Next Article in Journal
Statistical Machine-Learning Methods for Genomic Prediction Using the SKM Library
Next Article in Special Issue
Potential Applications of Nanoparticles in Improving the Outcome of Lung Cancer Treatment
Previous Article in Journal
The GJB2 (Cx26) Gene Variants in Patients with Hearing Impairment in the Baikal Lake Region (Russia)
Previous Article in Special Issue
Radiation-Induced Intestinal Normal Tissue Toxicity: Implications for Altered Proteome Profile
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TLR4—A Pertinent Player in Radiation-Induced Heart Disease?

by
Basveshwar Gawali
1,
Vijayalakshmi Sridharan
2,
Kimberly J. Krager
2,
Marjan Boerma
2 and
Snehalata A. Pawar
1,3,*
1
Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
2
Division of Radiation Health, College of Pharmacy, the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
3
Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
*
Author to whom correspondence should be addressed.
Submission received: 27 March 2023 / Revised: 18 April 2023 / Accepted: 26 April 2023 / Published: 28 April 2023

Abstract

:
The heart is one of the organs that is sensitive to developing delayed adverse effects of ionizing radiation (IR) exposure. Radiation-induced heart disease (RIHD) occurs in cancer patients and cancer survivors, as a side effect of radiation therapy of the chest, with manifestation several years post-radiotherapy. Moreover, the continued threat of nuclear bombs or terrorist attacks puts deployed military service members at risk of exposure to total or partial body irradiation. Individuals who survive acute injury from IR will experience delayed adverse effects that include fibrosis and chronic dysfunction of organ systems such as the heart within months to years after radiation exposure. Toll-like receptor 4 (TLR4) is an innate immune receptor that is implicated in several cardiovascular diseases. Studies in preclinical models have established the role of TLR4 as a driver of inflammation and associated cardiac fibrosis and dysfunction using transgenic models. This review explores the relevance of the TLR4 signaling pathway in radiation-induced inflammation and oxidative stress in acute as well as late effects on the heart tissue and the potential for the development of TLR4 inhibitors as a therapeutic target to treat or alleviate RIHD.

1. Introduction

Radiotherapy (RT) is used to treat more than 70% of all cancer types. Improvements in cancer therapy have led to an ever-increasing cancer cure rate. However, both RT and many chemotherapies are associated with side effects due to injury of normal (non-cancer) tissues that are sometimes severe and almost always reduce the quality of life of cancer patients and survivors. From a clinical perspective, the dose of ionizing radiation (IR) that can be delivered is limited by the radiation tolerance of normal tissue [1]. As a result, the impact of these side-effects of RT on the quality of life of cancer survivors due to normal tissue complications is now coming to attention as they represent a growing medical problem [2,3]. An improved understanding of normal tissue injury is therefore required to develop pharmaceutical interventions that increase the safety of cancer therapy [4]. Exposure of the heart to high doses of radiation has been known to cause cardiac dysfunction. In clinic, RT has been widely used for the treatment of thoracic tumors, Hodgkin’s lymphoma, and breast cancer. As every therapy has side effects and limitations, an important side effect is on the cardiac system known as radiation-induced heart disease (RIHD) [5,6,7,8,9,10,11]. Military service members continue to be at risk for exposure to IR because of nuclear warfare, terrorism, or rescue operations after nuclear or radiological events. Moreover, military service members and veterans who are treated for thoracic cancers may also be at risk of cardiac side effects of their cancer treatment.
For decades, the heart was thought to be resistant to radiation doses below those administered in RT [12]. The heart was considered to be safe when less than 10–40% of its volume was exposed to 25 Gy (in conventional RT dose fractions) [12]. However, recent analyses of atomic bomb survivors showed a significant increase in the incidence of cardiovascular disease in individuals exposed to IR (mostly in the form of γ-rays) at doses as low as 2 Gy [13,14,15,16,17]. The estimated radiation-related risk per Gy (ERR Gy−1) was 0.14 (95% confidence interval 0.06–0.23) with a linear dose response and doses greater than 0.5 Gy were associated with an elevated risk of stroke and heart disease [16,18,19]. While these studies suggest there is an elevated risk of cardiovascular disease after doses below ~2 Gy, more research in populations of low-dose radiation exposure is required to determine dose-outcome relationships [19]. Additional epidemiological studies on low-dose exposures due to occupation or medical treatment confirm that cardiovascular alterations occur after lower doses of IR than was previously thought, and these findings have re-opened the debate on the radiation sensitivity of the heart [20,21,22,23].

2. RIHD

RIHD appears several months to years after RT [6,24,25]. The earliest evidence of RIHD was available from data from cancer patients in the 1980s, the period when a higher range of radiation doses was used to treat cancers and a large part of the heart was exposed to radiation [24,26,27,28,29]. RIHD is characterized by structural and functional pathophysiological abnormal changes to the various parts of the heart like the conduction system, pericardium, myocardium, valves, and coronary vessels [10,30,31]. RIHD manifests different forms of cardiac dysfunction, such as accelerated atherosclerosis, adverse myocardial remodeling, conduction abnormalities, fibrosis, and acute pneumonitis risk [8,24,32,33]. In breast cancer patients receiving a dose of 5 Gy to the left ventricular volume, a 7.4% increase in the rate of coronary-related events was found per Gy mean dose to the heart [34]. When the radiation dose is increased from 5 to 9 Gy and greater than 10 Gy, the risk of developing ischemic heart diseases like acute myocardial injury and angina pectoris doubles [35,36].
Acute pericarditis was the first RIHD to appear in thoracic cancer patients who had undergone RT [37]. Patients developed a fever, chest pain, and abnormalities in their electrocardiogram due to an increase in oxidative stress and changes in the conduction system [38]. The early chronic phase is characterized by several compensatory mechanisms such as hypertrophy of cardiomyocytes and proliferation of endothelial cells that manage to survive the initial sub-lethal damage [39]. Insufficiency of these compensatory mechanisms, in turn, drives chronic inflammation and oxidative stress. These processes lead to increased proliferation/activation of fibroblasts which remodel the extracellular matrix causing tissue fibrosis, ultimately in alterations of tissue structure and function [39]. Chronic RIHD includes chronic pericarditis, ischemic heart disease, cardiomyopathy and heart failure, valvular heart disease, and chronic conduction system abnormalities [39,40].
Cardiomyopathy is a condition that develops several years after exposure to radiation therapy and is characterized as heart failure with preserved ejection fraction, a characteristic pathological condition that develops in the first phase of heart failure due to compensatory left ventricular hypertrophy and diastolic dysfunction [39,41]. Ten years after RT, there are several structural changes that occur in the heart, such as cardiac fibrosis, and leaflet thickening regurgitation of mitral valves. If untreated, this leads to over 20 years of stenosis and valvular heart disease [30,42]. Chronic conduction system abnormalities are a pathological condition with QTc prolongation, ventricular extrasystole, and atrioventricular block. Changes in the microvessel density, fibrotic lesions, and chronic conduction system abnormalities occur in up to 5% of patients [37]. IR causes a significant increase in the left ventricular wall thickness reducing the decrease in inner wall diameter. This further progresses to diastolic dysfunction, developing heart failure with elevated left ventricular filling pressures and preserved ejection fraction. This stage tends to lead to heart failure with reduced ejection fraction, which in the later stage involves depositions of collagen and cardiac fibrosis [39,41].
The molecular mechanisms by which IR injures the heart are largely unknown, and pharmaceutical interventions are currently unavailable. It is crucial to manage the cardiovascular complications of RIHD as it impacts not only the quality of life but also increases the cost of healthcare for cancer survivors and military personnel that may be exposed to IR. An improved understanding of RIHD is therefore necessary for developing pharmaceutical interventions to prevent or treat this complication.

