• Open Access
    Review

    The head of invasive cardiologists as a target of professional exposure to ionizing radiation

    Andrea Borghini *

    Explor Cardiol. 2024;2:224–240 DOI: https://doi.org/10.37349/ec.2024.00036

    Received: August 22, 2024 Accepted: October 11, 2024 Published: November 05, 2024

    Academic Editor: Quirino Ciampi, Fatebenefratelli Hospital of Benevento, Italy.

    Abstract

    Exposure to ionizing radiation has recognized detrimental cancer and non-cancer health effects. These effects are now well-proven not only for high doses > 1,000 millisieverts (mSv) associated with head radiotherapy but also for moderate (100–1,000 mSv) and even low (< 100 mSv) doses, of interest for professionally exposed cardiologists. The head of interventional cardiologists is highly exposed to ionizing radiation, with possible damage to the eye and brain. Unprotected interventional cardiologists experience head radiation doses up to ten times greater than chest doses below lead aprons, with marked exposure to the left hemisphere of the brain reaching up to 2 Sv—equivalent to 10,000 chest X-rays over a professional lifetime. This narrative review aims to provide an overview of the background of radioprotection, the biological mechanisms involved, and the epidemiological evidence regarding the health effects of head exposure to ionizing radiation in invasive cardiologists. These health effects include cataracts, brain cancer, cerebrovascular diseases, neurodegeneration, and mood disorders. The evidence gathered from other exposed populations, which experienced similar eye and brain doses, has also been reviewed. This is important because the doses, risks, and effects are consistent in cases of repeated exposures, which occur more frequently for patients, and in situations involving chronic low doses, as seen with interventional cardiologists. Despite these risks, effective protective measures—such as suspended lead ceilings, curtains, and specialized eyewear—can reduce radiation exposure to near-zero levels. In some fields, like interventional cardiac electrophysiology, a groundbreaking near-zero radiation approach using non-fluoroscopic methods has been created, eliminating radiation exposure and alleviating orthopedic stress and operational discomfort. The race to zero radiation in interventional cardiology is ongoing.

    Keywords

    Alzheimer’s disease, glioblastoma, ionizing radiation, meningioma, Parkinson’s disease

    Introduction

    Ionizing radiation from medical applications is a very significant source of exposure for patients and healthcare professionals working in the cardiac catheterization laboratory. For patients, the exposure per capita per year totaled the effective dose of 3.0 millisievert (mSv) in the USA in the radiological year 2006, corresponding to the biologic risk of 150 chest X-rays. The exposure declined to 2.29 mSv (120 chest X-rays) in the radiologic year 2016, thanks to technological upgrades, greater awareness of radiologic risk, and the development of non-ionizing alternatives such as stress echocardiography and magnetic resonance imaging for the diagnosis of cardiovascular disease [1]. Cardiovascular testing accounts for about 50% of all radiological exposures for patients, and invasive cardiologists are the most exposed health professionals, 3 times more exposed than diagnostic radiologists or nuclear physicians [2]. Ionizing radiation has a wide range of health effects, mostly cancer [3]. However, other non-cancer effects such as cardiovascular and cerebrovascular diseases are also important at the individual and population levels [4]. The head is a highly exposed target for invasive cardiologists and also the potential victim of a variety of damaging clinical effects mediated by different, disease-specific cell populations (Table 1). Damage of cell and mitochondrial DNA is a common initiating molecular step, and low-grade inflammation is a secondary cellular mechanism of progression of cancer, atherosclerotic, and neurodegenerative disease [57]. Chronic low-grade neuroinflammation may eventually lead to irreversible damage of neuronal structure and function, hitting cholinergic neurons in Alzheimer’s or dopaminergic neurons in Parkinson’s disease [8]. Ionizing radiation acts not directly on neurons but on microglial cells, key regulators of neuroinflammation and oxidative stress in both these neurodegenerative diseases [9].

    Different cell subtypes are involved in main cancer and non-cancer effects on the brain

    DiseaseLECEC and SMCArachnoid cellsAstrocytesSchwann cellsMicroglial cells
    Cataract    √
    Stroke    √
    Meningioma    √
    Glioblastoma    √
    Neuroma    √
    Alzheimer    √
    Parkinson    √
    Display full size

    LEC: lens epithelial cells; EC: endothelial cells; SMC: smooth muscle cells; √: involved

    These effects were first demonstrated after high dose (> 1,000 mSv) of head radiotherapy in cancer, but in the last decade increasingly recognized after moderate (100–1,000 mSv) and even low dose (< 100 mSv) exposures. Consequences of radiation with moderate or low dose exposures appear after a long latency time, and can be less easy to recognize, but are present and linearly related to the dose exposure, probably without a threshold effect. Every dose counts, there is no safe dose, and the low doses at low dose rates typically met in medically exposed patients and professionals, tend to be more toxic than acute high-dose exposures [10]. Tissue reactions (such as skin effects with erythema and ulcers from high radiation exposure in patients during intensive cardiac interventions) require medium to high doses of radiation on the target tissue, recognizing a threshold dose, and a latency period of days or weeks [4]. Tissue reactions were traditionally considered tissue reactions (previously named deterministic effects), conceptually different from stochastic effects, such as cancer and hereditary effects. However, in the last 15 years evidence has accumulated that eye, cardiovascular, cerebrovascular, and neurologic effects of radiation exposure tend to deviate from the definition of tissue reactions, show significant risks also at low dose and for low-dose rates, and their effects are apparent at doses and dose rates substantially lower than previously thought, sometimes many years after exposure [10].

    This narrative review will briefly describe the radioprotection background, the biological basis, and the epidemiological evidence supporting the health effects of head exposure to ionizing radiation in invasive cardiologists. The evidence obtained in patients, for similar eye and brain doses, has also been reviewed since the doses, the risks, and the effects are very much the same in the case of repeated exposures as more frequently happens for patients and in the case of chronic, low-dose, low-dose-rate refracted doses as happens with interventional cardiologists. Doctors may become patients, and the exposures as patients add up to professional exposures.

    Brain organ dose: patients and doctors

    For patients, head computed tomography or diagnostic cerebral angiography with whole-body exposures of 5 mSv to 10 mSv are associated with brain organ dose exposure in the range of 50 mSv to 100 mSv per scan [11]. Interventional neuroradiology intervention with endovascular procedures such as closure of cerebral aneurysm involves a brain dose exposure between 200 mSv and 1,000 mSv [1215].

