Next Article in Journal
Environmental Assessment of Friable Asbestos from Soil to Air Using the Releasable Asbestos Sampler (RAS)
Next Article in Special Issue
Deteriorative Effects of Radiation Injury Combined with Skin Wounding in a Mouse Model
Previous Article in Journal
In Silico Prediction of the Toxicity of Nitroaromatic Compounds: Application of Ensemble Learning QSAR Approach
Previous Article in Special Issue
Radiation Attenuation Assessment of Serpentinite Rocks from a Geological Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Organ-Specific Endothelial Dysfunction Following Total Body Irradiation Exposure

by
Guru Prasad Sharma
1 and
Heather A. Himburg
1,2,*
1
Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
2
Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
*
Author to whom correspondence should be addressed.
Toxics 2022, 10(12), 747; https://doi.org/10.3390/toxics10120747
Submission received: 29 October 2022 / Revised: 28 November 2022 / Accepted: 30 November 2022 / Published: 1 December 2022
(This article belongs to the Special Issue Radiation Exposure and Health Effects)

Abstract

:
As the single cell lining of the heart and all blood vessels, the vascular endothelium serves a critical role in maintaining homeostasis via control of vascular tone, immune cell recruitment, and macromolecular transit. For victims of acute high-dose radiation exposure, damage to the vascular endothelium may exacerbate the pathogenesis of acute and delayed multi-organ radiation toxicities. While commonalities exist between radiation-induced endothelial dysfunction in radiosensitive organs, the vascular endothelium is known to be highly heterogeneous as it is required to serve tissue and organ specific roles. In keeping with its organ and tissue specific functionality, the molecular and cellular response of the endothelium to radiation injury varies by organ. Therefore, in the development of medical countermeasures for multi-organ injury, it is necessary to consider organ and tissue-specific endothelial responses to both injury and candidate mitigators. The purpose of this review is to summarize the pathogenesis of endothelial dysfunction following total or near total body irradiation exposure at the level of individual radiosensitive organs.

1. Introduction

Victims of whole or near whole body radiation exposure will experience multi-organ acute and late toxicities. This review focuses on the potential role of the vascular endothelium in the progression of organ specific sequelae following the type of high dose exposure that may be likely to occur in a catastrophic nuclear or radiological event [1]. While injury to the vascular endothelium is an important dose-limiting toxicity for radiotherapy (RT), these mechanisms have been reviewed elsewhere [2,3]. Additionally, whole or near whole body irradiation exposure introduces comorbidities that are likely to exacerbate tissue damage to each individual organ system [4]. For example, survivors of the atomic bomb are known to suffer from lifelong immune impairment that is likely to impact vascular function [5].
At the cellular level, ionizing radiation is known to induce immediate endothelial damage characterized by apoptotic cell death, the release of pro-inflammatory cytokines, expression of cellular adhesion molecules, and loss of endothelial barrier function [6,7]. Data from genetic endothelial gain and loss of function models suggest endothelial damage contributes to the severity of the acute radiation subsyndromes of the bone marrow [8,9] and gastrointestinal tract [10,11,12]. In addition to acute endothelial damage, ionizing radiation induces long-term functional changes in the endothelium which may contribute to the pathogenesis of a range of late morbidities including neurological, cardiopulmonary, gastrointestinal, and endocrine disorders. Here, we first review common pathological changes in the endothelium which may be relevant to both acute and late responding organs. We then focus on radiation injury at the tissue and organ level as the endothelium is known to be highly heterogeneous with tissue-specific differences in structure, phenotype, and function [13,14,15].

2. Radiation-Induced Endothelial Cell Injury

In general, ionizing radiation induces direct cellular injury via three mechanisms: induction of DNA damage, damage to the plasma membrane, and generation of reactive oxygen species (ROS) as illustrated in Figure 1. The formation of radiation-induced double stranded DNA breaks (DSBs) is known to increase linearly with radiation dose [16]. DNA damage is detected by the PI3K- related kinase (PIKK) family of kinases which includes ataxia-telangiectasia mutated (ATM) [17]. ATM then activates the DNA damage signaling pathway which includes activation of master transcription regulator p53. P53 regulates transcription of a number of downstream targets involved in cell growth arrest (p21), DNA repair, and the intrinsic apoptotic pathway [18]. P53-mediated induction of the intrinsic apoptotic pathway occurs in highly proliferative tissues such as the hematopoietic system, hair follicles, thymus, and intestinal epithelium, p53 [19].
In endothelial cells, radiation-induced endothelial p53 activation typically promotes p21-mediated cell cycle arrest and the development of a senescent cell phenotype [20]. Although viable, senescent cells are in permanent cell-cycle arrest and can be phenotypically characterized by increased beta-galactosidase activity (SA-B gal) [19]. Several models support radiation-induced senescence downstream of p53 activation in the endothelium. This was clearly evidenced in endothelial conditional p53 knockout models which showed loss of p53 led to an increase in endothelial radiosensitivity rather than resistance to apoptotic cell death [21]. This finding is also supported by in vitro data in pulmonary artery endothelial cells which showed accelerated cellular senescence is the primary cause of clonogenic cell death following in vitro radiation of pulmonary artery endothelial cells demonstrated by upregulation of p21, loss of SIRT1, and increased SA-B gal [22]. While the senescent endothelial cell is not capable of replication, the presence of senescent endothelial cells is thought to contribute to chronic inflammation and vascular dysfunction as senescent cells are known to exhibit inflammatory cytokines [23]. Consistent with this idea is the observation that radiation induces a concurrent increase in SA-B gal and monocyte adhesion [24].
In addition to DNA damage, ionizing radiation causes direct damage to the cell membrane causes direct damage to the cellular membrane by lipid peroxidation and fragmentation. Radiation induces hydrolysis of the membrane lipid sphingomyelin to ceramide by the membrane bound lipid enzyme acidic sphingomyelinase (ASMase) [10,25]. Ceramide accumulation in the membrane occurs within minutes of radiation injury [26] and has been shown to positively regulate both the extrinsic and intrinsic apoptotic pathways [27,28]. Early studies from the Kolesnick laboratory demonstrated sphingomyelin-ceramide mediated induced apoptosis is the primary mechanism for radiation-induced endothelial cell apoptosis. In these experiments, radiation induced the immediate conversion of sphingomyelin to ceramide which occurred independent of DNA damage [26]. Independence of the ceramide pathway from p53-mediated intrinsic apoptotic cell death was confirmed in genetic ASMase loss of function mouse models. In these studies, radiation-induced apoptosis of the endothelium was absent in the thymus, lung, and intestines of irradiated ASMase knockout mice [29,30,31].
Finally, radiation injury is known to induce the generation of ROS. Although ROS are generated within seconds following radiation injury [32], only a transient amount of ROS species are generated by the hydrolysis of water from ionizing radiation [33]. The majority of radiation-induced ROS species are thought to be generated by loss of mitochondrial integrity as cells lacking functional mitochondria do not produce ROS in response to radiation [34]. Radiation injury at lower doses (2 Gy in vitro exposure) has been shown to induce mitochondrial dysfunction without increasing intracellular ROS [35]. In this study, radiation reduced the number of endothelial mitochondria and altered mitochondrial structure [35]. Interestingly, pretreatment with the compound rosiglitazone which stimulates mitochondrial biogenesis and oxidative phosphorylation, protected mitochondria from radiation-induced injury and apoptotic death [35].