3. Pathophysiology of RIHD

Several studies have demonstrated the role of radiation-induced oxidative/nitrosative stress as the initiating event in RIHD [39]. Exposure to IR results in the radiolysis of water molecules within the cell which causes the generation of reactive oxygen species (ROS), which interact with the cellular lipids, proteins, and deoxyribonucleic acid (DNA) [38,43]. The generation of free radicals results from just a few hours after exposure to radiation; however, its effects can last for several years [44,45]. RT interferes with the normal mitochondrial mechanism thereby disrupting the mitochondrial respiratory chain. Various antioxidant enzymes in the human body, like glutathione reductase, superoxide dismutase (SOD), heme oxygenase, glutathione peroxidase, and catalase, are known to have free radical scavenging abilities, but at this step because of excessive generation of ROS and reactive nitrogen species (RNS), these enzymes are incapable to neutralize excessive ROS [46]. This increase in ROS levels post-IR exposure exacerbates pre-existing imbalances in the antioxidant/oxidant system leading to apoptotic cell death. Similarly, oxidative stress also results in the activation of inflammatory cytokines like Interleukin-1 beta (IL1-β) and tumor necrosis factor-alpha (TNF-α) that interfere with cellular homeostatic processes like the Krebs cycle, cellular Ca2+ levels, and metabolism [47]. IR exposure also results in the upregulation of inducible nitric oxide synthase (iNOS) to generate nitric oxide (NO) and promotes radiation-induced tissue injury [48]. Peroxynitrite is produced through the interactions of NO and ROS and constitutes the RNS which causes tissue injury by nitrosylation of tyrosine residues of cellular proteins and rendering them non-functional [49]. The acute phase of RIHD is thus characterized by oxidative and nitrosative damage of the cellular proteins, lipids, and DNA leading to cell death, and results in the release of damage-associated molecular patterns (DAMPs) to stimulate the expression of cytokines and chemokines, which recruit immune cells for clearing the dying/dead cells [39].
The vascular system also plays an important role in the pathophysiology of RIHD, which is characterized by senescence and dysfunction of endothelial cells. In vitro studies in human microvascular endothelial cells showed increased levels of pro-inflammatory cytokines intercellular adhesion molecule 1 (ICAM-1), Interleukin-6 (IL-6), and Interleukin-8 (IL-8) in the absence of immune cells after exposure to a single dose of 15 Gy [50]. This shows the relation between the pro-inflammatory state and the vascular endothelium. Activation of pro-inflammatory cytokines and matrix metalloproteinases (MMP), specifically MMP-1 and MMP-2 in endothelial cells by increased oxidative stress, leads to protease enzyme involvement, recruiting macrophages and neutrophils which results in vasoconstriction and tissue hypoxia. Exposure to IR results in endothelial dysfunction, due to inhibition of thrombomodulin, increased levels of von Willebrand factor, resulting in improper fibrinolysis [51]. Various coagulation factors, like thrombin, cause secretion of chemokines like IL-8 and monocyte chemoattractant peptide which results in chemotaxis of neutrophils in a series of events causing vascular inflammation [52].
IR causes the induction of endoplasmic reticulum (ER) stress, which involves the unfolded protein response which is caused by an imbalance between correctly or incorrectly folded proteins [53]. ER-stimulated cardiomyocytes cause the release of Ca2+ into the cytoplasm which further leads to increased levels of Ca2+ in mitochondria and activation of pro-apoptotic proteins like Bax [54], while surviving cardiomyocytes show compensatory hypertrophy [53].
Fibrosis is the result of chronic radiation-induced damage and is a dynamic process involving oxidative stress, chronic hypoxia, pro-fibrotic cytokines, and alterations at the phenotypic level [55]. Fibrosis is one of the main processes which cause delayed radiation-induced damage, mainly involving the deposition of collagen by activated fibroblasts. After irradiation, neutrophils secrete pro-fibrotic factors like platelet-derived growth factor (PDGF), transforming growth factor-beta (TGF-β), Interleukin-13 (IL-13), and Interleukin-14 (IL-14). Especially TGF-β has a crucial role in the accumulation of myofibroblasts and inhibition of the enzyme collagenase, which is important for breaking down collagen I and collagen III. Connective tissue growth factor, which is induced by TGF-β causes the differentiation of mesenchymal cells and fibroblasts into pro-fibrotic myofibroblasts [56].
Literature review suggests that modification in epigenetics has a significant role in normal tissue injury, specifically fibrosis [57]. Different mechanisms by which epigenetic modifications are achieved are regulation of the non-coding RNA and modification of the histone residue involving acetylation, methylation, phosphorylation, and DNA methylation. Epigenetic reprogramming contributes to radiation-induced fibrosis, particularly via DNA methylation. α-smooth muscle actin (α-SMA), a gene of myofibroblast differentiation, is reported to be regulated by methylation of CpG islands [57]. Fibrosis can be promoted by hypermethylation of genes responsible for apoptosis resulting in decreased cell death [58]. Therefore, methylation-inhibiting agents may be promising new treatment strategies in RIHD. Similarly, irregularity in microRNA activity and histone modifications, which are also types of epigenetic modulation, are linked with fibrosis in the heart and other tissues [57].
Interestingly, several clinical and pre-clinical studies have shown a critical role of the innate immune receptor, Toll-like receptor 4 (TLR4), in cardiovascular disease conditions such as acute myocardial infarction, ischemic heart injury, heart failure, dilated cardiomyopathy, and septic shock [59,60,61,62,63,64,65,66]. In this review, we discuss further the role of Toll-like receptors (TLRs) in the heart and specifically the role of TLR4 in the pathophysiology of the heart and its potential role in RIHD.

4. Role of TLRs in the Heart

Discovered in the year 1996 by Lemaitre et al. in the Drosophila melanogaster, the TLRs are being explored for their role in various pathological conditions as well as the normal mechanism of the human body [67]. These receptors are linked to innate immunity by playing a key role as pattern recognition receptors (PRR). Activation of PRR is caused by DAMPs or PAMPs, leading to an inflammatory cytokine cascade [68]. TLRs are innate immune pattern recognition receptors expressed in immune cells that respond to extrinsic ligands produced by various pathogens, also termed pathogen-associated molecular patterns (PAMPs), and to intrinsic ligands associated with tissue injury, also known as DAMPs [69]. They are also expressed on endothelial, smooth muscle cells, fibroblasts, and cardiac myocytes and are able to release and sense DAMPs releases in response to cellular stress and tissue injury [70]. Currently, a total of ten TLRs in humans and thirteen TLR families in mice have been identified [71]. Several stimuli that activate the TLRs include endogenous ligands like human cardiac myosin, high-mobility group box 1 protein, DNA, messenger RNA, small interfering RNA, fibronectin, hyaluronan, and heat shock proteins, and exogenous agents, like lipopolysaccharides from Gram-negative bacteria, bacterial flagellin and single-stranded RNA [72,73,74,75,76,77,78,79,80,81]. TLRs are activated by the myeloid differentiation primary response gene 88 (MyD88) and Toll-IL1R domain-containing adapter (TIR) and induce transcription of genes required for inflammatory responses. While signaling via TLRs plays a key role in innate immune cells, such as macrophages, to induce inflammation for pathogen clearance, they can also propagate inflammation in response to tissue injury and play a key role in cardiovascular diseases [59].
TLR2 is highly expressed in cardiac myocytes, and its over-activation is implicated in myocardial fibrosis, cardiac dysfunction, activation of apoptosis, and cardiac inflammation [82]. Studies demonstrate the involvement of ROS in TLR2 activation [83]. TLR3 is involved in the regeneration of cardiomyocytes in neonates and plays a crucial role in the activation of NF-kB and Interferon-regulatory factors 3 and 7 [84]. Studies in TLR3-knockout mice revealed that reduced expression of caspase 2 and 3 led to improved cardiac function [84]. TLR3 is an endogenous sensor of necrosis during acute inflammation and an inducer of necroptosis via the TIR-domain-containing adapter-inducing interferon-β (TRIF)-RIP1 pathway [73,85]. TLR3 plays a deleterious role in cardiac dysfunction during polymicrobial sepsis and in animal models of myocardial infarction [86,87]. TLR5 promotes damage by increased expression of inflammatory mediators, such as IL-6, monocyte chemoattractant protein 1, and macrophage inflammatory protein 2 in cardiac dysfunction. TLR7 takes part in nucleotide sensing of the Toll/IL-1 family and is involved in the cardiac inflammation process via activation of MyD88 and TNF receptor-associated factor 6 (TRAF6), resulting in nuclear factor-κB (NF-κB) mediated inflammatory response [88,89]. TLR4 is expressed in different innate immune cells and mainly in heart cardiomyocytes, vascular smooth muscle cells, and fibroblasts [90]. TLR4 causes the activation of inflammatory pathways via the activation of leukocytes, and secretion of cytokines, leading to apoptosis and necrosis of cardiomyocytes [91,92]. Among TLR4 adaptor molecules, MyD88 was the first adaptor molecule discovered to be critical for TLR signaling (MyD88-dependent pathways) [93]. Activated TLR4 signals through the canonical NF-κB pathway increase the expression of pro-inflammatory cytokines, such as TNF-α and interleukin-1β (IL-1β) [94].

5. TLR4 Upregulation Is Associated with Heart Pathologies

Inflammation plays a critical role in the case of several types of cardiac dysfunction [95]. Dysregulation of TLR4 signaling plays a critical role in the initiation and/or progression of cardiovascular diseases, such as ischemia-reperfusion injury, atherosclerosis, myocardial infarction, heart failure, dilated cardiomyopathy, hypertension, sepsis neuropsychiatric, and neurodegenerative disorders [64,66,96,97,98,99,100,101,102,103,104]. For instance, the upregulation of TLR4 causes inflammation and aggravates heart failure [105,106]. Another pathophysiological condition associated with increased TLR4 activation is cardiac arrhythmias [64]. It was shown that activation of TLR4 decreased the action potential duration of cardiomyocytes via the IRF-3 pathway, a MyD88-independent pathway that leads to cardiac arrhythmias [107]. The expression levels of TLR4 mRNA and protein were found to be elevated in mononuclear cells of patients with coronary artery disease, acute myocardial ischemia, atherosclerosis, acute myocardial infarction, heart failure, and dilated cardiomyopathy [62,108,109,110]. TLR4 upregulation was also associated with high levels of ROS and increased production of iNOS that led to damage of the myocardium and to myocardial ischemia-related injuries [111]. In another study, it was found that patients with a history of acute myocardial ischemia showed increased plasma levels of TLR4, which correlated with elevated expression of inflammatory cytokines, such as IL-6, IL-2, IL-8, IL-10, and TNF-α [102,112]. Reports also suggest the involvement of TLR4 in the pathophysiology of acute cardiac dysfunction, which is caused by septic shock as well as myocardial ischemia. Chimeric mice treated with TLR4-deficient leukocytes show a marked reduction in the function of myocardial contraction when treated with lipopolysaccharide (LPS). This demonstrates the involvement of TLR4 in the leukocytes in promoting myocyte dysfunction [103]. Recent studies in patients with SARS-CoV-2 infection also revealed activation of TLR4 in the heart and lungs to cause aberrant TLR4 signaling via the MyD88-dependent pathway that contributes to acute lung injury, myocarditis, cardiac complications, and other severe inflammatory complications, such as the hyper inflammation observed in patients with severe COVID-19 [113].