    Brain organ doses are derived from recent literature dating back to the 1990s, but it is important to apply correction factors for earlier exposures. Specifically, a correction factor of 1.5 is necessary for radiographic procedures conducted in the 1980s, and a factor of 2.0 applies to nuclear medicine scans performed prior to 1980 [16].

    Regarding interventional cardiologists, the cumulative lifetime professional exposure ranges from 100 mSv to 200 mSv [17], with 1 Sv associated with right-sided cranial radiation and up to 2 Sv for left-sided cranial radiation [18, 19]. In fact, in a typical cardiac catheterization room, the cardiologist stands on the right side of the patient, and the left side of the head of the operator is closer to the scatter radiation coming from the patient [20]. The organ dose is far in excess to the low dose range (below 100 mSv or 5,000 chest X-rays). In addition, the reassuring concept that refracted low-dose exposures can be less damaging because there is time to repair damaged DNA has been disproved and the low, low-dose-rate exposures are at least equally, and probably more damaging than acute high-dose exposures, possibly because low-dose it is more likely that the cells suffer non-lethal DNA damage that can be passed to next cell generations [2123].

    The doses of medical or professional exposures are lower than the doses employed for head and neck cancer radiotherapy (Table 2). In the case of radiotherapy, high doses (> 1,000 mSv) are involved, with brain organ doses in the range of hundreds of thousands of chest X-rays. Following head radiotherapy, various essential neurological structures, including the brain, brainstem, spinal cord, cranial nerves, nerve plexuses, autonomic pathways, brain vasculature, and neurosensory organs, are exposed to varying degrees of radiation. This exposure can lead to a wide array of long-term neurological side effects in survivors. These complications often cause permanent symptoms, negatively impact patients’ quality of life, and account for a significant number of non-cancer-related deaths [24].

    Organ dose to the body and the brain (from refs. [6, 7, 10, 11, 15])

    SubjectsWhole body dose (mSv)Brain organ dose (mSv)
    Patients
    Skull X-ray1.80.9
    Thyroid isotope scan3.21
    Cerebral angiography85
    Head CT1.620
    Interventional Neuroradiology20–100200–1,000
    Doctors
    Per procedure0.030.3
    Lifetime (40 years)2002,000
    Radiotherapy
    Per cycle1,000–40,00010,000–400,000
    Display full size

    The effects of low-dose radiation exposure on brain cells

    Ionizing radiation exposure can lead to significant cerebrovascular and neuronal damage in the brain, primarily through a cascade of molecular and cellular mechanisms. These effects are often seen in both therapeutic settings, such as radiation therapy for brain tumors, and in accidental or occupational exposure to radiation.

    At the molecular level, ionizing radiation primarily causes damage by generating reactive oxygen species [25]. These highly reactive molecules can damage cellular components, including lipids, proteins, and DNA. The endothelial cells lining the blood-brain barrier are particularly vulnerable. DNA damage in these cells can lead to apoptosis (programmed cell death) or senescence (a state of permanent cell cycle arrest), disrupting the integrity of the blood-brain barrier. This disruption allows for the extravasation of blood components into the brain parenchyma, leading to inflammation and further tissue damage.

    One critical pathway activated by ionizing radiation is the DNA damage response (DDR) pathway, which includes the activation of ataxia-telangiectasia mutated and p53 proteins. These proteins initiate cell cycle arrest, DNA repair, or apoptosis depending on the extent of damage. Persistent activation of the DDR pathway can result in endothelial cell dysfunction, contributing to chronic cerebrovascular damage [6, 2628].

    At the cellular level, ionizing radiation induces changes in the microenvironment of the brain’s vasculature. This includes endothelial cell activation, which promotes the expression of adhesion molecules such as intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1). These molecules facilitate the adhesion and infiltration of inflammatory cells like macrophages and lymphocytes into the brain tissue, exacerbating inflammation and vascular damage [20, 26].

    Radiation also impacts astrocytes, the star-shaped glial cells in the brain that play a key role in maintaining and supporting neuronal function. Astrocytes exposed to ionizing radiation can undergo reactive gliosis, a process characterized by hypertrophy and proliferation. Reactive astrocytes secrete cytokines and chemokines that further promote inflammation and can alter the function of the blood-brain barrier [26].

    Microglia, the resident immune cells of the brain, also respond to ionizing radiation. Activated microglia release pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6, which contribute to a pro-inflammatory milieu. This chronic inflammatory state can perpetuate vascular damage and promote neurodegenerative processes [2628].

    The central nervous system is exceptionally radiosensitive and thus more liable to neurological insult even at low doses [26]. Neurogenesis occurs not only in the developing brain but also in the adult brain in specific regions such as the hippocampus and the impairment of adult neurogenesis has been implicated in depressive disorders such as anxiety and depression [29]. Fractionated low-dose irradiation has been shown to impair adult hippocampal neurogenesis and cognitive function in experimental animals [30].

    In summary, ionizing radiation exposure leads to cerebrovascular damage through a complex interplay of molecular and cellular mechanisms. DNA damage and reactive oxygen species generation trigger endothelial dysfunction, inflammation, and disruption of the blood-brain barrier. The involvement of various cell types, including endothelial cells, astrocytes, pericytes, and microglia, underscores the multifaceted nature of radiation-induced injury (Figure 1).

    From radiation exposure to the head through molecular and cellular effects to the clinical manifestations of radiation-induced disease

    A deeper understanding of the biological mechanisms involved is essential for developing protective strategies and treatments for individuals exposed to low-dose ionizing radiation. In this context, the use of three-dimensional (3D) brain culture models to investigate oxidative stress induced-DNA damage, telomere attrition, mitochondrial dysfunction, and neuroinflammation can offer valuable insights into the intricate cellular responses to ionizing radiation, serving as simplified representations of the structures and functions of in vivo organs. This is highlighted by recent results from Oyefeso et al. [31] that demonstrated the potential of using brain organoids to characterize cell-specific radiosensitivity and early radiation-induced gene expression changes within the human brain at low-to-moderate doses [31].

    Importantly, human induced pluripotent stem cells (hiPSCs) can serve as a powerful modeling system to elucidate the cellular and biological effects of radiation exposures on the central nervous system using conventional 2D arrays and more advanced tissue engineering approaches with organoids and other 3D models. The health effects of ionizing radiation may be influenced by genetic polymorphisms, which can make certain individuals more susceptible to its effects. Thus, since hiPSC lines reprogrammed from selected donors reflect their individual genetic backgrounds, iPSC disease modeling may reveal important gene-environment interactions that exacerbate cerebrovascular disease and predispose certain individuals to adverse outcomes.