3. Radiation-Induced Endothelial Functional Alterations

Endothelial cells which escape both radiation-induced senescence or apoptotic cell death, may have long-term functional impairment as illustrated in Figure 2 [23]. In their comprehensive review of radiation-induced endothelial injury, Baselet et al. describes the activation of the endothelium to a pro-inflammatory state in the absence of typical in vivo endogenous factors such as TNF-a and IL-6 which are known to induce endothelial activation. Such “sterile inflammation” is characterized by the release of multiple pro-inflammatory cytokines (IL-1, IL-6, TNF-alpha) and the cell-surface expression of immune cell-recruiting adhesion molecules (VCAM, ICAM, CD44). The induction of this pro-inflammatory state is mediated by a combination of radiation-induced mechanisms which include: DNA damage, release of damage-associated molecular patterns (DAMPs), and generation of reactive oxygen species (ROS). Each of these mechanisms is known to promote signaling cascades which lead to activation of the genotoxic stress-induced nuclear factor (NF)-κB pathway [36]. The NF-κB signaling cascade induces the expression of adhesion molecules ICAM, VCAM, and E-selectin and release of inflammatory cytokines [37,38].
The expression of cell adhesion molecules on the endothelium is a key step in the initiation and progression of inflammatory cardiovascular diseases as these adhesion molecules promote leukocyte adhesion and transendothelial migration of neutrophils [7]. Recent data from Soroush et al. demonstrate the serine/threonine kinase PKCδ regulates the radiation-induced expression of adhesion molecules P-selectin, ICAM-1, and VCAM-1 [39,40]. Radiation induces a dose-dependent phosphorylation and activation of PKCδ in endothelial cells exposed to physiological levels of fluid shear stress in a 3D microfluidic chamber. Importantly inhibition of PKCδ reduces transcription of adhesion molecules and reduces neutrophil adhesion [39,40]. Translationally, preclinical data suggest peptide inhibitors of PKCδ (PKCδ-TAT) improve survival in mouse H-ARS models [7].
Another important characteristic of functional impairment in the irradiated endothelium is a loss of endothelial barrier function. As will be discussed later, endothelial permeability varies in an organic-specific manner. In general, the endothelium serves as a selective barrier which regulates the transport of fluids, solutes, macromolecules, and cells from the blood to the tissue space. Barrier integrity is maintained by endothelial cell-specific junctional proteins of the adherens, tight, and gap junctions [41]. Several labs have documented radiation-induced impairment of endothelial barrier function in vitro and in vivo [39,42,43,44,45]. Impairment of barrier integrity following radiation injury has been associated with dysregulation of the key adherens junctional proteins platelet endothelial cell adhesion molecule (PECAM-1) and vascular endothelial cadherin (VE-Cadherin) [44,45]. Kouam et al. demonstrate a dose-dependent increase in endothelial monolayer permeability which occurs concurrently with degradation of the full-length form of VE-Cadherin and dislocalization of VE-Cadherin on the cell surface. Moreover, they demonstrate loss of VE-Cadherin is mediated by the metalloproteinase ADAM10 (a disintegrin and metalloproteinase) [44]. Additionally, radiation-induced regulation of endothelial integrity is regulated by serine/threonine kinase PKCδ [39].
Finally, dysfunction of the endothelium is generally associated with impaired endothelial nitric oxide synthase (eNOS) activity. Here, “uncoupled” eNOS favors production of the ROS super oxide (O2-) and peroxynitrite (ONOO−) instead of the vasodilator nitric oxide (NO). Superoxide dismutase (SOD) mimetics which act to scavenge ROS have shown promise in mitigation of lung and intestinal radiation injury [46,47,48]. More recently, work by Rabender et al. demonstrated treatment with sepiapterin, a cofactor for the production of nitric oxide (tetrahydrobiopterin (BH4)), mitigates against late radiation-induced lung and heart injury in mice [49].

4. Hematologic Acute Radiation Syndrome (H-ARS)

The hematopoietic system is acutely sensitive to radiation exposure. In human populations, whole body radiation doses greater than 2 Gy may lead to fatality 2–8 weeks following exposure due to acute bone marrow toxicity and the resulting loss of mature blood cells and platelets. Hematopoietic function including regeneration is tightly regulated by the bone marrow (BM) microenvironment which includes the endothelial cells (ECs) commonly divided into two vascular niches: the arteriolar and sinusoidal vessels. The arteriolar and sinusoidal endothelial cells defining these distinct vascular niches [50] are known to play highly specialized roles in the regulation of hematopoietic stem cells within the BM as reviewed by Ramalingam et al. [51].
Physically, the sinusoidal endothelium forms a network of permeable capillaries which serve to traffic nutrients and hematopoietic cells to and from the BM microenvironment [52]. Early transmission electron micrograph studies demonstrated sinusoidal ECs experienced membrane damage which results in loss of endothelial barrier function [53]. Subsequent studies have shown dose and time dependent changes including dilation of sinusoidal vessels, loss of sinusoidal vessels, increased permeability, and alterations in the molecular profile of the surviving EC fraction [54,55,56,57]. Relative to non-hematopoietic tissues including the retina, heart, skin, and small intestine, Chen et al. demonstrated the bone marrow sinusoidal endothelium is more sensitive to radiation injury [56]. Interestingly, the increased sensitivity of BM ECs to radiation may be due to reliance on the hematopoietic compartment for necessary EC growth factors such as VEGF as genetic depletion of hematopoietic cell compartment phenocopies radiation-induced vascular injury. Finally, Chen et al. also showed regeneration of the bone marrow sinusoidal endothelium is regulated by a rare and relative radiation-resistant population of sinusoidal ECs expressing the marker Apelin [56].
Bone marrow endothelial cells (BMECs) play a key role in restoring hematopoiesis following radiation injury in part by secretion of hematopoietic cytokines such as G-CSF, EGF, pleiotrophin, jagged-1, CCL5 and E-selection [13,58,59,60,61,62,63]. Indeed, genetic gain and loss of function models have demonstrated the necessity of the BM vascular niche for hematopoietic regeneration following radiation injury. Genetic protection of the endogenous endothelium from intrinsic apoptotic cell death is sufficient to rescue mice from lethal radiation injury [9]. Genetic or pharmacological inhibition of the sinusoidal endothelial-specific receptor VEGFR2 inhibits both structural recovery of sinusoidal vessels and reduces hematopoietic reconstitution [8]. Additionally, genetic deletion of endothelial secreted hematopoietic cytokines jagged-1 or pleiotrophin inhibits hematopoietic stem cell regeneration following radiation injury [58,64].
Collectively, these studies emphasize the fundamental significance of the endothelium in regulating hematopoietic regeneration following radiation injury. Accordingly, several studies have evaluated whether therapeutic delivery of endothelial cells can improve hematopoietic regeneration after injury. Multiple groups have demonstrated that infusion of endothelial cells or endothelial cell-derived extracellular vesicles (EVs) can accelerate hematopoietic recovery and rescues lethally irradiated mice from death due to hematopoietic syndrome [50,65,66,67,68,69]. Additionally, in an in vitro comparison, endothelial cell co-culture promoted better regeneration of hematopoietic stem cells than treatment with the established medical countermeasure (MCM) G-CSF alone and was efficacious even when treatment was delayed to 48 h [70].
In addition to endothelial cellular therapies, therapies targeting the endogenous bone marrow endothelium have also been evaluated as potential medical countermeasures for hematopoietic acute radiation syndrome. Notably, activated protein C (APC) is a factor which counters radiation-induced loss of endothelial thrombomodulin to reverse pro-thrombotic and pro-fibrogenic remodeling of the endothelium [71]. Among other mitigative roles discussed later, treatment with APC has been shown to reduce radiation-induced hematopoietic toxicity [72,73]. While the vascular endothelium exerts many protective and regenerative roles, in the context of injury it can release factors such as semaphorin 3A (sema3A) which promote endothelial cell apoptosis [74]. Targeted inhibition of this pro-apoptotic mechanism with an antibody specifically blocking sema3A binding to the endothelial receptor neuropilin 1 promotes concomitant endothelial & hematopoietic recovery [74].

5. Gastrointestinal Acute Radiation Syndrome (GI-ARS)

Persons exposed to whole body doses as low as 1.5 Gy may experience radiation injury to the gastrointestinal (GI) tract as evidenced by early symptoms of nausea, vomiting, and anorexia. Without treatment, doses above 5 Gy may result in GI death due to denudation of the intestinal mucosal barrier and subsequent fluid loss, hemorrhage, and/or sepsis [75,76]. The denudation of intestinal mucosal barrier results from an acute loss of the intestinal stem cell population which resides in the intestinal crypts and functions to replenish the epithelial layer [77]. Within the intestinal villi, intestinal endothelial cells form a dense vascular network within the gut epithelium. The intestinal vasculature plays a unique role as it serves as a selective permeability barrier (the gut vascular barrier or GVB) which allows the necessary absorption of nutrients while blocking the translocation of bacteria and large macromolecules [78].
In the setting of acute intestinal injury, it has been debated whether GI toxicity results from initiation of apoptotic death in the endothelium or the intestinal crypt [10,79]. While the relative contribution of the endothelium to the initiation of injury remains unclear, it is evident the intestinal endothelium plays a key role in the pathogenesis of acute GI injury as genetic protection of the intestinal endothelium against ceramide-mediated apoptotic cell death via the deletion of the acid sphingomyelinase gene [10] is protective against lethal GI toxicity. Subsequent studies have demonstrated this mechanism is critical to survival following lethal GI injury as pharmacologic neutralization of endothelial ceramide signaling promotes intestinal stem cell survival and effectively mitigates radiation-induced GI toxicity [11,12].