6. TLR4 Overexpression or Knockdown in Preclinical Models of Cardiovascular Diseases

TLR4 is expressed and elevated following ischemic injury in both leukocytes and cardiac myocytes [99]. Studies in isolated hearts and cardiomyocytes show increased apoptosis mediated by TLR4 in an ischemia-reperfusion model [82]. TLR4-mediated immune cell infiltration and inflammatory cytokines are implicated in the hypertrophy of cardiomyocytes [114]. Mice treated with a TLR4 inhibitor showed a significant decrease in all these parameters, which shows the involvement of TLR4 receptor signaling in cardiac hypertrophy [115]. LPS treatment of rat cardiomyocytes H9C2 in vitro and male Sprague Dawley (SD) rats in vivo revealed increased expression of TLR4 expression, caspase-7/caspase-9, oxidative stress, fibrosis markers, mitochondrial complex activity, and decreased levels of SIRT-2 protein [116]. These findings demonstrate that activation of TLR4 by LPS results in apoptosis, cardiac inflammation, mitochondrial dysfunction, and fibrosis both in vitro and in vivo [116]. TLR4 upregulation in cardiomyocytes is also implicated in a mouse model of doxorubicin-induced dilated cardiomyopathy [117]. An isoproterenol-induced cardiac hypertrophy model of male SD rats showed a significant increase in TLR4 expression along with mitochondrial dysfunction. A recent experimental study has reported that monocyte TLR4 expression is upregulated in patients with heart failure after acute myocardial infarction. The LPS treatment of chimeric mice that received TLR4−/− immune cells, showed significant reduction in the ventricular myocytes shortening. These results suggest that immune cells TLR4 promote cardiac dysfunction in an endotoxemia model [103]. Loss of TLR4 in mice attenuated-aging-associated cardiovascular dysfunction by suppressing oxidative stress by upregulation of Cu-Zn superoxide dismutase (SOD1) and Manganes-SOD (SOD2) and downregulation of senescence markers p16 and p53 [118].
TLR4 inhibitors or knockdown approaches have been shown to reduce cardiac injury in many animal models. Preclinical studies with TLR4 inhibition or gene knock-out approaches resulted in reduced cardiac injury in animal models, including cardiac dysfunction due to trauma-hemorrhage models [59,119]. In a model of hypertension, male SD rats treated with a TLR4 inhibitor showed a significant reduction in myocardial inflammation and expression of iNOS. These results show that TLR4 blockade inhibits the hypertensive response causing downregulation in the myocardial inflammatory activity [120]. Knockdown of TLR4 showed significant improvement in left ventricular remodeling and reduced cell death of cardiomyocytes [106]. Another study revealed reduced infarct size in the case of TLR4-deficient (C3H/HeJ) mice [96]. In myocardial injury models, it was found that TLR4-deficient mice had smaller infarctions and decreased damage to the myocardium, indicating that TLR4 plays a key role in promoting myocardial damage [121]. In the case of TLR4-deficient mice, all the autophagy genes Beclin-1, Atg7, Atg-5, LC-2, and α-SMA were found to be downregulated [106,122]. In spontaneously hypertensive rats, it was shown that silencing of TLR4 inhibited atrial fibrosis and susceptibility to atrial fibrillation through downregulation of the Nod-like receptors’ family pyrin domain containing 3 (NLRP3)-TGF-β [123].

7. Role of TLR4 in RIHD in Rodent Models

Different types of radiation approaches are being applied in pre-clinical settings like partial heart irradiation, whole thorax, and whole heart radiation with radiation delivery methods like conformal systems and single beam with lead shielding [124]. Recent studies in rodent models have confirmed that adverse cardiac remodeling as delayed effects of acute radiation exposure (DEARE) after total body irradiation (TBI) or partial body irradiation (PBI) can indeed be reproduced in animal models [125,126,127,128,129]. Inflammation is thought to play an important role in the development of RIHD [7]. However, very few studies have examined the effect of IR on the expression of TLR4 in the heart. Some recent studies in mouse models revealed a key role of TLR4 activation in acute as well as delayed adverse effects of IR-promoting pulmonary fibrosis, radiation pneumonitis, radiation-induced liver disease, and radiation-induced oral mucositis [130,131,132,133,134]. Another study using a combination of immunotherapy using the PD-1 inhibitor and RT revealed upregulation of TLR4 in mouse hearts [135]. Studies from our group revealed upregulation of left ventricular expression of TLR4 in both male and female C57BL/6 mice at 6 months after exposure to 9.5 Gy gamma rays in a mouse model where both the hind limbs were shielded from radiation to promote long-term survival and allow the study of DEARE [136]. We recently reported an elevated expression of TLR4 in mouse hearts at 6 months post-local heart or partial heart irradiation which correlated with increased markers of cardiac fibrosis [137].
Since previously published studies by us and others consistently show an upregulation of TLR4 in the irradiated heart, we hypothesize that TLR4 upregulation may play an essential role in IR-induced inflammation and oxidative stress in promoting RIHD (Figure 1). Since the heart tissue comprises a significant number of fibroblasts, cardiomyocytes, endothelial, and immune cells, it will be critical to identify the specific cell types involved in dysregulated TLR4 signaling and their role in the development of the sequelae of RIHD. Apart from this, further investigations are also needed to identify the specific type of DAMPs generated in response to radiation that promote the upregulation of TLR4. Radiation induces DAMPs, such as high mobility group box 1 (HMGB1), calreticulin, adenosine triphosphate, and heat shock proteins (HSPs) [138,139]. Apart from the well-known TLR4 ligand-lipopolysaccharide (LPS), DAMPs, such as HMGB1, heat shock factors 60, HSP70, fibrinogen, fibronectin, hyaluronan that are also implicated in cardiovascular diseases, are reported to play a role in the activation of TLR4 [77,82,140,141].
In addition, as it is well-known that inflammation in the early time points post-IR may be essential to recruit immune cells for clearing dying/damaged cells, further investigations will be needed to address the optimal timing for administration of TLR4 inhibitors or antagonists post-radiation exposure in order to see beneficial effects on alleviating RIHD. Interestingly, studies with TLR2 agonists (bacterial lipoprotein, CBL502) and TLR4 agonists (LPS, or its derivatives) treated prior to radiation showed significant protection against acute injury to hematopoietic and intestine tissues [141,142,143,144,145,146,147]. However, the role of these agonists in the context of DEARE has not been investigated and whether they may impart radioprotective effects needs further investigation. In the case of Doxorubicin-associated cardiotoxicity in human patients, it was also observed that elevated serum levels of TLR4 correlated with increased myocardial iron load and left ventricular dysfunction with decreased ejection fraction [148]. In the case of RIHD, a similar follow-up of patients prior to and post-RT is needed to determine whether elevated serum levels of TLR4 correlate positively with delayed adverse effects on the heart.

8. Conclusions

RIHD remains an important complication of cancer RT regimens. The only available approach currently is to limit complications from RT through efforts to reduce cardiac exposure to IR. Despite technological improvements in treatment planning and delivery of RT, patients with cancers in the lung, esophagus, and proximal stomach can still receive significant doses of IR to the heart and continue to be at risk of IR-induced cardiomyopathy [149,150,151]. Currently, there are no approved radiation countermeasures to alleviate or prevent RIHD.
Various pre-clinical and clinical models have shown the importance of TLR4 in promoting inflammation and oxidative stress in cardiovascular diseases. Similar work by us and others show that mouse models of localized radiation therapy as well as TBI resulted in elevated expression of TLR4 in mouse hearts and correlated with markers of adverse tissue remodeling and cardiac dysfunction. This suggests that TLR4 antagonists or inhibitors may be suitable to block the downstream effects of the TLR4/MyD88-dependent pathway in inflammation and oxidative/nitrosative stress and alleviate the radiation-related adverse effects on the heart (Figure 1). Further research is needed to identify the mechanistic role of the TLR4 pathway and the optimal timing for the administration of a TLR4 antagonist or inhibitor to treat or prevent RIHD. The effects of TLR4 inhibition on other organ systems also need to be assessed before the clinical development of TLR4 inhibitors for treatments of RIHD.

Author Contributions

Conceptualization, B.G. and S.A.P.; literature review: B.G., S.A.P., M.B., V.S., K.J.K. and M.B.; writing—original draft preparation, B.G. and S.A.P.; writing—review and editing, M.B. and S.A.P.; supervision, S.A.P. and M.B.; project administration, M.B. and S.A.P.; funding acquisition, S.A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Department of Defense, grant number PR181863, contract number W81XWH1910737, and the National Institute of General Medical Sciences, grant number P20 GM109005.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Illustrations in Figure 1 were created with www.biorender.com accessed on 20 April 2023, publication licensing rights agreement number DN25AQ9CWR.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

List of Abbreviations

Alpha-smooth muscle actinα-SMA
Cu-Zn superoxide dismutase SOD1
Damage-associated molecular patternDAMP
Delayed effects of acute radiation exposure DEARE
Endoplasmic reticulumER
Heat shock proteins HSPs
High mobility group box 1 HMGB1
Ionizing RadiationIR
Inducible nitric oxide synthase iNOS
Intercellular adhesion molecule 1ICAM-1
Interleukin-1 betaIL-1β
Interleukin-6IL-6
Interleukin-8 IL-8
Interleukin-13 IL-13
Interleukin-14 IL-14
LipopolysaccharideLPS
Manganese-SOD SOD2
Matrix metalloproteinases MMP
Myeloid differentiation primary response 88MyD88
Nitric oxide NO
Nod-like receptors family pyrin domain containing 3 NLRP3
Nuclear factor-kappa B NF-κB
Partial Body Irradiation PBI
Pattern Recognition Receptor PRR
Pathogen-associated molecular patternPAMP
Platelet-derived growth factor PDGF
Radiation-induced heart diseaseRIHD
RadiotherapyRT
Reactive oxygen speciesROS
Reactive Nitrogen SpeciesRNS
Sprague DawleySD
Toll-like receptorTLR
Toll-like receptor 2TLR2
Toll-like receptor 3TLR3
Toll-like receptor 4TLR4
Toll-like receptor 5TLR5
Toll-like receptor 7TLR7
Toll-IL1R domain-containing adapterTIR
Total body irradiationTBI
Tumor necrosis factor alphaTNF-α
TNF receptor-associated factor 6 TRAF6
TIR-domain-containing adapter-inducing interferon-βTRIF
Transforming growth factor betaTGF-β

