cancer Science. 89(8):825-828. Shirazi A, Haddadi GH, Ghazi-Khansari M, Abolhassani F, Mahdavi SR, Eshraghyan MR. 2009. Evaluation of melatonin for prevention of radiation myelopathy in irradiated cervical spinal cord. Cell J. 11(1):43-48. Shirazi A, Mihandoost E, Ghobadi G, Mohseni M, Ghazi-khansari M. 2013. Evaluation of Radio-Protective Effect of Melatonin on Whole Body Irradiation Induced Liver Tissue Damage. Cell Journal (Yakhteh). 14(4):292-297. Shukuwa K, Kume K, Yamasaki M, Yoshikawa I, Otsuki M. 2007. Argon plasma coagulation therapy for a hemorrhagic radiation-induced gastritis in patient with pancreatic cancer. Internal Medicine. 46(13):975977. Silva MT. 2010. Secondary necrosis: The natural outcome of the complete apoptotic program. FEBS letters. 584(22):4491-4499.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Siva S, MacManus M, Kron T, Best N, Smith J, Lobachevsky P, Ball D, Martin O. 2014. A pattern of early radiation-induced inflammatory cytokine expression is associated with lung toxicity in patients with nonsmall cell lung cancer. Soloviev AI, Tishkin SM, Parshikov AV, Ivanova IV, Goncharov EV, Gurney AM. 2003. Mechanisms of endothelial dysfunction after ionized radiation: selective impairment of the nitric oxide component of endothelium-dependent vasodilation. British Journal of Pharmacology. 138(5):837-844. Soloviev AI, Tishkin SM, Parshikov AV, Ivanova IV, Goncharov EV, Gurney AM. 2003. Mechanisms of endothelial dysfunction after ionized radiation: selective impairment of the nitric oxide component of endothelium‐dependent vasodilation. British journal of pharmacology. 138(5):837-844. Son Y, Lee HJ, Rho JK, Chung SY, Lee CG, Yang K, Kim SH, Lee M, Shin IS, Kim JS. 2015. The ameliorative effect of silibinin against radiation-induced lung injury: protection of normal tissue without decreasing therapeutic efficacy in lung cancer [journal article]. BMC Pulmonary Medicine. 15(1):68. Song W, Wang X. 2015. The role of TGFβ1 and LRG1 in cardiac remodelling and heart failure. Biophysical Reviews. 7:91-104. Sonis ST. 2004. The pathobiology of mucositis. Nature Reviews Cancer. 4(4):277-284. Sonis ST. 2007. Pathobiology of oral mucositis: novel insights and opportunities. J Support Oncol. 5(9 Suppl 4):3-11. Sorbara MT, Girardin SE. 2011. Mitochondrial ROS fuel the inflammasome. Cell research. 21(4):558. Soussain C, Ricard D, Fike JR, Mazeron J-J, Psimaras D, Delattre J-Y. 2009. CNS complications of radiotherapy and chemotherapy. The Lancet. 374(9701):1639-1651. Springer IN, Niehoff P, Warnke PH, Böcek G, Kovács G, Suhr M, Wiltfang J, Açil Y. 2005. Radiation caries—radiogenic destruction of dental collagen. Oral oncology. 41(7):723-728. Sprung CN, Forrester HB, Siva S, Martin OA. 2015. Immunological markers that predict radiation toxicity. Cancer letters. 368(2):191-197. Stacey R, Green JT. 2013. Radiation-induced small bowel disease: latest developments and clinical guidance. Therapeutic advances in chronic disease.2040622313510730. Stewart F, Hoving S, Russell N. 2010. Vascular damage as an underlying mechanism of cardiac and cerebral toxicity in irradiated cancer patients. Radiation research. 