6. Late Responding Organs

Survivors of high-dose acute radiation exposure are at increased risk for developing a range of late multi-organ morbidities which are collectively referred to as the delayed effects of acute radiation exposure (DEARE). Sustained vascular dysfunction is a common trait which has been observed in multiple late radiation-sensitive tissues including the lung [80,81,82,83,84,85], kidneys [86,87], heart [88,89,90,91,92], brain [93], and gastrointestinal tract [71]. Late vascular dysfunction is characterized by structural changes in the vasculature including malformities [93], and vessel regression [81,82]. In a cohort of non-human primate (NHP) survivors of total body irradiation doses between 1.1–8.5 Gy at Wake Forest University, the incidence of vascular brain lesions was non-invasively observed with MR imaging for more than 10 years post-irradiation. While most of the cohort exhibited early brain lesions (12/16 NHPs), there was a notable increase in the number of new lesions during surveillance [93]. Histopathological analysis of deceased animals demonstrated MRI lesions were correlated with abnormalities of the cerebral vasculature and remodeling of the vascular wall [93]. Together, these data suggest that survivors of acute radiation exposure are at elevated risk of developing vascular injury for many years following exposure.

7. Radiation Enteropathy

In addition to life-threatening acute GI syndrome, survivors of acute radiation exposure may experience radiation enteropathy a late-radiation toxicity characterized by mucosal loss, fibrosis, and sustained vascular dysfunction [71]. The mechanism which initiates late injury is thought to be distinct from acute toxicity as early epithelial damage does not necessarily correlate with late intestinal fibrosis in animal models [94]. Recently, Lee et al. demonstrated that while endothelial deletion of p53 had no effect on the acute GI syndrome, loss of endothelial p53 resulted in an increased susceptibility to late injury characterized by increased vascular permeability, tissue hypoxia, and reduced intestinal length [95]. A key mechanism regulating vascular dysfunction in radiation enteropathy is the thrombomodulin-protein C system. In the intestinal microvasculature, endothelial thrombomodulin production is markedly reduced following radiation injury [96,97]. The radiation-induced deficiency of thrombomodulin both increases levels of thrombin and reduces activated protein C (APC) [71]. The sustained dysregulation of this signaling pathway leads to increased blood clotting, inflammation, and pro-fibrotic collagen production [71]. For this reason, pharmacologic studies to target these pathways have been evaluated and suggest either recombinant thrombomodulin or therapies which promote thrombomodulin expression can mitigate intestinal injury in a rat model [98,99]. Interestingly, although APC treatment has established mitigative effects for acute injury [73], long-term genetic upregulation of APC had little effect on the progression of late vascular injury [100].

8. Radiation-Induced Lung Injury

Radiation-induced lung injury (RILI) can occur in two phases: an acute inflammatory stage (pneumonitis) 8–16 weeks following exposure and a late fibrotic injury which can take months to years to evolve [101]. These are two distinct mechanisms of injury as fibrosis can occur independently of overt pneumonitis [102]. However, endothelial dysfunction is common to both injury phases and contributes to the pathogenesis of both pneumonitis and fibrosis. The lung microvascular ECs are highly specialized and exist in close proximity to the alveolar epithelium to facilitate gas exchange between the circulation and alveoli.
Radiation pneumonitis is primarily an inflammatory condition characterized by pulmonary edema, immune cell infiltration, and clinical symptoms of labored breathing/cough. Animal models have been instructive in characterizing the acute vascular changes during pneumonitis. At one month following 10 Gy thoracic irradiation, marked changes in the pulmonary vasculature have been observed including a loss of the pulmonary vessels and increased vascular resistance [82]. Concurrent with the loss in vasculature, increased vascular permeability is observed in the lung of irradiated rats at 6 weeks following high dose radiation exposure [43]. Marked improvement in vascular function and survival during pneumonitis have been observed in rats treated with the angiotensin converting enzyme (ACE) inhibitor lisinopril [43,103]. While this effect may be mediated in part by the well-established vasodilatory function of lisinopril, recent data also suggest ACE inhibition suppresses immune-mediated production of inflammatory cytokines [104].
Pathologic alterations in vascular permeability during pneumonitis are attributable in part to alteration of lung endothelial sphingolipids [105]. Similar to intestinal injury, radiation induces an increase in the pro-apoptotic sphingolipid ceramide relative to the barrier protective sphingolipid sphingosine 1-phosphate (S1P) in lung endothelial cells [106]. Consistent with this observation, genetic deletion of S1P results in exacerbation of radiation-induced loss of pulmonary endothelial barrier function and treatment with S1P analogs prior to radiation exposure protected against loss of barrier function [106]. Finally, it has been shown that treatment with soy isoflavones may mediate clearance of damaged, pro-inflammatory lung endothelial cells immune cells which express adhesion molecules and release the DAMPs including HMGB1 [107].

9. Heart Disease

Schultz-Hector and Trott provided a comprehensive review of the manifestation of cardiovascular diseases in both clinical cohorts exposed to heart irradiation and epidemiologic analysis of the Life Span Study of Japanese atomic bomb survivors [108]. This review highlighted the increased risk for development of a range of cardiovascular diseases including ischemic heart disease, arteriosclerosis, and cardiomyopathy following radiation exposure [108]. Atomic bomb survivors have been shown to have increased circulating levels of pro-inflammatory cytokines IL-6 and C-reactive protein (CRP) [109] which is suggestive of sustained and systemic endothelial dysfunction. In a long-term analysis of the Life Span Study cohort, doses above 0.5 Gy were associated with an elevated risk of both heart disease and stroke [110]. A higher incidence of cerebrovascular disease was also observed in the Mayak Production Association cohort comprised of 18,797 workers exposed at the Mayak nuclear facility [111].

10. Radiation Nephropathy

Survivors of acute radiation syndrome are at increased risk for developing late kidney injury or radiation nephropathy. In a cohort analysis of Japanese atomic bomb survivors, radiation dose was significantly associated with chronic kidney disease [112]. The glomerular endothelium has been identified as the primary site of radiation damage in the kidney [113]. The glomerular endothelium is another highly specialized endothelial cell type which functions to filter massive volumes of plasma through transcellular pores (fenestrae) [114]. Radiation injury induces structural changes to glomerular endothelium including swelling and separation from the basement membrane [113,115]. Functionally, this results in a decrease in glomerular filtration rate and a progressive decline in renal function. In a rat model, vascular regression in the kidney was associated with a loss of Notch ligand, jagged1. Treatment with the ACE inhibitor lisinopril has been observed to both improve renal function following radiation injury in rat models and normalize renal jagged1 expression [87].

11. Conclusions

Radiation-induced EC injury is a major mediator and regulator of wider tissue damage in the different vascular beds. The data reviewed here highlight the role of the highly heterogeneous endothelium in the progression of multi-organ injury. While the endothelium of radiosensitive organs is highly specialized, there may be common mechanisms which can be targeted to systemically promote normalization of the endothelium post radiation injury. Several aforementioned therapies (summarized in Table 1 below) include peptide inhibitors of PKCδ, exogenous endothelial cell products, ACE inhibitors, factors targeting the thrombomodulin-activated protein C pathway, anti-ceramide antibodies, S1P analogs, SOD mimetics, and sepiapterin.