References

  1. Thompson, M.K.; Poortmans, P.; Chalmers, A.J.; Faivre-Finn, C.; Hall, E.; Huddart, R.A.; Lievens, Y.; Sebag-Montefiore, D.; Coles, C.E. Practice-changing radiation therapy trials for the treatment of cancer: Where are we 150 years after the birth of Marie Curie? Br. J. Cancer 2018, 119, 389–407. [Google Scholar] [CrossRef]
  2. Zheng, H.C.; Onderko, L.; Francis, S.A. Cardiovascular Risk in Survivors of Cancer. Curr. Cardiol. Rep. 2017, 19, 64–65. [Google Scholar] [CrossRef] [PubMed]
  3. Siegel, R.; DeSantis, C.; Virgo, K.; Stein, K.; Mariotto, A.; Smith, T.; Cooper, D.; Gansler, T.; Lerro, C.; Fedewa, S.; et al. Cancer treatment and survivorship statistics, 2012. CA A Cancer J. Clin. 2012, 62, 220–241. [Google Scholar] [CrossRef] [PubMed]
  4. Prasanna, P.; Stone, H.; Wong, R.; Capala, J.V.B.; Coleman, C. Normal tissue protection for improving radiotherapy: Where are the Gaps? Transl. Cancer Res. 2012, 1, 35–48. [Google Scholar] [PubMed]
  5. Boerma, M.; Sridharan, V.; Mao, X.W.; Nelson, G.A.; Cheema, A.K.; Koturbash, I.; Singh, S.P.; Tackett, A.J.; Hauer-Jensen, M. Effects of ionizing radiation on the heart. Mutat. Res. Rev. Mutat. Res. 2016, 770, 319–327. [Google Scholar] [CrossRef]
  6. Darby, S.C.; Cutter, D.J.; Boerma, M.; Constine, L.S.; Fajardo, L.F.; Kodama, K.; Mabuchi, K.; Marks, L.B.; Mettler, F.A.; Pierce, L.J.; et al. Radiation-related heart disease: Current knowledge and future prospects. Int. J. Radiat. Oncol. Biol. Phys. 2010, 76, 656–665. [Google Scholar] [CrossRef]
  7. Ellahham, S.; Khalouf, A.; Elkhazendar, M.; Dababo, N.; Manla, Y. An overview of radiation-induced heart disease. Radiat. Oncol. J. 2022, 40, 89–102. [Google Scholar] [CrossRef]
  8. Martinou, M.; Gaya, A. Cardiac complications after radical radiotherapy. Semin. Oncol. 2013, 40, 178–185. [Google Scholar] [CrossRef]
  9. Yusuf, S.W.; Sami, S.; Daher, I.N. Radiation-induced heart disease: A clinical update. Cardiol. Res. Pract. 2011, 2011, 317659. [Google Scholar] [CrossRef]
  10. Šteiner, I. Pathology of radiation induced heart disease. Rep. Pract. Oncol. Radiother. 2020, 25, 178–181. [Google Scholar] [CrossRef]
  11. Dess, R.T.; Sun, Y.; Matuszak, M.M.; Sun, G.; Soni, P.D.; Bazzi, L.; Murthy, V.L.; Hearn, J.W.D.; Kong, F.M.; Kalemkerian, G.P.; et al. Cardiac Events After Radiation Therapy: Combined Analysis of Prospective Multicenter Trials for Locally Advanced Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2017, 35, 1395–1402. [Google Scholar] [CrossRef]
  12. Emami, B.; Lyman, J.; Brown, A.; Coia, L.; Goitein, M.; Munzenrider, J.E.; Shank, B.; Solin, L.J.; Wesson, M. Tolerance of normal tissue to therapeutic irradiation. Int. J. Radiat. Oncol. Biol. Phys. 1991, 21, 109–122. [Google Scholar] [CrossRef] [PubMed]
  13. Ozasa, K.; Shimizu, Y.; Suyama, A.; Kasagi, F.; Soda, M.; Grant, E.J.; Sakata, R.; Sugiyama, H.; Kodama, K. Studies of the mortality of atomic bomb survivors, Report 14, 1950–2003: An overview of cancer and noncancer diseases. Radiat. Res. 2012, 177, 229–243. [Google Scholar] [CrossRef] [PubMed]
  14. Preston, D.L.; Shimizu, Y.; Pierce, D.A.; Suyama, A.; Mabuchi, K. Studies of mortality of atomic bomb survivors. Report 13: Solid cancer and noncancer disease mortality: 1950–1997. Radiat. Res. 2003, 160, 381–407. [Google Scholar] [CrossRef]
  15. Adams, M.J.; Grant, E.J.; Kodama, K.; Shimizu, Y.; Kasagi, F.; Suyama, A.; Sakata, R.; Akahoshi, M. Radiation dose associated with renal failure mortality: A potential pathway to partially explain increased cardiovascular disease mortality observed after whole-body irradiation. Radiat. Res. 2012, 177, 220–228. [Google Scholar] [CrossRef] [PubMed]
  16. Ozasa, K.; Takahashi, I.; Grant, E.J. Radiation-related risks of non-cancer outcomes in the atomic bomb survivors. Ann. ICRP 2016, 45, 253–261. [Google Scholar] [CrossRef]
  17. Little, M.P.; Azizova, T.V.; Hamada, N. Low- and moderate-dose non-cancer effects of ionizing radiation in directly exposed individuals, especially circulatory and ocular diseases: A review of the epidemiology. Int. J. Radiat. Biol. 2021, 97, 782–803. [Google Scholar] [CrossRef]
  18. Shimizu, Y.; Kodama, K.; Nishi, N.; Kasagi, F.; Suyama, A.; Soda, M.; Grant, E.J.; Sugiyama, H.; Sakata, R.; Moriwaki, H.; et al. Radiation exposure and circulatory disease risk: Hiroshima and Nagasaki atomic bomb survivor data, 1950–2003. BMJ 2010, 340, b5349. [Google Scholar] [CrossRef]
  19. Simon, S.L.; Kendall, G.M.; Bouffler, S.D.; Little, M.P. The Evidence for Excess Risk of Cancer and Non-Cancer Disease at Low Doses and Dose Rates. Radiat. Res. 2022, 198, 615–624. [Google Scholar] [CrossRef] [PubMed]
  20. Little, M.P.; Tawn, E.J.; Tzoulaki, I.; Wakeford, R.; Hildebrandt, G.; Paris, F.; Tapio, S.; Elliott, P. A Systematic Review of Epidemiological Associations between Low and Moderate Doses of Ionizing Radiation and Late Cardiovascular Effects, and Their Possible Mechanisms. Radiat. Res. 2008, 169, 99–109, 111. [Google Scholar] [CrossRef]
  21. Schultz-Hector, S.; Trott, K.R. Radiation-induced cardiovascular diseases: Is the epidemiologic evidence compatible with the radiobiologic data? Int. J. Radiat. Oncol. Biol. Phys. 2007, 67, 10–18. [Google Scholar] [CrossRef] [PubMed]
  22. Stewart, A.M.; Kneale, G.W. A-bomb survivors: Factors that may lead to a re-assessment of the radiation hazard. Int. J. Epidemiol. 2000, 29, 708–714. [Google Scholar] [CrossRef]
  23. Azimzadeh, O.; Azizova, T.; Merl-Pham, J.; Subramanian, V.; Bakshi, M.V.; Moseeva, M.; Zubkova, O.; Hauck, S.M.; Anastasov, N.; Atkinson, M.J.; et al. A dose-dependent perturbation in cardiac energy metabolism is linked to radiation-induced ischemic heart disease in Mayak nuclear workers. Oncotarget 2017, 8, 9067–9078. [Google Scholar] [CrossRef]
  24. Darby, S.C.; Ewertz, M.; McGale, P.; Bennet, A.M.; Blom-Goldman, U.; Brønnum, D.; Correa, C.; Cutter, D.; Gagliardi, G.; Gigante, B. Risk of ischemic heart disease in women after radiotherapy for breast cancer. N. Engl. J. Med. 2013, 368, 987–998. [Google Scholar] [CrossRef] [PubMed]
  25. Sarfati, D.; Koczwara, B.; Jackson, C. The impact of comorbidity on cancer and its treatment. CA A Cancer J. Clin. 2016, 66, 337–350. [Google Scholar] [CrossRef] [PubMed]
  26. Friedlander, A.H.; Sung, E.C.; Child, J.S. Radiation-induced heart disease after Hodgkin’s disease and breast cancer treatment: Dental implications. J. Am. Dent. Assoc. 2003, 134, 1615–1620. [Google Scholar] [CrossRef]
  27. Adams, M.J.; Lipsitz, S.R.; Colan, S.D.; Tarbell, N.J.; Treves, S.T.; Diller, L.; Greenbaum, N.; Mauch, P.; Lipshultz, S.E. Cardiovascular status in long-term survivors of Hodgkin’s disease treated with chest radiotherapy. J. Clin. Oncol. 2004, 22, 3139–3148. [Google Scholar] [CrossRef]
  28. Sardaro, A.; Petruzzelli, M.F.; D’Errico, M.P.; Grimaldi, L.; Pili, G.; Portaluri, M. Radiation-induced cardiac damage in early left breast cancer patients: Risk factors, biological mechanisms, radiobiology, and dosimetric constraints. Radiother. Oncol. 2012, 103, 133–142. [Google Scholar] [CrossRef]
  29. Menezes, K.M.; Wang, H.; Hada, M.; Saganti, P.B. Radiation Matters of the Heart: A Mini Review. Front. Cardiovasc. Med. 2018, 5, 83–83. [Google Scholar] [CrossRef]
  30. Taunk, N.K.; Haffty, B.G.; Kostis, J.B.; Goyal, S. Radiation-induced heart disease: Pathologic abnormalities and putative mechanisms. Front. Oncol. 2015, 5, 39. [Google Scholar] [CrossRef]
  31. Wei, T.; Cheng, Y. The cardiac toxicity of radiotherapy—A review of characteristics, mechanisms, diagnosis, and prevention. Int. J. Radiat. Biol. 2021, 97, 1333–1340. [Google Scholar] [CrossRef] [PubMed]
  32. Heidenreich, P.A.; Hancock, S.L.; Lee, B.K.; Mariscal, C.S.; Schnittger, I. Asymptomatic cardiac disease following mediastinal irradiation. J. Am. Coll. Cardiol. 2003, 42, 743–749. [Google Scholar] [CrossRef]