174(6b):865-869. Stoecklein VM, Osuka A, Ishikawa S, Lederer MR, Wanke-Jellinek L, Lederer JA. 2015. Radiation exposure induces inflammasome pathway activation in immune cells. The Journal of Immunology. 194(3):11781189. Stokman M, Oude Nijhuis C, Spijkervet F, De Bont E, Dijkstra P, Daenen S, Gietema J, van der Graaf W, Groen H, Vellenga E. 2006. The role of oral mucositis on the systemic inflammation parameter IL-8 in febrile neutropenic cancer patients. Cancer investigation. 24(5):479-483. Sugihara T, Hattori Y, Yamamoto Y, Qi F, Ichikawa R, Sato A, Liu M-Y, Abe K, Kanno M. 1999. Preferential impairment of nitric oxide–mediated endothelium-dependent relaxation in human cervical arteries after irradiation. Circulation. 100(6):635-641. Suman S, Maniar M, Fornace Jr AJ, Datta K. 2012. Administration of ON 01210. Na after exposure to ionizing radiation protects bone marrow cells by attenuating DNA damage response. Radiation oncology (London, England). 7(6). Sun Y. 2002. The Renin‐Angiotensin‐Aldosterone System and Vascular Remodeling. Congestive Heart Failure. 8(1):11-16. Sun Y, Cheng MK, Griffiths TR, Mellon JK, Kai B, Kriajevska M, Manson MM. 2013. Inhibition of STAT signalling in bladder cancer by diindolylmethane: relevance to cell adhesion, migration and proliferation. Curr Cancer Drug Targets. 13(1):57-68. eng. Symonds R, McIlroy P, Khorrami J, Paul J, Pyper E, Alcock S, McCallum I, Speekenbrink A, McMurray A, Lindemann E. 1996. The reduction of radiation mucositis by selective decontamination antibiotic pastilles: a placebo-controlled double-blind trial. British journal of cancer. 74(2):312.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Tahamtan R, Shabestani Monfared A, Tahamtani Y, Tavassoli A, Akmali M, Mosleh-Shirazi MA, Naghizadeh MM, Ghasemi D, Keshavarz M, Haddadi GH. 2015. Radioprotective Effect of Melatonin on Radiation-Induced Lung Injury and Lipid Peroxidation in Rats. Cell Journal (Yakhteh). 17(1):111-120. Tamir S, Burney S, Tannenbaum SR. 1996. DNA damage by nitric oxide. Chemical research in toxicology. 9(5):821-827. Theis V, Sripadam R, Ramani V, Lal S. 2010. Chronic radiation enteritis. Clinical Oncology. 22(1):70-83. Tian W, Yin X, Wang L, Wang J, Zhu W, Cao J, Yang H. 2015. The key role of miR-21-regulated SOD2 in the medium-mediated bystander responses in human fibroblasts induced by alpha-irradiated keratinocytes. Mutation research. 780:77-85. eng. Tsao MN, Li YQ, Lu G, Xu Y, Wong CS. 1999. Upregulation of vascular endothelial growth factor is associated with radiation-induced blood-spinal cord barrier breakdown. Journal of neuropathology and experimental neurology. 58(10):1051-1060. eng. Tsatsanis CS, DEMETRIOS A. 2000. The role of oncogenic kinases in human cancer (Review). International journal of molecular medicine. 5:583-590. Tsihlis ND, Oustwani CS, Vavra AK, Jiang Q, Keefer LK, Kibbe MR. 2011. Nitric oxide inhibits vascular smooth muscle cell proliferation and neointimal hyperplasia by increasing the ubiquitination and degradation of UbcH10. Cell biochemistry and biophysics. 