Author Contributions

Writing—original draft preparation, G.P.S. and H.A.H.; Writing—review and editing, G.P.S. and H.A.H. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by NIH/NIAID grants U01AI138331 and U01AI33594 to H.A.H.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Satyamitra, M.M.; DiCarlo, A.L.; Taliaferro, L. Understanding the Pathophysiology and Challenges of Development of Medical Countermeasures for Radiation-Induced Vascular/Endothelial Cell Injuries: Report of a NIAID Workshop, August 20, 2015. Radiat. Res. 2016, 186, 99–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. 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] [PubMed]
  3. Belzile-Dugas, E.; Eisenberg, M.J. Radiation-Induced Cardiovascular Disease: Review of an Underrecognized Pathology. J. Am. Heart Assoc. 2021, 10, e021686. [Google Scholar] [CrossRef] [PubMed]
  4. MacVittie, T.J.; Farese, A.M.; Parker, G.A.; Bennett, A.W.; Jackson, W.E., 3rd. Acute Radiation-induced Lung Injury in the Non-human Primate: A Review and Comparison of Mortality and Co-morbidities Using Models of Partial-body Irradiation with Marginal Bone Marrow Sparing and Whole Thorax Lung Irradiation. Health Phys. 2020, 119, 559–587. [Google Scholar] [CrossRef]
  5. Kusunoki, Y.; Hayashi, T. Long-lasting alterations of the immune system by ionizing radiation exposure: Implications for disease development among atomic bomb survivors. Int. J. Radiat. Biol. 2008, 84, 1–14. [Google Scholar] [CrossRef]
  6. 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] [Green Version]
  7. Wijerathne, H.; Langston, J.C.; Yang, Q.; Sun, S.; Miyamoto, C.; Kilpatrick, L.E.; Kiani, M.F. Mechanisms of radiation-induced endothelium damage: Emerging models and technologies. Radiother. Oncol. 2021, 158, 21–32. [Google Scholar] [CrossRef]
  8. Hooper, A.T.; Butler, J.M.; Nolan, D.J.; Kranz, A.; Iida, K.; Kobayashi, M.; Kopp, H.-G.; Shido, K.; Petit, I.; Yanger, K.; et al. Engraftment and Reconstitution of Hematopoiesis Is Dependent on VEGFR2-Mediated Regeneration of Sinusoidal Endothelial Cells. Cell Stem Cell 2009, 4, 263–274. [Google Scholar] [CrossRef] [Green Version]
  9. Doan, P.L.; Russell, J.L.; Himburg, H.A.; Helms, K.; Harris, J.R.; Lucas, J.; Holshausen, K.C.; Meadows, S.K.; Daher, P.; Jeffords, L.B.; et al. Tie2+ Bone Marrow Endothelial Cells Regulate Hematopoietic Stem Cell Regeneration Following Radiation Injury. Stem Cells 2013, 31, 327–337. [Google Scholar] [CrossRef] [Green Version]
  10. Paris, F.; Fuks, Z.; Kang, A.; Capodieci, P.; Juan, G.; Ehleiter, D.; Haimovitz-Friedman, A.; Cordon-Cardo, C.; Kolesnick, R. Endothelial Apoptosis as the Primary Lesion Initiating Intestinal Radiation Damage in Mice. Science 2001, 293, 293–297. [Google Scholar] [CrossRef]
  11. Rotolo, J.A.; Fong, C.S.; Bodo, S.; Nagesh, P.K.; Fuller, J.; Sharma, T.; Piersigilli, A.; Zhang, Z.; Fuks, Z.; Singh, V.K.; et al. Anti-ceramide single-chain variable fragment mitigates radiation GI syndrome mortality independent of DNA repair. JCI Insight 2021, 6, e145380. [Google Scholar] [CrossRef]
  12. Rotolo, J.; Stancevic, B.; Zhang, J.; Hua, G.; Fuller, J.; Yin, X.; Haimovitz-Friedman, A.; Kim, K.; Qian, M.; Cardó-Vila, M.; et al. Anti-ceramide antibody prevents the radiation gastrointestinal syndrome in mice. J. Clin. Investig. 2012, 122, 1786–1790. [Google Scholar] [CrossRef] [Green Version]
  13. Rafii, S.; Butler, J.M.; Ding, B.-S. Angiocrine functions of organ-specific endothelial cells. Nature 2016, 529, 316–325. [Google Scholar] [CrossRef] [Green Version]
  14. Nolan, D.J.; Ginsberg, M.; Israely, E.; Palikuqi, B.; Poulos, M.G.; James, D.; Ding, B.-S.; Schachterle, W.; Liu, Y.; Rosenwaks, Z.; et al. Molecular Signatures of Tissue-Specific Microvascular Endothelial Cell Heterogeneity in Organ Maintenance and Regeneration. Dev. Cell 2013, 26, 204–219. [Google Scholar] [CrossRef] [Green Version]
  15. Aird, W.C. Endothelial Cell Heterogeneity. Cold Spring Harb. Perspect. Med. 2012, 2, a006429. [Google Scholar] [CrossRef] [Green Version]
  16. Vilenchik, M.M.; Knudson, A.G. Endogenous DNA double-strand breaks: Production, fidelity of repair, and induction of cancer. Proc. Natl. Acad. Sci. USA 2003, 100, 12871–12876. [Google Scholar] [CrossRef] [Green Version]
  17. Bakkenist, C.J.; Kastan, M.B. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 2003, 421, 499–506. [Google Scholar] [CrossRef]
  18. Eriksson, D.; Stigbrand, T. Radiation-induced cell death mechanisms. Tumour Biol. 2010, 31, 363–372. [Google Scholar] [CrossRef]
  19. Jackson, J.G.; Post, S.M.; Lozano, G. Regulation of tissue- and stimulus-specific cell fate decisions by p53 in vivo. J. Pathol. 2011, 223, 127–137. [Google Scholar] [CrossRef]
  20. Lee, C.-L.; Blum, J.M.; Kirsch, D.G. Role of p53 in regulating tissue response to radiation by mechanisms independent of apoptosis. Transl. Cancer Res. 2013, 2, 412–421. [Google Scholar] [PubMed]
  21. Lee, C.-L.; Moding, E.J.; Cuneo, K.C.; Li, Y.; Sullivan, J.M.; Mao, L.; Washington, I.; Jeffords, L.B.; Rodrigues, R.C.; Ma, Y.; et al. p53 Functions in Endothelial Cells to Prevent Radiation-Induced Myocardial Injury in Mice. Sci. Signal. 2012, 5, ra52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Panganiban, R.A.M.; Mungunsukh, O.; Day, R.M. X-irradiation induces ER stress, apoptosis, and senescence in pulmonary artery endothelial cells. Int. J. Radiat. Biol. 2013, 89, 656–667. [Google Scholar] [CrossRef] [PubMed]
  23. Nagane, M.; Yasui, H.; Kuppusamy, P.; Yamashita, T.; Inanami, O. DNA damage response in vascular endothelial senescence: Implication for radiation-induced cardiovascular diseases. J. Radiat. Res. 2021, 62, 564–573. [Google Scholar] [CrossRef]
  24. Lowe, D.; Raj, K. Premature aging induced by radiation exhibits pro-atherosclerotic effects mediated by epigenetic activation ofCD44 expression. Aging Cell 2014, 13, 900–910. [Google Scholar] [CrossRef]
  25. Rodemann, H.P.; Blaese, M.A. Responses of Normal Cells to Ionizing Radiation. Semin. Radiat. Oncol. 2007, 17, 81–88. [Google Scholar] [CrossRef]
  26. Haimovitz-Friedman, A.; Kan, C.C.; Ehleiter, D.; Persaud, R.S.; McLoughlin, M.; Fuks, Z.; Kolesnick, R.N. Ionizing radiation acts on cellular membranes to generate ceramide and initiate apoptosis. J. Exp. Med. 1994, 180, 525–535. [Google Scholar] [CrossRef] [Green Version]