  33. Yu, L.; Feng, Z. The Role of Toll-Like Receptor Signaling in the Progression of Heart Failure. Mediat. Inflamm. 2018, 2018, 1–11. [Google Scholar] [CrossRef] [PubMed]
  34. Brown, K.N.; Hussain, K.; Richards, J.R. Radiation Induced Coronary Artery Disease. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2023. [Google Scholar]
  35. van den Bogaard, V.A.; Ta, B.D.; van der Schaaf, A.; Bouma, A.B.; Middag, A.M.; Bantema-Joppe, E.J.; van Dijk, L.V.; van Dijk-Peters, F.B.; Marteijn, L.A.; de Bock, G.H. Validation and modification of a prediction model for acute cardiac events in patients with breast cancer treated with radiotherapy based on three-dimensional dose distributions to cardiac substructures. J. Clin. Oncol. 2017, 35, 1171–1178. [Google Scholar] [CrossRef] [PubMed]
  36. van Nimwegen, F.A.; Schaapveld, M.; Cutter, D.J.; Janus, C.; Krol, A.; Hauptmann, M.; Kooijman, K.; Roesink, J.; van der Maazen, R.; Darby, S.C. Radiation dose-response relationship for risk of coronary heart disease in survivors of Hodgkin lymphoma. J. Clin. Oncol. 2015, 34, 235–243. [Google Scholar] [CrossRef] [PubMed]
  37. Cuomo, J.R.; Sharma, G.K.; Conger, P.D.; Weintraub, N.L. Novel concepts in radiation-induced cardiovascular disease. World J. Cardiol. 2016, 8, 504–519. [Google Scholar] [CrossRef]
  38. Ping, Z.; Peng, Y.; Lang, H.; Xinyong, C.; Zhiyi, Z.; Xiaocheng, W.; Hong, Z.; Liang, S. Oxidative Stress in Radiation-Induced Cardiotoxicity. Oxidative Med. Cell. Longev. 2020, 2020, 3579143. [Google Scholar] [CrossRef]
  39. Sárközy, M.; Varga, Z.; Gáspár, R.; Szűcs, G.; Kovács, M.G.; Kovács, Z.Z.; Dux, L.; Kahán, Z.; Csont, T. Pathomechanisms and therapeutic opportunities in radiation-induced heart disease: From bench to bedside. Clin. Res. Cardiol. 2021, 110, 507–531. [Google Scholar] [CrossRef]
  40. Zhuang, X.F.; Yang, Y.M.; Sun, X.L.; Liao, Z.K.; Huang, J. Late onset radiation-induced constrictive pericarditis and cardiomyopathy after radiotherapy: A case report. Medicine 2017, 96, e5932. [Google Scholar] [CrossRef]
  41. Saiki, H.; Moulay, G.; Guenzel, A.J.; Liu, W.; Decklever, T.D.; Classic, K.L.; Pham, L.; Chen, H.H.; Burnett, J.C.; Russell, S.J.; et al. Experimental cardiac radiation exposure induces ventricular diastolic dysfunction with preserved ejection fraction. American J. Physiol. -Heart Circ. Physiol. 2017, 313, H392–H407. [Google Scholar] [CrossRef]
  42. Cutter, D.J.; Schaapveld, M.; Darby, S.C.; Hauptmann, M.; Van Nimwegen, F.A.; Krol, A.D.; Janus, C.P.; van Leeuwen, F.E.; Aleman, B.M. Risk for valvular heart disease after treatment for Hodgkin lymphoma. JNCI J. Natl. Cancer Inst. 2015, 107, djv008. [Google Scholar] [CrossRef] [PubMed]
  43. Hall, E.J.; Giaccia, A.J. Radiobiology for the Radiologist, 8th ed.; Wolters Kluwer Health: Philadelphia, PA, USA, 2019. [Google Scholar]
  44. Yahyapour, R.; Motevaseli, E.; Rezaeyan, A.; Abdollahi, H.; Farhood, B.; Cheki, M.; Rezapoor, S.; Shabeeb, D.; Musa, A.E.; Najafi, M.; et al. Reduction-oxidation (redox) system in radiation-induced normal tissue injury: Molecular mechanisms and implications in radiation therapeutics. Clin. Transl. Oncol. 2018, 20, 975–988. [Google Scholar] [CrossRef] [PubMed]
  45. Spitz, D.R.; Hauer-Jensen, M. Ionizing radiation-induced responses: Where free radical chemistry meets redox biology and medicine. Antioxid Redox Signal 2014, 20, 1407–1409. [Google Scholar] [CrossRef] [PubMed]
  46. Azzam, E.I.; Jay-Gerin, J.-P.; Pain, D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012, 327, 48–60. [Google Scholar] [CrossRef]
  47. Spitz, D.R.; Azzam, E.I.; Jian Li, J.; Gius, D. Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation: A unifying concept in stress response biology. Cancer Metastasis Rev. 2004, 23, 311–322. [Google Scholar] [CrossRef]
  48. Zhao, W.; Robbins, M.E. Inflammation and chronic oxidative stress in radiation-induced late normal tissue injury: Therapeutic implications. Curr. Med. Chem. 2009, 16, 130–143. [Google Scholar] [CrossRef]
  49. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric Oxide and Peroxynitrite in Health and Disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef]
  50. Wondergem, J.; Wedekind, L.E.; Bart, C.I.; Chin, A.; van der Laarse, A.; Beekhuizen, H. Irradiation of mechanically-injured human arterial endothelial cells leads to increased gene expression and secretion of inflammatory and growth promoting cytokines. Atherosclerosis 2004, 175, 59–67. [Google Scholar] [CrossRef]
  51. Baselet, B.; Sonveaux, P.; Baatout, S.; Aerts, A. Pathological effects of ionizing radiation: Endothelial activation and dysfunction. Cell. Mol. Life Sci. 2019, 76, 699–728. [Google Scholar] [CrossRef]
  52. Venkatesulu, B.P.; Mahadevan, L.S.; Aliru, M.L.; Yang, X.; Bodd, M.H.; Singh, P.K.; Yusuf, S.W.; Abe, J.I.; Krishnan, S. Radiation-Induced Endothelial Vascular Injury: A Review of Possible Mechanisms. JACC Basic Transl. Sci. 2018, 3, 563–572. [Google Scholar] [CrossRef]
  53. Gotoh, T.; Endo, M.; Oike, Y. Endoplasmic Reticulum Stress-Related Inflammation and Cardiovascular Diseases. International J. Inflamm. 2011, 2011, 259462. [Google Scholar] [CrossRef]
  54. Livingston, K.; Schlaak, R.A.; Puckett, L.L.; Bergom, C. The role of mitochondrial dysfunction in radiation-induced heart disease: From bench to bedside. Front. Cardiovasc. Med. 2020, 7, 20. [Google Scholar] [CrossRef]
  55. Wang, B.; Wei, J.; Meng, L.; Wang, H.; Qu, C.; Chen, X.; Xin, Y.; Jiang, X. Advances in pathogenic mechanisms and management of radiation-induced fibrosis. Biomed Pharm. 2020, 121, 109560. [Google Scholar] [CrossRef] [PubMed]
  56. Pohlers, D.; Brenmoehl, J.; Löffler, I.; Müller, C.K.; Leipner, C.; Schultze-Mosgau, S.; Stallmach, A.; Kinne, R.W.; Wolf, G. TGF-β and fibrosis in different organs—Molecular pathway imprints. Biochim. Et Biophys. Acta (BBA)—Mol. Basis Dis. 2009, 1792, 746–756. [Google Scholar] [CrossRef]
  57. Weigel, C.; Schmezer, P.; Plass, C.; Popanda, O. Epigenetics in radiation-induced fibrosis. Oncogene 2015, 34, 2145–2155. [Google Scholar] [CrossRef]
  58. Tao, H.; Yang, J.-J.; Shi, K.-H.; Deng, Z.-Y.; Li, J. DNA methylation in cardiac fibrosis: New advances and perspectives. Toxicology 2014, 323, 125–129. [Google Scholar] [CrossRef]
  59. Moghimpour Bijani, F.; Vallejo, J.G.; Rezaei, N. Toll-like Receptor Signaling Pathways in Cardiovascular Diseases: Challenges and Opportunities. Int. Rev. Immunol. 2012, 31, 379–395. [Google Scholar] [CrossRef]
  60. Yang, Y.; Lv, J.; Jiang, S.; Ma, Z.; Wang, D.; Hu, W.; Deng, C.; Fan, C.; Di, S.; Sun, Y.; et al. The emerging role of Toll-like receptor 4 in myocardial inflammation. Cell Death Dis. 2016, 7, e2234. [Google Scholar] [CrossRef] [PubMed]
  61. Frantz, S.; Kobzik, L.; Kim, Y.D.; Fukazawa, R.; Medzhitov, R.; Lee, R.T.; Kelly, R.A. Toll4 (TLR4) expression in cardiac myocytes in normal and failing myocardium. J. Clin. Investig. 1999, 104, 271–280. [Google Scholar] [CrossRef] [PubMed]
  62. Wu, B.; Li, J.; Ni, H.; Zhuang, X.; Qi, Z.; Chen, Q.; Wen, Z.; Shi, H.; Luo, X.; Jin, B. TLR4 activation promotes the progression of experimental autoimmune myocarditis to dilated cardiomyopathy by inducing mitochondrial dynamic imbalance. Oxidative Med. Cell. Longev. 2018, 2018, 3181278. [Google Scholar] [CrossRef]
  63. Munoz-Rodriguez, C.; Fernandez, S.; Osorio, J.M.; Olivares, F.; Anfossi, R.; Bolivar, S.; Humeres, C.; Boza, P.; Vivar, R.; Pardo-Jimenez, V.; et al. Expression and function of TLR4- induced B1R bradykinin receptor on cardiac fibroblasts. Toxicol. Appl. Pharmacol. 2018, 351, 46–56. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, L.; Wang, Y.; Cao, Z.Y.; Wang, M.M.; Liu, X.M.; Gao, T.; Hu, Q.K.; Yuan, W.J.; Lin, L. Up-regulated TLR4 in cardiomyocytes exacerbates heart failure after long-term myocardial infarction. J. Cell. Mol. Med. 2015, 19, 2728–2740. [Google Scholar] [CrossRef] [PubMed]