    The eye as a radiosensitive target

    Cataract, or opacification of the lens, is often associated with visual impairment and may be classified into three main categories: nuclear, cortical, and posterior subcapsular, according to their anatomic location. Among the three major areas of age-related cataracts, posterior subcapsular is the least common but it is the one most frequently associated with ionizing radiation exposure. New exposure limits set by the International Commission on Radiological Protection (ICRP) in 2012 markedly reduced the allowed annual exposure of the eye from 150 mSv (ante-2012) to 20 mSv/year (since 2012). The eye is a very radiosensitive organ. Interventional cardiologists and staff have a 4-fold higher risk of posterior subcapsular opacity and cataracts compared to non-exposed controls [3237]. Although traditionally considered a deterministic effect with threshold dose, it is not clear if the cataract is explained by a stochastic effect (without threshold, like cancer) or a tissue reaction (with threshold, a deterministic effect, like skin ulcer after high dose exposure). Ionizing radiation can induce cataract formation through complex molecular and cellular mechanisms that disrupt lens transparency. The lens, composed of epithelial and fiber cells, is particularly vulnerable due to its high metabolic activity and lifelong accumulation of damage. Ionizing radiation causes direct DNA breaks and generates reactive oxygen species, leading to oxidative stress. This oxidative stress can result in DNA mutations, apoptosis, and cellular senescence. Reactive oxygen species can oxidize lens lipids and proteins, particularly crystallins, leading to protein aggregation and insolubilization. These aggregated proteins scatter light, impairing lens transparency [38, 39]. At the cellular level, ionizing radiation can cause epithelial cell death, DNA damage, and dysfunction, disrupting cell proliferation and differentiation necessary for lens fiber cell formation. This complex interplay of molecular and cellular damage disrupts lens transparency, resulting in cataract formation [38, 39]. There is emerging evidence that the risk of normal-tension glaucoma, diabetic retinopathy, and macular degeneration, may be increased after ionizing radiation exposure [40].

    Brain cancer disease

    In theory, the brain is a relatively radioresistant organ. A tissue weighting factor of 0.01 was applied to the brain by the ICRP in Document 103 released in 2007. The tissue weighting factor is 12 times lower for the brain compared to highly radiosensitive organs such as the lung, bone marrow, colon, and stomach [41]. To classify radiation-induced tumors, Cahan et al. [42] proposed back in 1948 certain criteria for radiation-induced carcinogenesis as follows: the tumor should arise in the field of irradiation, it must be histologically different from the disease that occasioned the irradiation; a suitable duration must elapse between the radiation and clinical onset of tumor; no other obvious predisposing conditions for oncogenesis be present [42].

    Brain cancer is a recognized complication of brain radiotherapy and has been increasingly recognized also after low to moderate doses of radiation in patients and occupationally exposed workers [3248]. The 3 types of brain cancer associated with chronic low-dose radiation exposure are acoustic neuroma, meningioma, and glioblastoma. Vestibular schwannomas (also known as acoustic neuromas) are benign, slow-growing tumors of the eighth cranial nerve, that arise from Schwann cells ensheathing the vestibular portion of the vestibulocochlear nerve. Ionizing radiation exposure is a recognized risk factor for schwannomas [4346].

    Meningioma is the most frequent type of brain cancer and is highly radiosensitive. Meninges are made of highly proliferating tissue, and host neural precursor cells with high mitotic activity. Meningiomas are mostly benign tumors originating from arachnoid cap cells [4756]. Glioblastoma originates from somatic mutations in neural stem cells and glial precursor cells. Repeated head CT with whole-body exposures of 5 mSv to 10 mSv are associated with brain organ dose exposure in the range of 50 mSv to 100 mSv per scan [57]. A large-scale study of close to 1 million patients with CT at a pediatric or young age shows a significant linear dose—response relationship for all brain cancers and gliomas individually [58]. Clusters of mostly left-sided meningioma and glioblastoma have been described by interventional radiologists [5961]. The famous cardiac surgeon and interventional cardiologist who pioneered hybrid coronary revascularization techniques Edward B. Diethrich developed a brain tumor (oligodendroglioma) in 2012, as well as cataracts in both eyes and dense, calcified plaque in his carotid artery [62]. He then made it his mission to educate other healthcare workers about the dangerous effects of radiation, joining the Organization for Occupational Radiation Safety in Interventional Fluoroscopy to release a documentary describing the effects that radiation had upon him [63].

    Interventional neuroradiology with endovascular procedures such as the closure of cerebral aneurysms involves a brain dose exposure between 500 mSv and 1,000 mSv and therefore also patients may develop brain cancer after diagnostic and therapeutic exposure [64].

    A 2019 meta-analysis of 26 studies supports a significant association between dental X-rays and thyroid cancer and meningioma [56], but recent large-scale studies and meta-analysis failed to find an association between diagnostic X-rays and brain cancer [65].

    Retinoblastoma is a malignant tumor of the developing retina that occurs in children, usually before age five years. Retinoblastoma may be unifocal or multifocal. About 60% of affected individuals have unilateral retinoblastoma with a mean age of diagnosis of 24 months; about 40% have bilateral retinoblastoma with a mean age of diagnosis of 15 months [66]. Maternal or paternal gonadal exposure (with radiologic or nuclear medicine diagnostic examinations) in the pre-conception time (9 weeks before conception in males) increases 3-fold to 6-fold the risk of retinoblastoma resulting from new germline mutation in offspring [67, 68]. This finding is of potential concern for interventional cardiologists since their cumulative gonadal exposure in a professional lifetime can range between 0.5 Sv and 1 Sv [69, 70].

    Cerebrovascular disease

    Radiation exposure increases cardiovascular disease and particularly cerebrovascular disease risk. This has long been known for radiotherapy doses. Still, now evidence has also accumulated with moderate (100–500 mSv) and low (< 100 mSv), and the atherosclerotic effect is considered proven for the dose ranges common for interventional cardiologists and for patients exposed to repeated head and neck radiology examinations.