  27. Oskouian, B.; Saba, J.D. Cancer Treatment Strategies Targeting Sphingolipid Metabolism. Adv. Exp. Med. Biol. 2010, 688, 185–205. [Google Scholar] [CrossRef] [Green Version]
  28. Kolesnick, R.; Fuks, Z. Radiation and ceramide-induced apoptosis. Oncogene 2003, 22, 5897–5906. [Google Scholar] [CrossRef] [Green Version]
  29. Santana, P.; Peña, L.A.; Haimovitz-Friedman, A.; Martin, S.; Green, D.; McLoughlin, M.; Cordon-Cardo, C.; Schuchman, E.H.; Fuks, Z.; Kolesnick, R. Acid Sphingomyelinase–Deficient Human Lymphoblasts and Mice Are Defective in Radiation-Induced Apoptosis. Cell 1996, 86, 189–199. [Google Scholar] [CrossRef] [Green Version]
  30. Peña, L.A.; Fuks, Z.; Kolesnick, R.N. Radiation-induced apoptosis of endothelial cells in the murine central nervous system: Protection by fibroblast growth factor and sphingomyelinase deficiency. Cancer Res. 2000, 60, 321–327. [Google Scholar]
  31. Paris, F.; Grassmé, H.; Cremesti, A.; Zager, J.; Fong, Y.; Haimovitz-Friedman, A.; Fuks, Z.; Gulbins, E.; Kolesnick, R. Natural Ceramide Reverses Fas Resistance of Acid Sphingomyelinase (-/-)Hepatocytes. J. Biol. Chem. 2001, 276, 8297–8305. [Google Scholar] [CrossRef] [PubMed]
  32. Mansilla, S.; Bataller, M. Mechanisms of Drug-Induced Mitotic Catastrophe in Cancer Cells. Curr. Pharm. Des. 2010, 16, 69–78. [Google Scholar] [CrossRef]
  33. Martins, I.; Galluzzi, L.; Kroemer, G. Hormesis, cell death and aging. Aging 2011, 3, 821–828. [Google Scholar] [CrossRef] [Green Version]
  34. Leach, J.K.; Van Tuyle, G.; Lin, P.S.; Schmidt-Ullrich, R.; Mikkelsen, R.B. Ionizing radiation-induced, mitochondria-dependent generation of reactive oxygen/nitrogen. Cancer Res 2001, 61, 3894–3901. [Google Scholar]
  35. Baselet, B.; Driesen, R.B.; Coninx, E.; Belmans, N.; Sieprath, T.; Lambrichts, I.; De Vos, W.H.; Baatout, S.; Sonveaux, P.; Aerts, A. Rosiglitazone Protects Endothelial Cells from Irradiation-Induced Mitochondrial Dysfunction. Front. Pharmacol. 2020, 11, 268. [Google Scholar] [CrossRef] [Green Version]
  36. Hellweg, C.E. The Nuclear Factor κB pathway: A link to the immune system in the radiation response. Cancer Lett. 2015, 368, 275–289. [Google Scholar] [CrossRef]
  37. Goebeler, M.; Gillitzer, R.; Kilian, K.; Utzel, K.; Bröcker, E.-B.; Rapp, U.R.; Ludwig, S. Multiple signaling pathways regulate NF-κB–dependent transcription of the monocyte chemoattractant protein-1 gene in primary endothelial cells. Blood J. Am. Soc. Hematol. 2001, 97, 46–55. [Google Scholar] [CrossRef] [Green Version]
  38. Kim, I.; Moon, S.-O.; Kim, S.H.; Kim, H.J.; Koh, Y.S.; Koh, G.Y. Vascular Endothelial Growth Factor Expression of Intercellular Adhesion Molecule 1 (ICAM-1), Vascular Cell Adhesion Molecule 1 (VCAM-1), and E-selectin through Nuclear Factor-κB Activation in Endothelial Cells. J. Biol. Chem. 2001, 276, 7614–7620. [Google Scholar] [CrossRef] [Green Version]
  39. Soroush, F.; Tang, Y.; Zaidi, H.M.; Sheffield, J.B.; Kilpatrick, L.E.; Kiani, M.F. PKCδ inhibition as a novel medical countermeasure for radiation-induced vascular damage. FASEB J. 2018, 32, fj201701099. [Google Scholar] [CrossRef]
  40. Soroush, F.; Zhang, T.; King, D.J.; Tang, Y.; Deosarkar, S.; Prabhakarpandian, B.; Kilpatrick, L.E.; Kiani, M.F. A novel microfluidic assay reveals a key role for protein kinase C δ in regulating human neutrophil–endothelium interaction. J. Leukoc. Biol. 2016, 100, 1027–1035. [Google Scholar] [CrossRef] [Green Version]
  41. Claesson-Welsh, L.; Dejana, E.; McDonald, D.M. Permeability of the Endothelial Barrier: Identifying and Reconciling Controversies. Trends Mol. Med. 2020, 27, 314–331. [Google Scholar] [CrossRef] [PubMed]
  42. Sharma, G.P.; Frei, A.C.; Narayanan, J.; Gasperetti, T.; Veley, D.; Amjad, A.; Albano, K.; Fish, B.L.; Himburg, H.A. Brain-derived neurotrophic factor promotes immune reconstitution following radiation injury via activation of bone marrow mesenchymal stem cells. PLoS ONE 2021, 16, e0259042. [Google Scholar] [CrossRef]
  43. Jagtap, J.; Audi, S.; Razeghi-Kondelaji, M.H.; Fish, B.L.; Hansen, C.; Narayan, J.; Gao, F.; Sharma, G.; Parchur, A.K.; Banerjee, A.; et al. A rapid dynamic in vivo near-infrared fluorescence imaging assay to track lung vascular permeability after acute radiation injury. Am. J. Physiol. Cell. Mol. Physiol. 2021, 320, L436–L450. [Google Scholar] [CrossRef]
  44. Kouam, P.N.; Rezniczek, G.A.; Adamietz, I.A.; Bühler, H. Ionizing radiation increases the endothelial permeability and the transendothelial migration of tumor cells through ADAM10-activation and subsequent degradation of VE-cadherin. BMC Cancer 2019, 19, 958. [Google Scholar] [CrossRef] [Green Version]
  45. Sharma, P.; Templin, T.; Grabham, P. Short term effects of gamma radiation on endothelial barrier function: Uncoupling of PECAM-1. Microvasc. Res. 2013, 86, 11–20. [Google Scholar] [CrossRef]
  46. Otterson, M.F.; Nie, L.; Schmidt, J.L.; Link, B.J.; Jovanovic, N.; Lyros, O.; Rafiee, P. EUK-207 protects human intestinal microvascular endothelial cells (HIMEC) against irradiation-induced apoptosis through the Bcl2 pathway. Life Sci. 2012, 91, 771–782. [Google Scholar] [CrossRef] [Green Version]
  47. Mahmood, J.; Jelveh, S.; Calveley, V.; Zaidi, A.; Doctrow, S.R.; Hill, R.P. Mitigation of radiation-induced lung injury by genistein and EUK-207. Int. J. Radiat. Biol. 2011, 87, 889–901. [Google Scholar] [CrossRef] [Green Version]
  48. Gao, F.; Fish, B.L.; Szabo, A.; Doctrow, S.R.; Kma, L.; Molthen, R.C.; Moulder, J.E.; Jacobs, E.R.; Medhora, M. Short-Term Treatment with a SOD/Catalase Mimetic, EUK-207, Mitigates Pneumonitis and Fibrosis after Single-Dose Total-Body or Whole-Thoracic Irradiation. Radiat. Res. 2012, 178, 468–480. [Google Scholar] [CrossRef]
  49. Rabender, C.S.; Mezzaroma, E.; Yakovlev, V.A.; Mauro, A.G.; Bonaventura, A.; Abbate, A.; Mikkelsen, R.B. Mitigation of Radiation-Induced Lung and Heart Injuries in Mice by Oral Sepiapterin after Irradiation. Radiat. Res. 2021, 195, 463–473. [Google Scholar] [CrossRef]
  50. Itkin, T.; Gur-Cohen, S.; Spencer, J.A.; Schajnovitz, A.; Ramasamy, S.K.; Kusumbe, A.P.; Ledergor, G.; Jung, Y.; Milo, I.; Poulos, M.G.; et al. Distinct bone marrow blood vessels differentially regulate haematopoiesis. Nature 2016, 532, 323–328. [Google Scholar] [CrossRef]