  65. Becher, P.M.; Hinrichs, S.; Fluschnik, N.; Hennigs, J.K.; Klingel, K.; Blankenberg, S.; Westermann, D.; Lindner, D. Role of Toll-like receptors and interferon regulatory factors in different experimental heart failure models of diverse etiology: IRF7 as novel cardiovascular stress-inducible factor. PLoS ONE 2018, 13, e0193844. [Google Scholar] [CrossRef] [PubMed]
  66. Avlas, O.; Bragg, A.; Fuks, A.; Nicholson, J.D.; Farkash, A.; Porat, E.; Aravot, D.; Levy-Drummer, R.S.; Cohen, C.; Shainberg, A.; et al. TLR4 Expression Is Associated with Left Ventricular Dysfunction in Patients Undergoing Coronary Artery Bypass Surgery. PLoS ONE 2015, 10, e0120175. [Google Scholar] [CrossRef] [PubMed]
  67. Komal, S.; Komal, N.; Mujtaba, A.; Wang, S.-H.; Zhang, L.-R.; Han, S.-N. Potential therapeutic strategies for myocardial infarction: The role of Toll-like receptors. Immunol. Res. 2022, 70, 607–623. [Google Scholar] [CrossRef]
  68. Roh, J.S.; Sohn, D.H. Damage-associated molecular patterns in inflammatory diseases. Immune Netw. 2018, 18, e27. [Google Scholar] [CrossRef]
  69. Chen, G.Y.; Nuñez, G. Sterile inflammation: Sensing and reacting to damage. Nat. Rev. Immunol. 2010, 10, 826–837. [Google Scholar] [CrossRef]
  70. Bezhaeva, T.; Karper, J.; Quax, P.H.A.; de Vries, M.R. The Intriguing Role of TLR Accessory Molecules in Cardiovascular Health and Disease. Front. Cardiovasc. Med. 2022, 9, 1–17. [Google Scholar] [CrossRef]
  71. Takeda, K.; Akira, S. Toll-like Receptors. Curr. Protoc. Immunol. 2015, 109, 14.12.1–14.12.10. [Google Scholar] [CrossRef]
  72. Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Criollo, A.; Ortiz, C.; Lidereau, R.; Mariette, C.; Chaput, N.; Mira, J.P.; Delaloge, S.; et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol. Rev. 2007, 220, 47–59. [Google Scholar] [CrossRef]
  73. Cavassani, K.A.; Ishii, M.; Wen, H.; Schaller, M.A.; Lincoln, P.M.; Lukacs, N.W.; Hogaboam, C.M.; Kunkel, S.L. TLR3 is an endogenous sensor of tissue necrosis during acute inflammatory events. J. Exp. Med. 2008, 205, 2609–2621. [Google Scholar] [CrossRef] [PubMed]
  74. Heil, F.; Hemmi, H.; Hochrein, H.; Ampenberger, F.; Kirschning, C.; Akira, S.; Lipford, G.; Wagner, H.; Bauer, S. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 2004, 303, 1526–1529. [Google Scholar] [CrossRef] [PubMed]
  75. Doddapattar, P.; Gandhi, C.; Prakash, P.; Dhanesha, N.; Grumbach, I.M.; Dailey, M.E.; Lentz, S.R.; Chauhan, A.K. Fibronectin splicing variants containing extra domain a promote atherosclerosis in mice through toll-like receptor 4. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 2391–2400. [Google Scholar] [CrossRef] [PubMed]
  76. Takeda, K.; Akira, S. Toll-like receptors in innate immunity. Int. Immunol. 2005, 17, 1–14. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, P.; Cox, C.J.; Alvarez, K.M.; Cunningham, M.W. Cutting edge: Cardiac myosin activates innate immune responses through TLRs. J. Immunol. 2009, 183, 27–31. [Google Scholar] [CrossRef]
  78. Lau, C.M.; Broughton, C.; Tabor, A.S.; Akira, S.; Flavell, R.A.; Mamula, M.J.; Christensen, S.R.; Shlomchik, M.J.; Viglianti, G.A.; Rifkin, I.R. RNA-associated autoantigens activate B cells by combined B cell antigen receptor/Toll-like receptor 7 engagement. J. Exp. Med. 2005, 202, 1171–1177. [Google Scholar] [CrossRef] [PubMed]
  79. Zughaier, S.M.; Zimmer, S.M.; Datta, A.; Carlson, R.W.; Stephens, D.S. Differential Induction of the Toll-Like Receptor 4-MyD88-Dependent and -Independent Signaling Pathways by Endotoxins. Infect. Immun. 2005, 73, 2940–2950. [Google Scholar] [CrossRef]
  80. Vijay-Kumar, M.; Aitken, J.D.; Sanders, C.J.; Frias, A.; Sloane, V.M.; Xu, J.; Neish, A.S.; Rojas, M.; Gewirtz, A.T. Flagellin Treatment Protects against Chemicals, Bacteria, Viruses, and Radiation. J. Immunol. 2008, 180, 8280–8285. [Google Scholar] [CrossRef]
  81. Prakash, P.; Kulkarni, P.P.; Lentz, S.R.; Chauhan, A.K. Cellular fibronectin containing extra domain A promotes arterial thrombosis in mice through platelet Toll-like receptor 4. Blood J. Am. Soc. Hematol. 2015, 125, 3164–3172. [Google Scholar] [CrossRef]
  82. Lin, L.; Knowlton, A.A. Innate immunity and cardiomyocytes in ischemic heart disease. Life Sci. 2014, 100, 1–8. [Google Scholar] [CrossRef]
  83. Tang, Y.; Xu, Z.; Chen, X.; Wang, N.; Deng, X.; Peng, L.; Chen, Q.; Cai, H. Effects of enalapril on TLR2/NF-κB signaling pathway and inflammatory factors in rabbits with chronic heart failure. Evid. -Based Complement. Altern. Med. 2021, 2021, 9594607. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, X.; Ha, T.; Liu, L.; Hu, Y.; Kao, R.; Kalbfleisch, J.; Williams, D.; Li, C. TLR3 mediates repair and regeneration of damaged neonatal heart through glycolysis dependent YAP1 regulated miR-152 expression. Cell Death Differ. 2018, 25, 966–982. [Google Scholar] [CrossRef] [PubMed]
  85. Kaiser, W.J.; Sridharan, H.; Huang, C.; Mandal, P.; Upton, J.W.; Gough, P.J.; Sehon, C.A.; Marquis, R.W.; Bertin, J.; Mocarski, E.S. Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J. Biol. Chem. 2013, 288, 31268–31279. [Google Scholar] [CrossRef]
  86. Shao, L.; Zhang, P.; Zhang, Y.; Lu, Q.; Ma, A. TLR3 and TLR4 as potential clinically biomarkers of cardiovascular risk in coronary artery disease (CAD) patients. Heart Vessel. 2014, 29, 690–698. [Google Scholar] [CrossRef]
  87. Fattahi, F.; Russell, M.W.; Malan, E.A.; Parlett, M.; Abe, E.; Zetoune, F.S.; Ward, P.A. Harmful Roles of TLR3 and TLR9 in Cardiac Dysfunction Developing during Polymicrobial Sepsis. BioMed Res. Int. 2018, 2018, 4302726. [Google Scholar] [CrossRef] [PubMed]
  88. Saiyang, X.; Qingqing, W.; Man, X.; Chen, L.; Min, Z.; Yun, X.; Wenke, S.; Haiming, W.; Xiaofeng, Z.; Si, C.; et al. Activation of Toll-like receptor 7 provides cardioprotection in septic cardiomyopathy-induced systolic dysfunction. Clin. Transl. Med. 2021, 11, e266. [Google Scholar] [CrossRef] [PubMed]
  89. Feng, Y.; Chen, H.; Cai, J.; Zou, L.; Yan, D.; Xu, G.; Li, D.; Chao, W. Cardiac RNA induces inflammatory responses in cardiomyocytes and immune cells via Toll-like receptor 7 signaling. J. Biol. Chem. 2015, 290, 26688–26698. [Google Scholar] [CrossRef]
  90. Zhang, S.P.; Yang, R.H.; Shang, J.; Gao, T.; Wang, R.; Peng, X.D.; Miao, X.; Pan, L.; Yuan, W.J.; Lin, L. FOXC1 up-regulates the expression of toll-like receptors in myocardial ischaemia. J. Cell. Mol. Med. 2019, 23, 7566–7580. [Google Scholar] [CrossRef]
  91. Beg, A.A. Endogenous ligands of Toll-like receptors: Implications for regulating inflammatory and immune responses. Trends Immunol. 2002, 23, 509–512. [Google Scholar] [CrossRef]
  92. Bhattacharyya, S.; Varga, J. Emerging roles of innate immune signaling and toll-like receptors in fibrosis and systemic sclerosis. Curr. Rheumatol. Rep. 2015, 17, 2. [Google Scholar] [CrossRef] [PubMed]
  93. Akira, S.; Hoshino, K. Myeloid Differentiation Factor 88—Dependent and—Independent Pathways in Toll-Like Receptor Signaling. J. Infect. Dis. 2003, 187, S356–S363. [Google Scholar] [CrossRef]
  94. Barton, G.M.; Medzhitov, R. Toll-like receptor signaling pathways. Science 2003, 300, 1524–1525. [Google Scholar] [CrossRef] [PubMed]
  95. Gomez-Guerrero, C.; Mallavia, B.; Egido, J. Targeting Inflammation in Cardiovascular Diseases. Still a Neglected field? Cardiovasc. Ther. 2012, 30, e189–e197. [Google Scholar] [CrossRef]
  96. Stapel, H.; Kim, S.C.; Osterkamp, S.; Knuefermann, P.; Hoeft, A.; Meyer, R.; Grohe, C.; Baumgarten, G. Toll-like receptor 4 modulates myocardial ischaemia-reperfusion injury: Role of matrix metalloproteinases. Eur. J. Heart Fail. 2006, 8, 665–672. [Google Scholar] [CrossRef] [PubMed]
  97. Baumgarten, G.; Kim, S.C.; Stapel, H.; Vervolgyi, V.; Bittig, A.; Hoeft, A.; Meyer, R.; Grohe, C.; Knuefermann, P. Myocardial injury modulates the innate immune system and changes myocardial sensitivity. Basic. Res. Cardiol. 2006, 101, 427–435. [Google Scholar] [CrossRef] [PubMed]