    From the epidemiologic viewpoint, large-scale meta-analyses have conclusively proven on millions of patients that the risk of cardiovascular disease is of the same order of magnitude as radiation-induced cancer risk [7174]. This risk is detectable at low doses, and probably low dose rates are even more dangerous for their pro-atherosclerotic effects than high-dose acute administration. Radiation is a risk factor for atherosclerosis and cardiovascular disease, especially cerebrovascular disease [75]. The level of risk is less uncertain at lower doses and lower dose rates, with the possibility that risk per unit dose is greater at lower doses and dose rates [76]. Recently, Cha et al. [77] found limited evidence in support of a positive association between occupational radiation exposure and the overall risk of circulatory disease over a short follow-up period among diagnostic medical radiation workers in South Korea. However, the excess relative risk of morbidity from circulatory diseases was significantly higher in female workers compared to male workers, and for cerebrovascular diseases compared to other subtypes of circulatory diseases [77]. In another study using data from the UK National Registry for Radiation Workers cohort, Hinksman et al. [78] identified an increased risk of mortality from cerebrovascular disease related to low-doses of ionizing radiation. This finding supports the notion that radiation exposure may significantly affect cerebrovascular health at doses lower than those currently recommended by ICRP protection guidelines [78].

    The epidemiological findings are supported by molecular epidemiology and imaging studies that reveal alterations in intermediate and preclinical markers of vascular aging and early atherosclerosis. These include telomere shortening in circulating leukocytes and early thickening of carotid intima-media thickness observed in exposed interventional cardiologists [79]. Telomere shortening is a well-known hallmark of both cellular senescence and organismal aging. Current research consistently indicates that telomere dysfunction plays a key role in the progression of age-related vascular diseases, particularly coronary atherosclerosis, myocardial infarction, ischemic heart disease, and stroke [80, 81]. DNA damage and critically shortened telomeres can induce senescence in both vascular endothelial and smooth muscle cells resulting in an enhanced pro-inflammatory response and endothelial activation [81].

    In vascular cells, ionizing radiation can initially induce irreparable DNA damage and telomere erosion, which activate permanent DDR-inducing vascular cellular senescence [82]. This, in turn, increases senescence-associated secretory phenotype signals, promoting a heightened pro-inflammatory response within the vessel wall and accelerating the progression of atherosclerosis, ultimately resulting in cardio-cerebrovascular complications [82].

    Neurodegeneration

    Systematic reviews and meta-analyses concordantly show an association between ionizing radiation exposure and increased risk for neurodegenerative disease for both patients and doctors. Srivastava et al. [83] identified five types of exposed populations: 1. survivors of atomic bombings in Japan; 2. patients treated with radiation therapy for cancer or other diseases; 3. occupationally exposed workers; 4. those exposed to environmental radiation; 5. patients exposed to radiation from diagnostic radiation imaging procedures. The meta-analysis of over 18 studies shows an increased risk (of 10–17% for every 100 mSv exposure) for Alzheimer’s disease incidence and mortality [83]. The pooled analysis included the One Million Worker study including nuclear power plant workers, industrial radiography workers, exposed military personnel, and 109,019 medical radiation workers with 2,779,838 person-years of follow-up, with a median exposure dose of 9.8 mSv and only 1% with an exposure > 100 mSv. The association remained significant when considering only Parkinson’s disease mortality as a distinct outcome [84]. For cognitive effects, the detrimental effects of low-dose exposure are even more marked than acute high-dose exposures [85, 86]. The epidemiological data are also corroborated by molecular epidemiology and functional testing data showing changes in intermediate proximal, preclinical biomarkers of neurodegeneration such as brain-specific microRNA expression and early neurocognitive changes with reduction of olfactory acuity in exposed interventional cardiologists [8789]. The brain-specific microRNA-134 is involved in synapse development and is directly connected to learning and memory. It has been shown to be dysregulated in conditions such as Alzheimer’s disease, bipolar disorder, oligodendrogliomas, and glioblastomas [90]. Notably, a recent study provided the first evidence that the microRNA-134-mediated post-transcriptional regulation of cAMP response element-binding protein 1 (CREB-1) and brain-derived neurotrophic factor (BDNF) is an important molecular mechanism underlying the plasticity deficit in Alzheimer’s disease, underscoring the critical role of miR-134-5p as a potential therapeutic target for restoring plasticity in Alzheimer’s disease condition [91]. Given its significant dysregulation in interventional cardiologists, it strongly indicates that brain damage could be a major long-term risk of unprotected head irradiation, potentially leading to enduring cognitive impairments [87].

    Mood disorders

    Long-term, low-dose-rate radiation exposure early in life might cause subsequent psychological stress and an increased risk of depression decades later. The radiation-induced dysfunction of the cortico-limbic system in the left dominant hemisphere of the human brain with specific involvement of the impaired neurogenesis in the hippocampus is considered to be the key cerebral basis of post-radiation organic brain damage [92, 93]. The risk of diseases rises with radiation dose. Radiation risks are revealed for organic psychoses, non-psychotic organic brain damage, and acute and chronic cerebrovascular pathology in exposed populations such as Chernobyl liquidators [9498]. Although there is a tremendous biological complexity and multifactorial nature of these symptoms, from stress to sleep deprivation to reduced sunlight exposure, the possibility that these mood disorders exist and have a plausible biological mechanism should be considered in planning future studies and looking for the missing convincing evidence. X-ray stimulation induced the generation of reactive oxygen species in the prefrontal cortex in a dose-dependent manner, leading to the occurrence of depression-like behaviors in the mice, resulting in neuron death triggered by pro-inflammatory signals both in vivo and in vitro [99]. In a survey by Andreassi et al. [100] in interventional cardiologists, mood disorders such as anxiety/depression were 6 times more frequent in exposed professionals compared to unexposed controls.

    Protection as a definitive therapy: ceilings, caps, and curtains

    Despite the abundant evidence of detrimental long-term health effects of chronic low-dose, low-dose-rate radiation exposure, radiation unawareness, and suboptimal protection have been for a long time the rule rather than the exception in the catheterization laboratory [101103]. Genuine lack of information, additive work discomfort and orthopedic strain connected to wearing protection, dose limits perceived as a barrier to personal productivity, and underestimation of radiation risk also encouraged by limited direct evidence in medically exposed populations contributed to exposure above and beyond all regulatory limits of a first generation of invasive cardiologists now suffering an epidemic of radiation-induced damage. Now it is mainstream knowledge that a culture of respect for radiation hazards is needed to enter the cardiac catheterization laboratory [104105] and the primary duty of the invasive cardiologist must protect the operators, the patients, and the staff from the negative effects of chronic occupational exposure to radiation [106]. These effects include significant effects on the eye, brain cancer, cerebrovascular disease, neurodegeneration, and mood disorders.

    Known, proven, and uncertain, possible eye and brain effects of radiation exposure have all the same therapy: prevention through protection. Therefore, a legitimate question is how, not if and when, to protect the head of invasive cardiologists from ionizing radiation (Table 3).