  51. Ramalingam, P.; Butler, J.M.; Poulos, M.G. Vascular Regulation of Hematopoietic Stem Cell Homeostasis, Regeneration, and Aging. Curr. Stem Cell Rep. 2021, 7, 194–203. [Google Scholar] [CrossRef] [PubMed]
  52. Kunisaki, Y.; Frenette, P.S. Influences of vascular niches on hematopoietic stem cell fate. Int. J. Hematol. 2014, 99, 699–705. [Google Scholar] [CrossRef] [PubMed]
  53. Shirota, T.; Tavassoli, M. Alterations of bone marrow sinus endothelium induced by ionizing irradiation: Implications in the homing of intravenously transplanted marrow cells. Blood cells 1992, 18, 197–214. [Google Scholar]
  54. Li, X.-M.; Hu, Z.; Jorgenson, M.L.; Wingard, J.R.; Slayton, W.B. Bone marrow sinusoidal endothelial cells undergo nonapoptotic cell death and are replaced by proliferating sinusoidal cells in situ to maintain the vascular niche following lethal irradiation. Exp. Hematol. 2008, 36, 1143–1156.e3. [Google Scholar] [CrossRef]
  55. Huang, W.; Yang, Y.; Sun, Z.; Zeng, X. Early Radiation-induced Bone Marrow Injury: Serial MR Imaging During Initial 4 Weeks After Irradiation. Acad. Radiol. 2009, 16, 733–738. [Google Scholar] [CrossRef]
  56. Chen, Q.; Liu, Y.; Jeong, H.-W.; Stehling, M.; Dinh, V.V.; Zhou, B.; Adams, R.H. Apelin+ Endothelial Niche Cells Control Hematopoiesis and Mediate Vascular Regeneration after Myeloablative Injury. Cell Stem Cell 2019, 25, 768–783.e6. [Google Scholar] [CrossRef] [Green Version]
  57. Cary, L.; Noutai, D.; Salber, R.; Fadiyimu, O.; Gross, A.; Almeida-Porada, G.; Kidane, Y.; Whitnall, M. Bone Marrow Endothelial Cells Influence Function and Phenotype of Hematopoietic Stem and Progenitor Cells after Mixed Neutron/Gamma Radiation. Int. J. Mol. Sci. 2019, 20, 1795. [Google Scholar] [CrossRef] [Green Version]
  58. Poulos, M.G.; Guo, P.; Kofler, N.M.; Pinho, S.; Gutkin, M.C.; Tikhonova, A.; Aifantis, I.; Frenette, P.S.; Kitajewski, J.; Rafii, S.; et al. Endothelial Jagged-1 Is Necessary for Homeostatic and Regenerative Hematopoiesis. Cell Rep. 2013, 4, 1022–1034. [Google Scholar] [CrossRef] [Green Version]
  59. Himburg, H.A.; Muramoto, G.G.; Daher, P.; Meadows, S.K.; Russell, J.L.; Doan, P.; Chi, J.-T.; Salter, A.B.; Lento, W.E.; Reya, T.; et al. Pleiotrophin regulates the expansion and regeneration of hematopoietic stem cells. Nat. Med. 2010, 16, 475–482. [Google Scholar] [CrossRef]
  60. Himburg, H.A.; Yan, X.; Doan, P.L.; Quarmyne, M.; Micewicz, E.; McBride, W.; Chao, N.J.; Slamon, D.J.; Chute, J.P. Pleiotrophin mediates hematopoietic regeneration via activation of RAS. J. Clin. Investig. 2014, 124, 4753–4758. [Google Scholar] [CrossRef] [Green Version]
  61. Doan, P.L.; Himburg, H.A.; Helms, K.; Russell, J.L.; Fixsen, E.; Quarmyne, M.; Harris, J.R.; Deoliviera, D.; Sullivan, J.M.; Chao, N.J.; et al. Epidermal growth factor regulates hematopoietic regeneration after radiation injury. Nat. Med. 2013, 19, 295–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Piryani, S.O.; Kam, A.Y.F.; Vu, U.T.; Chao, N.J.; Doan, P.L. CCR5 Signaling Promotes Murine and Human Hematopoietic Regeneration following Ionizing Radiation. Stem Cell Rep. 2019, 13, 76–90. [Google Scholar] [CrossRef] [Green Version]
  63. Winkler, I.G.; Barbier, V.; Nowlan, B.; Jacobsen, R.N.; Forristal, C.E.; Patton, J.T.; Magnani, J.L.; Lévesque, J.-P. Vascular niche E-selectin regulates hematopoietic stem cell dormancy, self renewal and chemoresistance. Nat. Med. 2012, 18, 1651–1657. [Google Scholar] [CrossRef]
  64. Himburg, H.A.; Termini, C.M.; Schlussel, L.; Kan, J.; Li, M.; Zhao, L.; Fang, T.; Sasine, J.P.; Chang, V.Y.; Chute, J.P. Distinct Bone Marrow Sources of Pleiotrophin Control Hematopoietic Stem Cell Maintenance and Regeneration. Cell Stem Cell 2018, 23, 370–381.e5. [Google Scholar] [CrossRef] [Green Version]
  65. Chute, J.P.; Muramoto, G.G.; Salter, A.B.; Meadows, S.K.; Rickman, D.W.; Chen, B.; Himburg, H.A.; Chao, N.J. Transplantation of vascular endothelial cells mediates the hematopoietic recovery and survival of lethally irradiated mice. Blood 2007, 109, 2365–2372. [Google Scholar] [CrossRef] [Green Version]
  66. Poulos, M.G.; Ramalingam, P.; Gutkin, M.C.; Llanos, P.; Gilleran, K.; Rabbany, S.Y.; Butler, J.M. Endothelial transplantation rejuvenates aged hematopoietic stem cell function. J. Clin. Investig. 2017, 127, 4163–4178. [Google Scholar] [CrossRef] [Green Version]
  67. Li, B.; Bailey, A.S.; Jiang, S.; Liu, B.; Goldman, D.C.; Fleming, W.H. Endothelial cells mediate the regeneration of hematopoietic stem cells. Stem Cell Res. 2010, 4, 17–24. [Google Scholar] [CrossRef] [Green Version]
  68. Piryani, S.O.; Jiao, Y.; Kam, A.Y.F.; Liu, Y.; Vo-Dinh, T.; Chen, B.J.; Chao, N.J.; Doan, P.L. Endothelial Cell-Derived Extracellular Vesicles Mitigate Radiation-Induced Hematopoietic Injury. Int. J. Radiat. Oncol Biol. Phys. 2019, 104, 291–301. [Google Scholar] [CrossRef]
  69. Kim, M.M.; Schlussel, L.; Zhao, L.; Himburg, H.A. Dickkopf-1 Treatment Stimulates Hematopoietic Regenerative Function in Infused Endothelial Progenitor Cells. Radiat. Res. 2019, 192, 53–62. [Google Scholar] [CrossRef]
  70. Zachman, D.K.; Leon, R.P.; Das, P.; Goldman, D.C.; Hamlin, K.L.; Guha, C.; Fleming, W.H. Endothelial cells mitigate DNA damage and promote the regeneration of hematopoietic stem cells after radiation injury. Stem Cell Res. 2013, 11, 1013–1021. [Google Scholar] [CrossRef] [Green Version]
  71. Wang, J.; Boerma, M.; Fu, Q.; Hauer-Jensen, M. Significance of endothelial dysfunction in the pathogenesis of early and delayed radiation enteropathy. World J. Gastroenterol. 2007, 13, 3047–3055. [Google Scholar] [CrossRef]
  72. Gur-Cohen, S.; Itkin, T.; Chakrabarty, S.; Graf, C.; Kollet, O.; Ludin, A.; Golan, K.; Kalinkovich, A.; Ledergor, G.; Wong, E.; et al. PAR1 signaling regulates the retention and recruitment of EPCR-expressing bone marrow hematopoietic stem cells. Nat. Med. 2015, 21, 1307–1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Geiger, H.; Pawar, S.A.; Kerschen, E.J.; Nattamai, K.J.; Hernandez, I.; Liang, H.P.; Fernández, J.A.; Cancelas, J.A.; Ryan, M.A.; Kustikova, O.; et al. Pharmacological targeting of the thrombomodulin–activated protein C pathway mitigates radiation toxicity. Nat. Med. 2012, 18, 1123–1129. [Google Scholar] [CrossRef] [PubMed]