  98. Ehrentraut, S.; Lohner, R.; Schwederski, M.; Ehrentraut, H.; Boehm, O.; Noga, S.; Langhoff, P.; Baumgarten, G.; Meyer, R.; Knuefermann, P. In Vivo Toll-Like Receptor 4 Antagonism Restores Cardiac Function During Endotoxemia. Shock 2011, 36, 613–620. [Google Scholar] [CrossRef] [PubMed]
  99. Fallach, R.; Shainberg, A.; Avlas, O.; Fainblut, M.; Chepurko, Y.; Porat, E.; Hochhauser, E. Cardiomyocyte Toll-like receptor 4 is involved in heart dysfunction following septic shock or myocardial ischemia. J. Mol. Cell. Cardiol. 2010, 48, 1236–1244. [Google Scholar] [CrossRef]
  100. Fang, Y.; Hu, J. Toll-like receptor and its roles in myocardial ischemic/reperfusion injury. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2011, 17, Ra100–Ra109. [Google Scholar] [CrossRef]
  101. Satoh, M.; Nakamura, M.; Akatsu, T.; Shimoda, Y.; Segawa, I.; Hiramori, K. Toll-like receptor 4 is expressed with enteroviral replication in myocardium from patients with dilated cardiomyopathy. Lab. Investig. A J. Tech. Methods Pathol. 2004, 84, 173–181. [Google Scholar] [CrossRef] [PubMed]
  102. Satoh, M.; Shimoda, Y.; Maesawa, C.; Akatsu, T.; Ishikawa, Y.; Minami, Y.; Hiramori, K.; Nakamura, M. Activated toll-like receptor 4 in monocytes is associated with heart failure after acute myocardial infarction. Int. J. Cardiol. 2006, 109, 226–234. [Google Scholar] [CrossRef]
  103. Tavener, S.A.; Long, E.M.; Robbins, S.M.; McRae, K.M.; Van Remmen, H.; Kubes, P. Immune cell Toll-like receptor 4 is required for cardiac myocyte impairment during endotoxemia. Circ. Res. 2004, 95, 700–707. [Google Scholar] [CrossRef] [PubMed]
  104. Sun, Y.; Liu, W.-Z.; Liu, T.; Feng, X.; Yang, N.; Zhou, H.-F. Role of toll-like receptors in myocardial infarction. J. Recept. Signal Transduct. 2015, 35, 420–422. [Google Scholar] [CrossRef] [PubMed]
  105. Huo, J.-Y.; Jiang, W.-Y.; Yin, T.; Xu, H.; Lyu, Y.-T.; Chen, Y.-Y.; Chen, M.; Geng, J.; Jiang, Z.-X.; Shan, Q.-J. Intestinal Barrier Dysfunction Exacerbates Neuroinflammation via the TLR4 Pathway in Mice With Heart Failure. Front. Physiol. 2021, 12, 1–10. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, X.; Xu, S.; Zhao, C.; Liu, B. Role of TLR4/NADPH oxidase 4 pathway in promoting cell death through autophagy and ferroptosis during heart failure. Biochem. Biophys. Res. Commun. 2019, 516, 37–43. [Google Scholar] [CrossRef]
  107. Monnerat-Cahli, G.; Alonso, H.; Gallego, M.; Alarcón, M.L.; Bassani, R.A.; Casis, O.; Medei, E. Toll-like receptor 4 activation promotes cardiac arrhythmias by decreasing the transient outward potassium current (Ito) through an IRF3-dependent and MyD88-independent pathway. J. Mol. Cell. Cardiol. 2014, 76, 116–125. [Google Scholar] [CrossRef]
  108. Geng, H.; Lu, H.; Zhang, L.; Zhang, H.; Zhou, L.; Wang, H.; Zhong, R. Increased expression of Toll like receptor 4 on peripheral-blood mononuclear cells in patients with coronary arteriosclerosis disease. Clin. Exp. Immunol. 2006, 143, 269–273. [Google Scholar] [CrossRef]
  109. Timmers, L.; Sluijter, J.P.G.; Keulen, J.K.v.; Hoefer, I.E.; Nederhoff, M.G.J.; Goumans, M.-J.; Doevendans, P.A.; Echteld, C.J.A.V.; Joles, J.A.; Quax, P.H.; et al. Toll-Like Receptor 4 Mediates Maladaptive Left Ventricular Remodeling and Impairs Cardiac Function After Myocardial Infarction. Circ. Res. 2008, 102, 257–264. [Google Scholar] [CrossRef]
  110. Methe, H.; Kim, J.-O.; Kofler, S.; Weis, M.; Nabauer, M.; Koglin, J. Expansion of circulating Toll-like receptor 4–positive monocytes in patients with acute coronary syndrome. Circulation 2005, 111, 2654–2661. [Google Scholar] [CrossRef]
  111. Gill, R.; Tsung, A.; Billiar, T. Linking oxidative stress to inflammation: Toll-like receptors. Free Radic. Biol. Med. 2010, 48, 1121–1132. [Google Scholar] [CrossRef]
  112. Wu, B.; Ni, H.; Li, J.; Zhuang, X.; Zhang, J.; Qi, Z.; Chen, Q.; Wen, Z.; Shi, H.; Luo, X. The impact of circulating mitochondrial DNA on cardiomyocyte apoptosis and myocardial injury after TLR4 activation in experimental autoimmune myocarditis. Cell. Physiol. Biochem. 2017, 42, 713–728. [Google Scholar] [CrossRef]
  113. Pannucci, P.; Jefferson, S.R.; Hampshire, J.; Cooper, S.L.; Hill, S.J.; Woolard, J. COVID-19-Induced Myocarditis: Pathophysiological Roles of ACE2 and Toll-like Receptors. Int. J. Mol. Sci. 2023, 24, 5374. [Google Scholar] [CrossRef]
  114. Xiao, Z.; Kong, B.; Yang, H.; Dai, C.; Fang, J.; Qin, T.; Huang, H. Key Player in Cardiac Hypertrophy, Emphasizing the Role of Toll-Like Receptor 4. Front. Cardiovasc. Med. 2020, 7, 579036. [Google Scholar] [CrossRef]
  115. Katare, P.B.; Bagul, P.K.; Dinda, A.K.; Banerjee, S.K. Toll-like receptor 4 inhibition improves oxidative stress and mitochondrial health in isoproterenol-induced cardiac hypertrophy in rats. Front. Immunol. 2017, 8, 719. [Google Scholar] [CrossRef] [PubMed]
  116. Katare, P.B.; Nizami, H.L.; Paramesha, B.; Dinda, A.K.; Banerjee, S.K. Activation of toll like receptor 4 (TLR4) promotes cardiomyocyte apoptosis through SIRT2 dependent p53 deacetylation. Sci. Rep. 2020, 10, 19232. [Google Scholar] [CrossRef] [PubMed]
  117. Ma, Y.; Zhang, X.; Bao, H.; Mi, S.; Cai, W.; Yan, H.; Wang, Q.; Wang, Z.; Yan, J.; Fan, G.; et al. Toll-Like Receptor (TLR) 2 and TLR4 Differentially Regulate Doxorubicin Induced Cardiomyopathy in Mice. PLoS ONE 2012, 7, e40763. [Google Scholar] [CrossRef]
  118. Liu, H.; Chu, S.; Wu, Z. Loss of toll-like receptor 4 ameliorates cardiovascular dysfunction in aged mice. Immun. Ageing 2021, 18, 42. [Google Scholar] [CrossRef]
  119. Lucas, K.; Maes, M. Role of the Toll Like Receptor (TLR) Radical Cycle in Chronic Inflammation: Possible Treatments Targeting the TLR4 Pathway. Mol. Neurobiol. 2013, 48, 190–204. [Google Scholar] [CrossRef]
  120. Dange, R.B.; Agarwal, D.; Masson, G.S.; Vila, J.; Wilson, B.; Nair, A.; Francis, J. Central blockade of TLR4 improves cardiac function and attenuates myocardial inflammation in angiotensin II-induced hypertension. Cardiovasc. Res. 2014, 103, 17–27. [Google Scholar] [CrossRef] [PubMed]
  121. Oyama, J.-i.; Blais Jr, C.; Liu, X.; Pu, M.; Kobzik, L.; Kelly, R.A.; Bourcier, T. Reduced myocardial ischemia-reperfusion injury in toll-like receptor 4-deficient mice. Circulation 2004, 109, 784–789. [Google Scholar] [CrossRef]
  122. Hu, N.; Zhang, Y. TLR4 knockout attenuated high fat diet-induced cardiac dysfunction via NF-κB/JNK-dependent activation of autophagy. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2017, 1863, 2001–2011. [Google Scholar] [CrossRef]
  123. Ge, C.; Zhao, Y.; Liang, Y.; He, Y. Silencing of TLR4 Inhibits Atrial Fibrosis and Susceptibility to Atrial Fibrillation via Downregulation of NLRP3-TGF-β in Spontaneously Hypertensive Rats. Dis. Markers 2022, 2022, 2466150. [Google Scholar] [CrossRef] [PubMed]
  124. Schlaak, R.A.; SenthilKumar, G.; Boerma, M.; Bergom, C. Advances in Preclinical Research Models of Radiation-Induced Cardiac Toxicity. Cancers 2020, 12, 415. [Google Scholar] [CrossRef]
  125. Lee, C.L.; Lee, J.W.; Daniel, A.R.; Holbrook, M.; Hasapis, S.; Wright, A.O.; Brownstein, J.; Da Silva Campos, L.; Ma, Y.; Mao, L.; et al. Characterization of cardiovascular injury in mice following partial-heart irradiation with clinically relevant dose and fractionation. Radiother. Oncol. 2021, 157, 155–162. [Google Scholar] [CrossRef] [PubMed]
  126. Dreyfuss, A.D.; Velalopoulou, A.; Avgousti, H.; Bell, B.I.; Verginadis, I.I. Preclinical models of radiation-induced cardiac toxicity: Potential mechanisms and biomarkers. Front. Oncol. 2022, 12, 920867. [Google Scholar] [CrossRef]
  127. Unthank, J.L.; Miller, S.J.; Quickery, A.K.; Ferguson, E.L.; Wang, M.; Sampson, C.H.; Chua, H.L.; DiStasi, M.R.; Feng, H.; Fisher, A.; et al. Delayed Effects of Acute Radiation Exposure in a Murine Model of the H-ARS: Multiple-Organ Injury Consequent to <10 Gy Total Body Irradiation. Health Phys. 2015, 109, 511–521. [Google Scholar] [CrossRef]