60(1-2):89-97. eng. Tsoutsou PG, Koukourakis MI. 2006. Radiation pneumonitis and fibrosis: mechanisms underlying its pathogenesis and implications for future research. International Journal of Radiation Oncology* Biology* Physics. 66(5):1281-1293. Tyldesley S, Boyd C, Schulze K, Walker H, Mackillop WJ. 2001. Estimating the need for radiotherapy for lung cancer: an evidence-based, epidemiologic approach. International Journal of Radiation Oncology* Biology* Physics. 49(4):973-985. Ulff E, Maroti M, Serup J, Falkmer U. 2013. A potent steroid cream is superior to emollients in reducing acute radiation dermatitis in breast cancer patients treated with adjuvant radiotherapy. A randomised study of betamethasone versus two moisturizing creams. Radiotherapy and Oncology. 108(2):287-292. Veiko NN. 2013. Oxidized extracellular DNA as a stress signal in human cells. Oxidative medicine and cellular longevity. 2013. Vissink A, Burlage F, Spijkervet F, Jansma J, Coppes R. 2003. Prevention and treatment of the consequences of head and neck radiotherapy. Critical Reviews in Oral Biology & Medicine. 14(3):213225. Vissink A, Jansma J, Spijkervet F, Burlage F, Coppes R. 2003. Oral sequelae of head and neck radiotherapy. Critical Reviews in Oral Biology & Medicine. 14(3):199-212. Von Bültzingslöwen I, Brennan MT, Spijkervet FK, Logan R, Stringer A, Raber-Durlacher JE, Keefe D. 2006. Growth factors and cytokines in the prevention and treatment of oral and gastrointestinal mucositis. Supportive Care in Cancer. 14(6):519-527. Wambi C, Sanzari J, Wan XS, Nuth M, Davis J, Ko Y-H, Sayers CM, Baran M, Ware JH, Kennedy AR. 2008. Dietary antioxidants protect hematopoietic cells and improve animal survival after total-body irradiation. Radiation research. 169(4):384-396. Wang S, Liao Z, Wei X, Liu HH, Tucker SL, Hu C-s, Mohan R, Cox JD, Komaki R. 2006. Analysis of clinical and dosimetric factors associated with treatment-related pneumonitis (TRP) in patients with non–smallcell lung cancer (NSCLC) treated with concurrent chemotherapy and three-dimensional conformal radiotherapy (3D-CRT). International Journal of Radiation Oncology* Biology* Physics. 66(5):1399-1407. Wang TJC, Wu C-C, Chai Y, Lam RKK, Hamada N, Kakinuma S, Uchihori Y, Yu PKN, Hei TK. 2015. Induction of Non-Targeted Stress Responses in Mammary Tissues by Heavy Ions. PloS one. 10(8):e0136307. Wang XS, Shi Q, Williams LA, Mao L, Cleeland CS, Komaki RR, Mobley GM, Liao Z. 2010. Inflammatory cytokines are associated with the development of symptom burden in patients with NSCLC undergoing concurrent chemoradiation therapy. Brain, behavior, and immunity. 24(6):968-974.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Wang Y, Liu L, Pazhanisamy SK, Li H, Meng A, Zhou D. 2010. Total body irradiation causes residual bone marrow injury by induction of persistent oxidative stress in murine hematopoietic stem cells. Free Radic Biol Med. 48(2):348-356. Ward PA, Hunninghake GW. 1998. Lung inflammation and fibrosis. American journal of respiratory and critical care medicine. 157(4):S123-S129. Ward WF, Molteni A, Hinz JM. 1990. Captopril reduces collagen and mast cell accumulation in irradiated rat lung. International Journal of Radiation Oncology* Biology* Physics. 