    Protection of the head of interventional cardiologists

    Types of protectionLogisticsProtectionComfort
    Cap, collar, glassesSimple50% to 90%Low
    Suspended ceilingsComplex> 90%Intermediate
    CabinAdvanced99%High
    Zero radiationVery advanced100%Very high
    Display full size

    Once a culture of respect towards radiation hazards has been built, the therapy is simple: to apply the principle of justification, dose optimization, and responsibility embedded in the law and to protect ourselves and the staff most efficiently. There are now many different effective solutions, although probably not all equally effective: the leaded cap, the suspended ceiling, the cabin, and even robotic intervention with remote control from the operator away from the patient and the radiation source [107118]. There is also the most radical and effective solution to move from ionizing radiation imaging to zero-fluoroscopy and ionizing radiation-free methods, now the standard state-of-the-art in cardiac electrophysiology. Each protection system has different logistics, learning curve, cost, and efficacy, and the best solution may vary according to these variables, but the worst solution is to ignore the radiation issue important for the safety of doctors and patients. The final protection is the progressive shift to zero radiation as it has already been achieved for electrophysiology procedures [119122]. In the meantime, careful optimization practices can reduce 10-fold the dose to patients and doctors in the catheterization laboratory [123]. The radiation exposure is dramatically reduced with intravascular ultrasound complementing or replacing fluoroscopy and fluorography [124]. Robotic interventional cardiology allows one to operate at a distance with remote control of catheters [125]. The race to near-zero radiation cardiology continues.

    Conclusions

    In summary, epidemiological evidence suggests a connection between exposure to ionizing radiation and an increased risk of brain cancer, cerebrovascular disease, and neurodegenerative disorders in both patients and doctors. These findings are supported by molecular epidemiology and imaging studies that reveal early changes in intermediate and preclinical disease markers. Despite these risks, effective protective measures such as the use of lead-lined ceilings, curtains, and specialized eyewear can significantly minimize radiation exposure to near-zero levels.

    However, large-scale studies that involve individual dose reconstruction and thorough data collection on potential confounding factors are necessary to more accurately assess the risk of disease. Future research should investigate the extent of brain effects resulting from the interplay of various traditional and environmental risk factors, particularly the potential synergistic effects between pollution and medical radiation exposure.

    Detailed research into the biological mechanisms by which ionizing radiation impacts brain cells is also essential. For instance, employing brain organoids to study neuroinflammation, DNA damage, telomere attrition, and mitochondrial dysfunction could significantly enhance our understanding of these processes. Since genetic research has provided evidence of variability in individual responses to ionizing radiation—demonstrating that some individuals are more susceptible than others—hiPSC disease modeling derived from selected donors may reveal important gene-environment interactions that exacerbate cerebrovascular and neurodegenerative disorders, predisposing certain individuals to adverse outcomes. This approach will be crucial not only for deepening our knowledge of brain disease risks following low-dose ionizing radiation exposure but also for developing effective protective strategies.

    Abbreviations

    3D:

    three-dimensional

    DDR:

    DNA damage response

    hiPSCs:

    human induced pluripotent stem cells

    ICRP:

    International Commission on Radiological Protection

    mSv:

    millisieverts

    Declarations

    Author contributions

    AB: Writing—original draft, Conceptualization, Validation, Supervision. The author read and approved the submitted version.

    Conflicts of interest

    Andrea Borghini who is the Editorial Board Member of Exploration of Cardiology had no involvement in the decision-making or the review process of this manuscript.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2024.