  128. Boerma, M.; Sridharan, V.; Krager, K.J.; Pawar, S.A. Small animal models of localized heart irradiation. Methods Cell. Biol. 2022, 168, 221–234. [Google Scholar] [CrossRef] [PubMed]
  129. Hanson, K.M.; Hernady, E.B.; Reed, C.K.; Johnston, C.J.; Groves, A.M.; Finkelstein, J.N. Apoptosis Resistance in Fibroblasts Precedes Progressive Scarring in Pulmonary Fibrosis and Is Partially Mediated by Toll-Like Receptor 4 Activation. Toxicol. Sci. Off. J. Soc. Toxicol. 2019, 170, 489–498. [Google Scholar] [CrossRef]
  130. Garcia, A.N.; Casanova, N.G.; Valera, D.G.; Sun, X.; Song, J.H.; Kempf, C.L.; Moreno-Vinasco, L.; Burns, K.; Bermudez, T.; Valdez, M.; et al. Involvement of eNAMPT/TLR4 signaling in murine radiation pneumonitis: Protection by eNAMPT neutralization. Transl. Res. J. Lab. Clin. Med. 2022, 239, 44–57. [Google Scholar] [CrossRef]
  131. Zheng, L.; Zhu, Q.; Xu, C.; Li, M.; Li, H.; Yi, P.Q.; Xu, F.F.; Cao, L.; Chen, J.Y. Glycyrrhizin mitigates radiation-induced acute lung injury by inhibiting the HMGB1/TLR4 signalling pathway. J. Cell. Mol. Med. 2020, 24, 214–226. [Google Scholar] [CrossRef]
  132. Zhi-Feng, W.; Le-Yuan, Z.; Xiao-Hui, Z.; Ya-Bo, G.; Jian-Ying, Z.; Yong, H.; Zhao-Chong, Z. TLR4-Dependent Immune Response Promotes Radiation-Induced Liver Disease by Changing the Liver Tissue Interstitial Microenvironment during Liver Cancer Radiotherapy. Radiat. Res. 2014, 182, 674–682. [Google Scholar] [CrossRef]
  133. Ji, L.; Hao, S.; Wang, J.; Zou, J.; Wang, Y. Roles of Toll-Like Receptors in Radiotherapy- and Chemotherapy-Induced Oral Mucositis: A Concise Review. Front. Cell. Infect. Microbiol. 2022, 12, 1–11. [Google Scholar] [CrossRef]
  134. Bai, J.; Wu, B.; Zhao, S.; Wang, G.; Su, S.; Lu, B.; Hu, Y.; Geng, Y.; Guo, Z.; Wan, J.; et al. The Effect of PD-1 Inhibitor Combined with Irradiation on HMGB1-Associated Inflammatory Cytokines and Myocardial Injury. J. Inflamm. Res. 2022, 15, 6357–6371. [Google Scholar] [CrossRef] [PubMed]
  135. Sridharan, V.; Johnson, K.A.; Landes, R.D.; Cao, M.; Singh, P.; Wagoner, G.; Hayar, A.; Sprick, E.D.; Eveld, K.A.; Bhattacharyya, A. Sex-dependent effects of genetic upregulation of activated protein C on delayed effects of acute radiation exposure in the mouse heart, small intestine, and skin. PLoS ONE 2021, 16, e0252142. [Google Scholar] [CrossRef] [PubMed]
  136. Sridharan, V.; Krager, K.; Pawar, S.A.; Bansal, S.; Li, Y.; Cheema, A.K.; Boerma, M. Effects of Whole and Partial Heart Irradiation on Collagen, Mast Cells, and Toll-like Receptor 4 in the Mouse Heart. Cancers 2023, 2, 406. [Google Scholar] [CrossRef]
  137. Schaue, D.; Micewicz, E.D.; Ratikan, J.A.; Xie, M.W.; Cheng, G.; McBride, W.H. Radiation and inflammation. Semin. Radiat. Oncol. 2015, 25, 4–10. [Google Scholar] [CrossRef] [PubMed]
  138. Ratikan, J.A.; Micewicz, E.D.; Xie, M.W.; Schaue, D. Radiation takes its Toll. Cancer Lett. 2015, 368, 238–245. [Google Scholar] [CrossRef]
  139. Klee, N.S.; McCarthy, C.G.; Martinez-Quinones, P.; Webb, R.C. Out of the frying pan and into the fire: Damage-associated molecular patterns and cardiovascular toxicity following cancer therapy. Ther. Adv. Cardiovasc. Dis. 2017, 11, 297–317. [Google Scholar] [CrossRef]
  140. Bolourani, S.; Brenner, M.; Wang, P. The interplay of DAMPs, TLR4, and proinflammatory cytokines in pulmonary fibrosis. J. Mol. Med. 2021, 99, 1373–1384. [Google Scholar] [CrossRef]
  141. Neta, R.; Oppenheim, J.J.; Schreiber, R.D.; Chizzonite, R.; Ledney, G.D.; MacVittie, T.J. Role of cytokines (interleukin 1, tumor necrosis factor, and transforming growth factor beta) in natural and lipopolysaccharide-enhanced radioresistance. J. Exp. Med. 1991, 173, 1177–1182. [Google Scholar] [CrossRef]
  142. Krivokrysenko, V.I.; Toshkov, I.A.; Gleiberman, A.S.; Krasnov, P.; Shyshynova, I.; Bespalov, I.; Maitra, R.K.; Narizhneva, N.V.; Singh, V.K.; Whitnall, M.H.; et al. The Toll-Like Receptor 5 Agonist Entolimod Mitigates Lethal Acute Radiation Syndrome in Non-Human Primates. PLoS ONE 2015, 10, e0135388. [Google Scholar] [CrossRef]
  143. Shakhov, A.N.; Singh, V.K.; Bone, F.; Cheney, A.; Kononov, Y.; Krasnov, P.; Bratanova-Toshkova, T.K.; Shakhova, V.V.; Young, J.; Weil, M.M.; et al. Prevention and Mitigation of Acute Radiation Syndrome in Mice by Synthetic Lipopeptide Agonists of Toll-Like Receptor 2 (TLR2). PLoS ONE 2012, 7, e33044. [Google Scholar] [CrossRef] [PubMed]
  144. Singh, V.K.; Ducey, E.J.; Fatanmi, O.O.; Singh, P.K.; Brown, D.S.; Purmal, A.; Shakhova, V.V.; Gudkov, A.V.; Feinstein, E.; Shakhov, A. CBLB613: A TLR 2/6 agonist, natural lipopeptide of Mycoplasma arginini, as a novel radiation countermeasure. Radiat. Res. 2012, 177, 628–642. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, Z.; Lei, X.; Li, X.; Cai, J.M.; Gao, F.; Yang, Y.Y. Toll-like receptors and radiation protection. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 31–39. [Google Scholar] [CrossRef] [PubMed]
  146. Feng, Z.; Xu, Q.; He, X.; Wang, Y.; Fang, L.; Zhao, J.; Cheng, Y.; Liu, C.; Du, J.; Cai, J. FG-4592 protects the intestine from irradiation-induced injury by targeting the TLR4 signaling pathway. Stem. Cell Res. Ther. 2022, 13, 271. [Google Scholar] [CrossRef]
  147. Xu, Y.; Chen, Y.; Liu, H.; Lei, X.; Guo, J.; Cao, K.; Liu, C.; Li, B.; Cai, J.; Ju, J.; et al. Heat-killed salmonella typhimurium (HKST) protects mice against radiation in TLR4-dependent manner. Oncotarget 2017, 8, 67082–67093. [Google Scholar] [CrossRef]
  148. Trofenciuc, N.-M.; Bordejevic, A.D.; Tomescu, M.C.; Petrescu, L.; Crisan, S.; Geavlete, O.; Mischie, A.; Onel, A.F.M.; Sasu, A.; Pop-Moldovan, A.L. Toll-like receptor 4 (TLR4) expression is correlated with T2* iron deposition in response to doxorubicin treatment: Cardiotoxicity risk assessment. Sci. Rep. 2020, 10, 17013. [Google Scholar] [CrossRef] [PubMed]
  149. Kole, T.P.; Aghayere, O.; Kwah, J.; Yorke, E.D.; Goodman, K.A. Comparison of heart and coronary artery doses associated with intensity-modulated radiotherapy versus three-dimensional conformal radiotherapy for distal esophageal cancer. Int. J. Radiat. Oncol. Biol. Phys. 2012, 83, 1580–1586. [Google Scholar] [CrossRef]
  150. Tillman, G.F.; Pawlicki, T.; Koong, A.C.; Goodman, K.A. Preoperative versus postoperative radiotherapy for locally advanced gastroesophageal junction and proximal gastric cancers: A comparison of normal tissue radiation doses. Dis. Esophagus 2008, 21, 437–444. [Google Scholar] [CrossRef]
  151. Wu, W.C.; Chan, C.L.; Wong, Y.W.; Cuijpers, J.P. A study on the influence of breathing phases in intensity-modulated radiotherapy of lung tumours using four-dimensional CT. Br. J. Radiol. 2010, 83, 252–256. [Google Scholar] [CrossRef]
Figure 1. Potential role for TLR4 as a therapeutic target to alleviate RIHD. Left: hypothesized role of the TLR4 pathway in the development of RIHD. Right: TLR4 inhibition may interfere with the events of adverse tissue remodeling in RIHD.
Figure 1. Potential role for TLR4 as a therapeutic target to alleviate RIHD. Left: hypothesized role of the TLR4 pathway in the development of RIHD. Right: TLR4 inhibition may interfere with the events of adverse tissue remodeling in RIHD.
Genes 14 01002 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gawali, B.; Sridharan, V.; Krager, K.J.; Boerma, M.; Pawar, S.A. TLR4—A Pertinent Player in Radiation-Induced Heart Disease? Genes 2023, 14, 1002. https://0-doi-org.brum.beds.ac.uk/10.3390/genes14051002

AMA Style

Gawali B, Sridharan V, Krager KJ, Boerma M, Pawar SA. TLR4—A Pertinent Player in Radiation-Induced Heart Disease? Genes. 2023; 14(5):1002. https://0-doi-org.brum.beds.ac.uk/10.3390/genes14051002

Chicago/Turabian Style

Gawali, Basveshwar, Vijayalakshmi Sridharan, Kimberly J. Krager, Marjan Boerma, and Snehalata A. Pawar. 2023. "TLR4—A Pertinent Player in Radiation-Induced Heart Disease?" Genes 14, no. 5: 1002. https://0-doi-org.brum.beds.ac.uk/10.3390/genes14051002

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

Article Metrics

Back to TopTop