19(6):1405-1409. Weaver DA, Hei TK, Hukku B, McRaven JA, Willey JC. 1997. Cytogenetic and molecular genetic analysis of tumorigenic human bronchial epithelial cells induced by radon alpha particles. Carcinogenesis. 18(6):1251-1257. eng. Weber C, Erl W, Pietsch A, Weber PC. 1995. Aspirin Inhibits Nuclear Factor–κB Mobilization and Monocyte Adhesion in Stimulated Human Endothelial Cells. Circulation. 91(7):1914-1917. Weintraub NL, Jones WK, Manka D. 2010. Understanding Radiation-Induced Vascular Disease. Journal of the American College of Cardiology. 55(12):10.1016/j.jacc.2009.1011.1053. Weintraub NL, Jones WK, Manka D. 2010. Understanding Radiation-Induced Vascular Disease⁎. Journal of the American College of Cardiology. 55(12):1237-1239. Willey JC, Hei TK, Piao CQ, Madrid L, Willey JJ, Apostolakos MJ, Hukku B. 1993. Radiation-induced deletion of chromosomal regions containing tumor suppressor genes in human bronchial epithelial cells. Carcinogenesis. 14(6):1181-1188. eng. Williams MS, Burk M, Loprinzi CL, Hill M, Schomberg PJ, Nearhood K, O'Fallon JR, Laurie JA, Shanahan TG, Moore RL. 1996. Phase III double-blind evaluation of an aloe vera gel as a prophylactic agent for radiation-induced skin toxicity. International Journal of Radiation Oncology* Biology* Physics. 36(2):345349. Wink DA, Laval J. 1994. The Fpg protein, a DNA repair enzyme, is inhibited by the biomediator nitric oxide in vitro and in vivo. Carcinogenesis. 15(10):2125-2129. Wirsdörfer F, Jendrossek V. 2016. The Role of Lymphocytes in Radiotherapy-Induced Adverse Late Effects in the Lung. Frontiers in immunology. 7. Wolf G. 2004. New insights into the pathophysiology of diabetic nephropathy: from haemodynamics to molecular pathology. European journal of clinical investigation. 34(12):785-796. eng. Wong CS, Van der Kogel AJ. 2004. Mechanisms of radiation injury to the central nervous system: implications for neuroprotection. Molecular interventions. 4(5):273. Wong CS, Van Dyk J, Milosevic M, Laperriere NJ. 1994. Radiation myelopathy following single courses of radiotherapy and retreatment. International journal of radiation oncology, biology, physics. 30(3):575581. eng. Wynn T. 2008. Cellular and molecular mechanisms of fibrosis. The Journal of pathology. 214(2):199-210. Wynn TA. 2007. Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. Journal of Clinical Investigation. 117(3):524. Wynn TA, Ramalingam TR. 2012. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nature medicine. 18(7):1028-1040. Xu S, Ding N, Pei H, Hu W, Wei W, Zhang X, Zhou G, Wang J. 2014. MiR-21 is involved in radiationinduced bystander effects. RNA biology. 11(9):1161-1170. eng. Yagi A, Kabash A, Okamura N, Haraguchi H, Moustafa S, Khalifa T. 2002. Antioxidant, free radical scavenging and anti-inflammatory effects of aloesin derivatives in Aloe vera. Planta medica. 68(11):957960. Yahyapour R, Amini P, Rezapoor S, Rezaeyan A, Farhood B, Cheki M, Fallah H, Najafi M. 2017. Targeting of Inflammation for Radiation Protection and Mitigation. Curr Mol Pharmacol. doi: 10.2174/1874467210666171108165641. [Epub ahead of print]