    References

    Mahesh M, Ansari AJ, Jr FAM. Patient Exposure from Radiologic and Nuclear Medicine Procedures in the United States and Worldwide: 2009-2018. Radiology. 2023;307:e239006. [DOI] [PubMed]
    Picano E, Vañó E, Rehani MM, Cuocolo A, Mont L, Bodi V, et al. The appropriate and justified use of medical radiation in cardiovascular imaging: a position document of the ESC Associations of Cardiovascular Imaging, Percutaneous Cardiovascular Interventions and Electrophysiology. Eur Heart J. 2014;35:66572. [DOI] [PubMed]
    United Nations Scientific Committee on the Effects of Atomic Radiation (UNSCEAR) Reports [Internet]. United Nations iLibrary; c2020 [cited 2024 August 20]. Available from: https://www.un-ilibrary.org/content/periodicals/24121428
    ICRP Aobo, Stewart FA, Akleyev AV, Hauer-Jensen M, Hendry JH, Kleiman NJ, et al. ICRP publication 118: ICRP statement on tissue reactions and early and late effects of radiation in normal tissues and organs--threshold doses for tissue reactions in a radiation protection context. Ann ICRP. 2012;41:1322. [DOI] [PubMed]
    Acharya MM, Lan ML, Kan VH, Patel NH, Giedzinski E, Tseng BP, et al. Consequences of ionizing radiation-induced damage in human neural stem cells. Free Radic Biol Med. 2010;49:184655. [DOI] [PubMed]
    Borghini A, Gianicolo EAL, Picano E, Andreassi MG. Ionizing radiation and atherosclerosis: current knowledge and future challenges. Atherosclerosis. 2013;230:407. [DOI] [PubMed]
    Kay J, Thadhani E, Samson L, Engelward B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair (Amst). 2019;83:102673. [DOI] [PubMed] [PMC]
    Giri PM, Banerjee A, Ghosal A, Layek B. Neuroinflammation in Neurodegenerative Disorders: Current Knowledge and Therapeutic Implications. Int J Mol Sci. 2024;25:3995. [DOI] [PubMed] [PMC]
    Gilmore SA, Phillips N, Liu KM, Houlé JD. Radiation-induced modulation of the microglial population in the normal and injured mature spinal cord. Exp Neurol. 2003;182:16979. [DOI] [PubMed]
    Hamada N. Noncancer Effects of Ionizing Radiation Exposure on the Eye, the Circulatory System and beyond: Developments made since the 2011 ICRP Statement on Tissue Reactions. Radiat Res. 2023;200:188216. [DOI] [PubMed]
    Martin CJ, Barnard M, Vocht Fd. Evaluation of risks of cardiovascular disease from radiation exposure linked to computed tomography scans in the UK. J Radiol Prot. 2024;44:011513. [DOI] [PubMed]
    Li X, Hirsch JA, Rehani MM, Yang K, Liu B. Effective Dose Assessment for Patients Undergoing Contemporary Fluoroscopically Guided Interventional Procedures. AJR Am J Roentgenol. 2020;214:15870. [DOI] [PubMed]
    Sanchez RM, Vano E, Fernández JM, Moreu M, Lopez-Ibor L. Brain radiation doses to patients in an interventional neuroradiology laboratory. AJNR Am J Neuroradiol. 2014;35:127680. [DOI] [PubMed] [PMC]
    Seguchi S, Saijou T, Ishikawa Y, Kojima T, Koyama S. Radiation Dose to Patients Undergoing X-ray Diagnosis, Treatment and Follow-up for Cerebral Aneurysms. No Shinkei Geka. 2015;43:4118. Japanese. [DOI] [PubMed]
    D'Alessio A, Strocchi S, Dalmasso F, Cannillo B, Matheoud R, Ponzetti A, et al. Effective and organ doses in patient undergoing interventional neuroradiology procedures: A multicentre study. Phys Med. 2024;122:103383. [DOI] [PubMed]
    Auvinen A, Cardis E, Blettner M, Moissonnier M, Sadetzki S, Giles G, et al. Diagnostic radiological examinations and risk of intracranial tumours in adults—findings from the Interphone Study. Int J Epidemiol. 2022;51:53746. [DOI] [PubMed] [PMC]
    Venneri L, Rossi F, Botto N, Andreassi MG, Salcone N, Emad A, et al. Cancer risk from professional exposure in staff working in cardiac catheterization laboratory: insights from the National Research Council's Biological Effects of Ionizing Radiation VII Report. Am Heart J. 2009;157:11824. [DOI] [PubMed]
    Vañó E, González L, Guibelalde E, Fernández JM, Ten JI. Radiation exposure to medical staff in interventional and cardiac radiology. Br J Radiol. 1998;71:95460. [DOI] [PubMed]
    Reeves RR, Ang L, Bahadorani J, Naghi J, Dominguez A, Palakodeti V, et al. Invasive Cardiologists Are Exposed to Greater Left Sided Cranial Radiation: The BRAIN Study (Brain Radiation Exposure and Attenuation During Invasive Cardiology Procedures). JACC Cardiovasc Interv. 2015;8:1197206. [DOI] [PubMed]
    Picano E, Vano E, Domenici L, Bottai M, Thierry-Chef I. Cancer and non-cancer brain and eye effects of chronic low-dose ionizing radiation exposure. BMC Cancer. 2012;12:157. [DOI] [PubMed] [PMC]
    Hamada N. Cancer and Non-Cancer Effects Following Ionizing Irradiation. Cancers (Basel). 2024;16:1141. [DOI] [PubMed] [PMC]
    Rühm W, Laurier D, Wakeford R. Cancer risk following low doses of ionising radiation - Current epidemiological evidence and implications for radiological protection. Mutat Res Genet Toxicol Environ Mutagen. 2022;873:503436. [DOI] [PubMed]
    Ma T, Li K, Sang W, Liu X, Luo Q, Peng Y, et al. Low-dose-rate induces more severe cognitive impairment than high-dose-rate in rats exposed to chronic low-dose γ-radiation. Front Public Health. 2024;12:1387330. [DOI] [PubMed] [PMC]
    Chow JCH, Ho JCS, Cheung KM, Johnson D, Ip BYM, Beitler JJ, et al. Neurological complications of modern radiotherapy for head and neck cancer. Radiother Oncol. 2024;194:110200. [DOI] [PubMed]
    Manenti G, Coppeta L, Kirev IV, Verno G, Garaci F, Magrini A, et al. Low-Dose Occupational Exposure to Ionizing Radiation and Cardiovascular Effects: A Narrative Review. Healthcare (Basel). 2024;12:238. [DOI] [PubMed] [PMC]
    Wang Q, Yin G, Huang J, Xi S, Qian F, Lee R, et al. Ionizing Radiation-Induced Brain Cell Aging and the Potential Underlying Molecular Mechanisms. Cells. 2021;10:3570. [DOI] [PubMed] [PMC]
    Baxter PS, Hardingham GE. Adaptive regulation of the brain's antioxidant defences by neurons and astrocytes. Free Radic Biol Med. 2016;100:14752. [DOI] [PubMed] [PMC]
    Mata-Garrido J, Tapia O, Casafont I, Berciano MT, Cuadrado A, Lafarga M. Persistent accumulation of unrepaired DNA damage in rat cortical neurons: nuclear organization and ChIP-seq analysis of damaged DNA. Acta Neuropathol Commun. 2018;6:68. [DOI] [PubMed] [PMC]