  74. Termini, C.M.; Pang, A.; Fang, T.; Roos, M.; Chang, V.Y.; Zhang, Y.; Setiawan, N.J.; Signaevskaia, L.; Li, M.; Kim, M.M.; et al. Neuropilin 1 regulates bone marrow vascular regeneration and hematopoietic reconstitution. Nat. Commun. 2021, 12, 6990. [Google Scholar] [CrossRef]
  75. MacVittie, T.J.; Farese, A.M.; Bennett, A.; Gelfond, D.; Shea-Donohue, T.; Tudor, G.; Booth, C.; McFarland, E.; Jackson, W., 3rd. The Acute Gastrointestinal Subsyndrome of the Acute Radiation Syndrome: A rhesus macaque model. Health Phys. 2012, 103, 411–426. [Google Scholar] [CrossRef]
  76. MacVittie, T.J.; Bennett, A.; Booth, C.; Garofalo, M.; Tudor, G.; Ward, A.; Shea-Donohue, T.; Gelfond, D.; McFarland, E.; Jackson, W., 3rd; et al. The Prolonged Gastrointestinal Syndrome in Rhesus Macaques: The relationship between gastrointestinal, hematopoietic, and delayed multi-organ sequelae following acute, potentially lethal, partial-body irradiation. Health Phys. 2012, 103, 427–453. [Google Scholar] [CrossRef] [Green Version]
  77. Santos, A.J.M.; Lo, Y.-H.; Mah, A.T.; Kuo, C.J. The Intestinal Stem Cell Niche: Homeostasis and Adaptations. Trends Cell Biol. 2018, 28, 1062–1078. [Google Scholar] [CrossRef]
  78. Spadoni, I.; Pietrelli, A.; Pesole, G.; Rescigno, M. Gene expression profile of endothelial cells during perturbation of the gut vascular barrier. Gut Microbes 2016, 7, 540–548. [Google Scholar] [CrossRef] [Green Version]
  79. Schuller, B.W.; Binns, P.J.; Riley, K.J.; Ma, L.; Hawthorne, M.F.; Coderre, J.A. Selective irradiation of the vascular endothelium has no effect on the survival of murine intestinal crypt stem cells. Proc. Natl. Acad. Sci. USA 2006, 103, 3787–3792. [Google Scholar] [CrossRef] [Green Version]
  80. Williams, J.P.; Jackson, I.L.; Shah, J.R.; Czarniecki, C.W.; Maidment, B.W.; DiCarlo, A.L. Animal Models and Medical Countermeasures Development for Radiation-Induced Lung Damage: Report from an NIAID Workshop. Radiat. Res. 2012, 177, e0025–e0039. [Google Scholar] [CrossRef] [Green Version]
  81. Mehrvar, S.; Mostaghimi, S.; Camara, A.K.; Foomani, F.; Narayanan, J.; Fish, B.; Medhora, M.; Ranji, M. Three-dimensional vascular and metabolic imaging using inverted autofluorescence. J. Biomed. Opt. 2021, 26, 076002. [Google Scholar] [CrossRef] [PubMed]
  82. Ghosh, S.N.; Wu, Q.; Mäder, M.; Fish, B.L.; Moulder, J.E.; Jacobs, E.R.; Medhora, M.; Molthen, R.C. Vascular Injury after Whole Thoracic X-Ray Irradiation in the Rat. Int. J. Radiat. Oncol. 2009, 74, 192–199. [Google Scholar] [CrossRef] [Green Version]
  83. Robbins, M.E.; Brunso-Bechtold, J.K.; Peiffer, A.M.; Tsien, C.I.; Bailey, J.E.; Marks, L.B. Imaging Radiation-Induced Normal Tissue Injury. Radiat. Res. 2012, 177, 449–466. [Google Scholar] [CrossRef] [PubMed]
  84. Farhood, B.; Aliasgharzadeh, A.; Amini, P.; Rezaeyan, A.; Tavassoli, A.; Motevaseli, E.; Shabeeb, D.; Musa, A.E.; Najafi, M. Mitigation of Radiation-Induced Lung Pneumonitis and Fibrosis Using Metformin and Melatonin: A Histopathological Study. Medicina 2019, 55, 417. [Google Scholar] [CrossRef] [Green Version]
  85. Graham, M.M.; Evans, M.L.; Dahlen, D.D.; Mahler, P.A.; Rasey, J.S. Pharmacological alteration of the lung vascular response to radiation. Int. J. Radiat. Oncol. Biol. Phys. 1990, 19, 329–339. [Google Scholar] [CrossRef]
  86. Klaus, R.; Niyazi, M.; Lange-Sperandio, B. Radiation-induced kidney toxicity: Molecular and cellular pathogenesis. Radiat. Oncol. 2021, 16, 43. [Google Scholar] [CrossRef]
  87. Gao, F.; Dong, W.; Liu, P.; Narayanan, J.; Fish, B.L.; Jacobs, E.R.; Medhora, M. Molecular Changes in miRNA in Irradiated Rat Kidneys: Role of miR-34a and its Vascular Targets in the Notch Pathway. Radiat. Res. 2021, 196, 611–622. [Google Scholar] [CrossRef]
  88. 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. 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. [Google Scholar]
  89. Unthank, J.L.; Ortiz, M.; Trivedi, H.; Pelus, L.M.; Sampson, C.H.; Sellamuthu, R.; Fisher, A.; Chua, H.L.; Plett, A.; Orschell, C.M.; et al. Cardiac and Renal Delayed Effects of Acute Radiation Exposure: Organ Differences in Vasculopathy, Inflammation, Senescence and Oxidative Balance. Radiat. Res. 2019, 191, 383–397. [Google Scholar] [CrossRef]
  90. Boerma, M.; Hauer-Jensen, M. Potential Targets for Intervention in Radiation-Induced Heart Disease. Curr. Drug Targets 2010, 11, 1405–1412. [Google Scholar] [CrossRef] [Green Version]
  91. Baker, J.E.; Fish, B.L.; Su, J.; Haworth, S.T.; Strande, J.L.; Komorowski, R.A.; Migrino, R.Q.; Doppalapudi, A.; Harmann, L.; Allen Li, X.; et al. 10 Gy total body irradiation increases risk of coronary sclerosis, degeneration of heart structure and function in a rat model. Int. J. Radiat. Biol. 2009, 85, 1089–1100. [Google Scholar] [CrossRef] [PubMed]
  92. van Luijk, P.; Novakova-Jiresova, A.; Faber, H.; Schippers, J.M.; Kampinga, H.H.; Meertens, H.; Coppes, R.P. Radiation Damage to the Heart Enhances Early Radiation-Induced Lung Function Loss. Cancer Res. 2005, 65, 6509–6511. [Google Scholar] [CrossRef] [Green Version]
  93. Andrews, R.N.; Bloomer, E.G.; Olson, J.D.; Hanbury, D.B.; Dugan, G.O.; Whitlow, C.T.; Cline, J.M. Non-Human Primates Receiving High-Dose Total-Body Irradiation are at Risk of Developing Cerebrovascular Injury Years Postirradiation. Radiat. Res. 2020, 194, 277–287. [Google Scholar] [CrossRef]
  94. Wang, J.; Zheng, H.; Kulkarni, A.; Ou, X.; Hauer-Jensen, M. Regulation of early and delayed radiation responses in rat small intestine by capsaicin-sensitive nerves. Int. J. Radiat. Oncol. Biol. Phys. 2006, 64, 1528–1536. [Google Scholar] [CrossRef]
  95. Lee, C.-L.; Daniel, A.R.; Holbrook, M.; Brownstein, J.; Silva Campos, L.D.; Hasapis, S.; Ma, Y.; Borst, L.B.; Badea, C.T.; Kirsch, D.G. Sensitization of Vascular Endothelial Cells to Ionizing Radiation Promotes the Development of Delayed Intestinal Injury in Mice. Radiat. Res. 2019, 192, 258–266. [Google Scholar] [CrossRef] [Green Version]
  96. Richter, K.K.; Fink, L.M.; Hughes, B.M.; Sung, C.-C.; Hauer-Jensen, M. Is the loss of endothelial thrombomodulin involved in the mechanism of chronicity in late radiation enteropathy? Radiother. Oncol. 1997, 44, 65–71. [Google Scholar] [CrossRef]
  97. Richter, K.K.; Fink, L.M.; Hughes, B.M.; Shmaysani, H.M.; Sung, C.-C.; Hauer-Jensen, M. Differential effect of radiation on endothelial cell function in rectal cancer and normal rectum. Am. J. Surg. 1998, 176, 642–647. [Google Scholar] [CrossRef]