Ac

ce

pt e

d

M

an

us

cr

ip

t

Yahyapour R, Motevaseli E, Rezaeyan A, Abdollahi H, Farhood B, Cheki M, Najafi M, Villa V. 2017. Mechanisms of Radiation Bystander and Non-Targeted Effects: Implications to Radiation Carcinogenesis and Radiotherapy. Curr Radiopharm. doi: 10.2174/1874471011666171229123130. [Epub ahead of print] Yahyapour R, Motevaseli E, Rezaeyan A, Abdollahi H, Farhood B, Cheki M, Rezapoor S, Shabeeb D, Musa AE, Najafi M et al. 2018. Reduction-oxidation (redox) system in radiation-induced normal tissue injury: molecular mechanisms and implications in radiation therapeutics. Clin Transl Oncol. doi: 10.1007/s12094-017-1828-6. [Epub ahead of print] Yamada M, Kubo H, Ota C, Takahashi T, Tando Y, Suzuki T, Fujino N, Makiguchi T, Takagi K, Suzuki T. 2013. The increase of microRNA-21 during lung fibrosis and its contribution to epithelial-mesenchymal transition in pulmonary epithelial cells. Respiratory research. 14(95). Yarnold J, Brotons M-CV. 2010. Pathogenetic mechanisms in radiation fibrosis. Radiotherapy and oncology. 97(1):149-161. Yeoh A, Gibson R, Yeoh E, Bowen J, Stringer A, Giam K, Logan R, Keefe D. 2006. Radiation therapyinduced mucositis: relationships between fractionated radiation, NF-κB, COX-1, and COX-2. Cancer treatment reviews. 32(8):645-651. Yi C, Zhang Y, Yu Z, Xiao Y, Wang J, Qiu H, Yu W, Tang R, Yuan Y, Guo W. 2014. Melatonin enhances the anti-tumor effect of fisetin by inhibiting COX-2/iNOS and NF-κB/p300 signaling pathways. Yonish-Rouach E, Resnitzky D, Lotem J, Sachs L, Kimchi A, Oren M. 1991. Wild-type p53 induces apoptosis of myeloid leukaemic cells that is inhibited by interleukin-6. Nature. 352(6333):345-347. eng. Yu H, Pardoll D, Jove R. 2009. STATs in cancer inflammation and immunity: a leading role for STAT3. Nature reviews Cancer. 9(11):798-809. eng. Yuan H, Gaber MW, McColgan T, Naimark MD, Kiani MF, Merchant TE. 2003. Radiation-induced permeability and leukocyte adhesion in the rat blood-brain barrier: modulation with anti-ICAM-1 antibodies. Brain research. 969(1-2):59-69. eng. Yun HG, Kim HY, Do Yeun Kim YJL. 2015. Successful Treatment of Intractable Bleeding Caused by Radiation-Induced Hemorrhagic Gastritis Using Oral Prednisolone: A Case Report. Cancer research and treatment: official journal of Korean Cancer Association. 47(2):334. Yusuf S, Reddy S, Ôunpuu S, Anand S. 2001. Global burden of cardiovascular diseases. Circulation. 104(23):2855-2864. Zaratiegui M, Irvine DV, Martienssen RA. 2007. Noncoding RNAs and gene silencing. Cell. 128(4):763776. Zhang L, Xia W-J, Zhang Z-S, Lu X-L. 2015. Growth hormone used to control intractable bleeding caused by radiation-induced gastritis. World journal of gastroenterology: WJG. 21(31):9453. Zhang L, Xie X-Y, Wang Y, Wang Y-H, Chen Y, Ren Z-G. 2012. Treatment of radiation-induced hemorrhagic gastritis with prednisolone: a case report. World journal of gastroenterology: WJG. 18(48):7402. Zhao W, Diz D, Robbins M. 2014. Oxidative damage pathways in relation to normal tissue injury. The British journal of radiology. Zhao W, Robbins ME. 2009. Inflammation and chronic oxidative stress in radiation-induced late normal tissue injury: therapeutic implications. Current medicinal chemistry. 16(2):130-143. Zhou H, Ivanov VN, Lien Y-C, Davidson M, Hei TK. 2008. Mitochondrial function and nuclear factor-κB– mediated signaling in radiation-induced bystander effects. Cancer research. 68(7):2233-2240. Zhou R, Yazdi AS, Menu P, Tschopp J. 2011. A role for mitochondria in NLRP3 inflammasome activation. Nature. 469(7329):221-225. Zhu Y, Zhou J, Tao G. 2011. Molecular aspects of chronic radiation enteritis. Clinical & Investigative Medicine. 34(3):119-124.

Ac

ce

pt e

d

M

an

us

cr

ip

t

Zimmermann L, Dudeck O, Schmitt J, Ricke J, Roessner A, Malfertheiner P, Mönkemüller K. 2009. Duodenal ulcer due to yttrium microspheres used for selective internal radiation therapy of hepatocellular cancer. Gastrointestinal endoscopy. 69(4):977-978. Zorov DB, Juhaszova M, Sollott SJ. 2006. Mitochondrial ROS-induced ROS release: an update and review. Biochimica et Biophysica Acta (BBA)-Bioenergetics. 1757(5):509-517.