    Gomes-Leal W. Adult Hippocampal Neurogenesis and Affective Disorders: New Neurons for Psychic Well-Being. Front Neurosci. 2021;15:594448. [DOI] [PubMed] [PMC]
    Schmal Z, Isermann A, Hladik D, Toerne Cv, Tapio S, Rübe CE. DNA damage accumulation during fractionated low-dose radiation compromises hippocampal neurogenesis. Radiother Oncol. 2019;137:4554. [DOI] [PubMed]
    Oyefeso FA, Goldberg G, Opoku NYPS, Vazquez M, Bertucci A, Chen Z, et al. Effects of acute low-moderate dose ionizing radiation to human brain organoids. PLoS One. 2023;18:e0282958. [DOI] [PubMed] [PMC]
    Narasimhamurthy RK, Mumbrekar KD, Rao BSS. Effects of low dose ionizing radiation on the brain- a functional, cellular, and molecular perspective. Toxicology. 2022;465:153030. [DOI] [PubMed]
    Khan DZ, Lacasse MC, Khan R, Murphy KJ. Radiation Cataractogenesis: The Progression of Our Understanding and Its Clinical Consequences. J Vasc Interv Radiol. 2017;28:4129. [DOI] [PubMed]
    Alhasan AS, Aalam WA. Eye lens opacities and cataracts among physicians and healthcare workers occupationally exposed to radiation: A systematic review and meta-analysis. Saudi Med J. 2022;43:66577. [DOI] [PubMed] [PMC]
    Elmaraezy A, Morra ME, Mohammed AT, Al-Habaa A, Elgebaly A, Ghazy AA, et al. Risk of cataract among interventional cardiologists and catheterization lab staff: A systematic review and meta-analysis. Catheter Cardiovasc Interv. 2017;90:19. [DOI] [PubMed]
    Hulthén M, Tsapaki V, Karambatsakidou A. Estimating brain and eye lens dose for the cardiologist in interventional cardiology—are the dose levels of concern? Br J Radiol. 2024;97:1191201. [DOI] [PubMed]
    Domienik J, Bissinger A, Grabowicz W, Jankowski Ł, Kręcki R, Makowski M, et al. The impact of various protective tools on the dose reduction in the eye lens in an interventional cardiology-clinical study. Journal of radiological protection: official journal of the Society for Radiological Protection. 2016;36:30918. [DOI]
    Ainsbury EA, Dalke C, Hamada N, Benadjaoud MA, Chumak V, Ginjaume M, et al. Radiation-induced lens opacities: Epidemiological, clinical and experimental evidence, methodological issues, research gaps and strategy. Environ Int. 2021;146:106213. [DOI] [PubMed]
    Ainsbury EA, Barnard S, Bright S, Dalke C, Jarrin M, Kunze S, et al. Ionizing radiation induced cataracts: Recent biological and mechanistic developments and perspectives for future research. Mutat Res Rev Mutat Res. 2016;770:23861. [DOI] [PubMed]
    Hamada N, Azizova TV, Little MP. An update on effects of ionizing radiation exposure on the eye. Br J Radiol. 2020;93:20190829. [DOI] [PubMed] [PMC]
    The 2007 Recommendations of the International Commission on Radiological Protection. ICRP publication 103. Ann ICRP. 2007;37:1332. [PubMed] [PMC]
    Cahan WG, Woodard HQ, Higinbotham NL, Stewart FW, Coley BL. Sarcoma arising in irradiated bone: report of eleven cases. Cancer. 1998;82:834. [DOI] [PubMed]
    Durham AR, Tooker EL, Patel NS, Gurgel RK. Epidemiology and Risk Factors for Development of Sporadic Vestibular Schwannoma. Otolaryngol Clin North Am. 2023;56:41320. [DOI] [PubMed]
    Rubinstein AB, Reichenthal E, Borohov H. Radiation-induced schwannomas. Neurosurgery. 1989;24:92932. [DOI] [PubMed]
    Salvati M, Ciappetta P, Raco A, Capone R, Artico M, Santoro A. Radiation-induced schwannomas of the neuraxis. Report of three cases. Tumori. 1992;78:1436. [DOI] [PubMed]
    Sogg RL, Nikoskelainen E. Parotid carcinoma and posterior fossa schwannoma following irradiation. Report of a patient treated in infancy for benign ear disease. JAMA. 1977;237:2098100. [PubMed]
    Al-Mefty O, Topsakal C, Pravdenkova S, Sawyer JR, Harrison MJ. Radiation-induced meningiomas: clinical, pathological, cytokinetic, and cytogenetic characteristics. J Neurosurg. 2004;100:100213. [DOI] [PubMed]
    Sadetzki S, Flint-Richter P, Ben-Tal T, Nass D. Radiation-induced meningioma: a descriptive study of 253 cases. J Neurosurg. 2002;97:107882. [DOI] [PubMed]
    Strojan P, Popović M, Jereb B. Secondary intracranial meningiomas after high-dose cranial irradiation: report of five cases and review of the literature. Int J Radiat Oncol Biol Phys. 2000;48:6573. [DOI] [PubMed]
    Beller AJ, Feinsod M, Sahar A. The possible relationship between small dose irradiation to the scalp and intracranial meningiomas. Neurochirurgia (Stuttg). 1972;15:13543. [DOI] [PubMed]
    Gosztonyi G, Slowik F, Pásztor E. Intracranial meningiomas developing at long intervals following low-dose X-ray irradiation of the head. J Neurooncol. 2004;70:5965. [DOI] [PubMed]
    Coca-Pelaz A, Mäkitie AA, Strojan P, Corry J, Eisbruch A, Beitler JJ, et al. Radiation-Induced Sarcomas of the Head and Neck: A Systematic Review. Adv Ther. 2021;38:90108. [DOI] [PubMed] [PMC]
    Omran AR, Shore RE, Markoff RA, Friedhoff A, Albert RE, Barr H, et al. Follow-up study of patients treated by X-ray epilation for tinea capitis: psychiatric and psychometric evaluation. Am J Public Health. 1978;68:5617. [DOI] [PubMed] [PMC]
    Ron E, Modan B, Jr JDB, Alfandary E, Stovall M, Chetrit A, et al. Tumors of the brain and nervous system after radiotherapy in childhood. N Engl J Med. 1988;319:10339. [DOI] [PubMed]
    Memon A, Rogers I, Paudyal P, Sundin J. Dental X-Rays and the Risk of Thyroid Cancer and Meningioma: A Systematic Review and Meta-Analysis of Current Epidemiological Evidence. Thyroid. 2019;29:157293. [DOI] [PubMed]
    Preston-Martin S. Descriptive epidemiology of primary tumors of the spinal cord and spinal meninges in Los Angeles County, 1972-1985. Neuroepidemiology. 1990;9:10611. [DOI] [PubMed]
    Preston-Martin S, Mack W, Henderson BE. Risk factors for gliomas and meningiomas in males in Los Angeles County. Cancer Res. 1989;49:613743. [PubMed]
    Hauptmann M, Byrnes G, Cardis E, Bernier M, Blettner M, Dabin J, et al. Brain cancer after radiation exposure from CT examinations of children and young adults: results from the EPI-CT cohort study. Lancet Oncol. 2023;24:4553. [DOI] [PubMed]
    Finkelstein MM. Is brain cancer an occupational disease of cardiologists? Can J Cardiol. 1998;14:13858. [PubMed]
    Roguin A, Goldstein J, Bar O. Brain tumours among interventional cardiologists: a cause for alarm? Report of four new cases from two cities and a review of the literature. EuroIntervention. 2012;7:10816. [DOI] [PubMed]