  98. Pathak, R.; Wang, J.; Garg, S.; Aykin-Burns, N.; Petersen, K.-U.; Hauer-Jensen, M. Recombinant Thrombomodulin (Solulin) Ameliorates Early Intestinal Radiation Toxicity in a Preclinical Rat Model. Radiat. Res. 2016, 186, 112–120. [Google Scholar] [CrossRef] [Green Version]
  99. Jang, H.; Kwak, S.-Y.; Park, S.; Kim, K.; Kim, Y.-H.; Na, J.; Kim, H.; Jang, W.-S.; Lee, S.-J.; Kim, M.J.; et al. Pravastatin Alleviates Radiation Proctitis by Regulating Thrombomodulin in Irradiated Endothelial Cells. Int. J. Mol. Sci. 2020, 21, 1897. [Google Scholar] [CrossRef] [Green Version]
  100. Sridharan, V.; Johnson, K.A.; Landes, R.D.; Cao, M.; Singh, P.; Wagoner, G.; Hayar, A.; Sprick, E.D.; Eveld, K.A.; Bhattacharyya, A.; et al. 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]
  101. Williams, J.P.; Brown, S.L.; Georges, G.E.; Hauer-Jensen, M.; Hill, R.P.; Huser, A.K.; Kirsch, D.G.; MacVittie, T.J.; Mason, K.A.; Medhora, M.M.; et al. Animal Models for Medical Countermeasures to Radiation Exposure. Radiat. Res. 2010, 173, 557–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Rubin, P.; Finkelstein, J.; Shapiro, D. Molecular biology mechanisms in the radiation induction of pulmonary injury syndromes: Interrelationship between the alveolar macrophage and the septal fibroblast. Int. J. Radiat. Oncol. Biol. Phys. 1992, 24, 93–101. [Google Scholar] [CrossRef] [PubMed]
  103. Ghosh, S.N.; Zhang, R.; Fish, B.L.; Semenenko, V.A.; Li, X.A.; Moulder, J.E.; Jacobs, E.R.; Medhora, M. Renin-Angiotensin System Suppression Mitigates Experimental Radiation Pneumonitis. Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 1528–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Sharma, G.P.; Fish, B.L.; Frei, A.C.; Narayanan, J.; Gasperetti, T.; Scholler, D.; Pierce, L.; Szalewski, N.; Blue, N.; Medhora, M.; et al. Pharmacologic ACE-Inhibition Mitigates Radiation-Induced Pneumonitis by Suppressing ACE-Expressing Lung Myeloid Cells. Int. J. Radiat. Oncol. Biol. Phys. 2022, 113, 177–191. [Google Scholar] [CrossRef] [PubMed]
  105. Jacobson, J.R. Sphingolipids as a Novel Therapeutic Target in Radiation-Induced Lung Injury. Cell Biochem. Biophys. 2021, 79, 509–516. [Google Scholar] [CrossRef]
  106. Mathew, B.; Jacobson, J.R.; Berdyshev, E.; Huang, Y.; Sun, X.; Zhao, Y.; Gerhold, L.M.; Siegler, J.; Evenoski, C.; Wang, T.; et al. Role of sphingolipids in murine radiation-induced lung injury: Protection by sphingosine 1-phosphate analogs. FASEB J. 2011, 25, 3388–3400. [Google Scholar] [CrossRef] [Green Version]
  107. Fountain, M.D.; McLellan, L.A.; Smith, N.L.; Loughery, B.F.; Rakowski, J.T.; Tse, H.Y.; Hillman, G.G. Isoflavone-mediated radioprotection involves regulation of early endothelial cell death and inflammatory signaling in Radiation-Induced lung injury. Int. J. Radiat. Biol. 2020, 96, 245–256. [Google Scholar] [CrossRef]
  108. 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]
  109. Hayashi, T.; Morishita, Y.; Kubo, Y.; Kusunoki, Y.; Hayashi, I.; Kasagi, F.; Hakoda, M.; Kyoizumi, S.; Nakachi, K. Long-term effects of radiation dose on inflammatory markers in atomic bomb survivors. Am. J. Med. 2005, 118, 83–86. [Google Scholar] [CrossRef]
  110. 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] [Green Version]
  111. Azizova, T.V.; Grigorieva, E.S.; Hunter, N.; Pikulina, M.V.; Moseeva, M.B. Risk of mortality from circulatory diseases in Mayak workers cohort following occupational radiation exposure. J. Radiol. Prot. 2015, 35, 517–538. [Google Scholar] [CrossRef]
  112. Sera, N.; Hida, A.; Imaizumi, M.; Nakashima, E.; Akahoshi, M. The Association Between Chronic Kidney Disease and Cardiovascular Disease Risk Factors in Atomic Bomb Survivors. Radiat. Res. 2013, 179, 46–52. [Google Scholar] [CrossRef] [Green Version]
  113. Jaenke, R.S.; Robbins, M.E.; Bywaters, T.; Whitehouse, E.; Rezvani, M.; Hopewell, J.W. Capillary endothelium. Target site of renal radiation injury. Lab. Investig. 1993, 68, 396–405. [Google Scholar]
  114. Obeidat, M.; Obeidat, M.; Ballermann, B.J. Glomerular endothelium: A porous sieve and formidable barrier. Exp. Cell Res. 2012, 318, 964–972. [Google Scholar] [CrossRef]
  115. Lawton, C.A.; Cohen, E.P.; Barber-Derus, S.W.; Murray, K.J.; Ash, R.C.; Casper, J.T.; Moulder, J.E. Late renal dysfunction in adult survivors of bone marrow transplantation. Cancer 1991, 67, 2795–2800. [Google Scholar] [CrossRef]
Figure 1. Schematic depiction of the main mechanisms of radiation-induced endothelial cell damage.
Figure 1. Schematic depiction of the main mechanisms of radiation-induced endothelial cell damage.
Toxics 10 00747 g001
Figure 2. Schematic depiction of the mechanisms which contribute to radiation-induced endothelial cell dysfunction.
Figure 2. Schematic depiction of the mechanisms which contribute to radiation-induced endothelial cell dysfunction.
Toxics 10 00747 g002
Table 1. Candidate Mitigators of Radiation-Induced Endothelial Cell Injury.
Table 1. Candidate Mitigators of Radiation-Induced Endothelial Cell Injury.
MitigatorMechanism of ActionRefs.
PKCδ-TATBlocks upregulation of adhesion molecules[7,39,40]
SOD MimeticsScavengers of ROS[46,47,48]
SepiapterinRestores eNOS “coupling” and reduces ROS [49]
ECs/VesiclesRestores hematopoiesis and rescues H-ARS lethality[65,66,67,68,69,70]
Anti-Sema3ABlocks SEMA3A binding to Neuropilin-1[74]
APCCorrects for thrombomodulin loss[72,73,98,99]
ACE inhibitorsReduces recruitment of inflammatory immune cells [103,104]
Anti-ceramide Blocks ceramide-mediated apoptotic death[10,11,12]
S1P AnalogsRestores endothelial barrier function[105,106]
Soy isoflavonesPromote clearance of damaged endothelium[107]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sharma, G.P.; Himburg, H.A. Organ-Specific Endothelial Dysfunction Following Total Body Irradiation Exposure. Toxics 2022, 10, 747. https://doi.org/10.3390/toxics10120747

AMA Style

Sharma GP, Himburg HA. Organ-Specific Endothelial Dysfunction Following Total Body Irradiation Exposure. Toxics. 2022; 10(12):747. https://doi.org/10.3390/toxics10120747

Chicago/Turabian Style

Sharma, Guru Prasad, and Heather A. Himburg. 2022. "Organ-Specific Endothelial Dysfunction Following Total Body Irradiation Exposure" Toxics 10, no. 12: 747. https://doi.org/10.3390/toxics10120747

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