    Roguin A, Goldstein J, Bar O, Goldstein JA. Brain and neck tumors among physicians performing interventional procedures. Am J Cardiol. 2013;111:136872. [DOI] [PubMed]
    Coselli JS, Preventza O. In memoriam: Edward B. Diethrich, MD, (1935–2017). Tex Heart Inst J. 2017; 44:1646. [DOI]
    Invisible Impact: The Risk of Ionizing Radiation on Cath Lab Staff [Internet]. OrsifOrg; c2020 [cited 2024 August 18]. Available from: https://www.youtube.com/watch?v=rXgt0bF3GJM
    Li J, Zhang X, Liu J, Su C, Cui J, Yang L, et al. Case report: Low-dose radiation-induced meningioma with a short latency period. Front Oncol. 2024;14:1413610. [DOI] [PubMed] [PMC]
    Lopes J, Baudin C, Leuraud K, Klokov D, Bernier M. Ionizing radiation exposure during adulthood and risk of developing central nervous system tumors: systematic review and meta-analysis. Sci Rep. 2022;12:16209. [DOI] [PubMed] [PMC]
    Lohmann DR, Gallie BL. Retinoblastoma. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, editors. GeneReviews®. Seattle (WA): University of Washington, Seattle; 1993–2024.
    Bunin GR, Felice MA, Davidson W, Friedman DL, Shields CL, Maidment A, et al. Medical radiation exposure and risk of retinoblastoma resulting from new germline RB1 mutation. Int J Cancer. 2011;128:2393404. [DOI] [PubMed] [PMC]
    Shakeel O, Pace N, Chambers TM, Scheurer ME, Ganguly AA, Lupo PJ, et al. Medical radiation exposure and risk of sporadic retinoblastoma. Pediatr Blood Cancer. 2020;67:e28633. [DOI] [PubMed]
    Theocharopoulos N, Damilakis J, Perisinakis K, Manios E, Vardas P, Gourtsoyiannis N. Occupational exposure in the electrophysiology laboratory: quantifying and minimizing radiation burden. Br J Radiol. 2006;79:64451. [DOI] [PubMed]
    Andreassi MG, Borghini A, Vecoli C, Piccaluga E, Guagliumi G, Greco MD, et al. Reproductive outcomes and Y chromosome instability in radiation-exposed male workers in cardiac catheterization laboratory. Environ Mol Mutagen. 2020;61:3618. [DOI] [PubMed]
    Jahng JWS, Little MP, No HJ, Jr BWL, Wu JC. Consequences of ionizing radiation exposure to the cardiovascular system. Nat Rev Cardiol. 2024. [DOI] [PubMed]
    Liu M, Ding C, Li Z, Yi R, Ma L, Ou X, et al. Multiple exposures to low-dose ionizing radiation induced the initiation and progression of pro-atherosclerotic phenotypes in mice and vascular endothelial cell damage. Sci Prog. 2024;107:368504241228668. [DOI] [PubMed] [PMC]
    Zheng X, Liu Z, Bin Y, Wang J, Rao X, Wu G, et al. Ionizing radiation induces vascular smooth muscle cell senescence through activating NF-κB/CTCF/p16 pathway. Biochim Biophys Acta Mol Basis Dis. 2024;1870:166994. [DOI] [PubMed]
    Little MP, Azizova TV, Bazyka D, Bouffler SD, Cardis E, Chekin S, et al. Systematic review and meta-analysis of circulatory disease from exposure to low-level ionizing radiation and estimates of potential population mortality risks. Environ Health Perspect. 2012;120:150311. [DOI] [PubMed] [PMC]
    Little MP, Azizova TV, Richardson DB, Tapio S, Bernier M, Kreuzer M, et al. Ionising radiation and cardiovascular disease: systematic review and meta-analysis. BMJ. 2023;380:e072924. [DOI] [PubMed] [PMC]
    Zablotska LB, Little MP, Hamada N. Revisiting an Inverse Dose-Fractionation Effect of Ionizing Radiation Exposure for Ischemic Heart Disease: Insights from Recent Studies. Radiat Res. 2024;202:806. [DOI] [PubMed]
    Cha ES, Zablotska LB, Bang YJ, Lee WJ. Occupational radiation exposure and morbidity of circulatory disease among diagnostic medical radiation workers in South Korea. Occup Environ Med. 2020;77:75260. [DOI] [PubMed]
    Hinksman CA, Haylock RGE, Gillies M. Cerebrovascular Disease Mortality after occupational Radiation Exposure among the UK National Registry for Radiation Workers Cohort. Radiat Res. 2022;197:45970. [DOI] [PubMed]
    Andreassi MG, Piccaluga E, Gargani L, Sabatino L, Borghini A, Faita F, et al. Subclinical carotid atherosclerosis and early vascular aging from long-term low-dose ionizing radiation exposure: a genetic, telomere, and vascular ultrasound study in cardiac catheterization laboratory staff. JACC Cardiovasc Interv. 2015;8:61627. [DOI] [PubMed]
    Schneider CV, Schneider KM, Teumer A, Rudolph KL, Hartmann D, Rader DJ, et al. Association of Telomere Length With Risk of Disease and Mortality. JAMA Intern Med. 2022;182:291300. [DOI] [PubMed] [PMC]
    Fyhrquist F, Saijonmaa O, Strandberg T. The roles of senescence and telomere shortening in cardiovascular disease. Nat Rev Cardiol. 2013;10:27483. [DOI] [PubMed]
    Andreassi MG. Low-doses ionizing radiation exposure: an emerging causal risk factor for cardiovascular disease. Explor Cardiol. 2023;1:1417. [DOI]
    Srivastava T, Chirikova E, Birk S, Xiong F, Benzouak T, Liu JY, et al. Exposure to Ionizing Radiation and Risk of Dementia: A Systematic Review and Meta-Analysis. Radiat Res. 2023;199:490505. [DOI] [PubMed] [PMC]
    Dauer LT, Walsh L, Mumma MT, Cohen SS, Golden AP, Howard SC, et al. Moon, Mars and Minds: Evaluating Parkinson’s disease mortality among U.S. radiation workers and veterans in the million person study of low-dose effects. Z Med Phys. 2024;34:10010. [DOI] [PubMed] [PMC]
    Acharya MM, Baulch JE, Klein PM, Baddour AAD, Apodaca LA, Kramár EA, et al. New Concerns for Neurocognitive Function during Deep Space Exposures to Chronic, Low Dose-Rate, Neutron Radiation. eNeuro. 2019;6:ENEURO.009419.2019. [DOI] [PubMed] [PMC]
    Kempf SJ, Moertl S, Sepe S, Toerne Cv, Hauck SM, Atkinson MJ, et al. Low-dose ionizing radiation rapidly affects mitochondrial and synaptic signaling pathways in murine hippocampus and cortex. J Proteome Res. 2015;14:205564. [DOI] [PubMed]
    Borghini A, Vecoli C, Mercuri A, Carpeggiani C, Piccaluga E, Guagliumi G, et al. Low-Dose Exposure to Ionizing Radiation Deregulates the Brain-Specific MicroRNA-134 in Interventional Cardiologists. Circulation. 2017;136:25168. [DOI] [PubMed]
    Tonacci A, Baldus G, Corda D, Piccaluga E, Andreassi M, Cremonesi A, et al. Olfactory non-cancer effects of exposure to ionizing radiation in staff working in the cardiac catheterization laboratory. Int J Cardiol. 2014;171:4613. [DOI] [PubMed]
    Marazziti D, Tomaiuolo F, Dell'Osso L, Demi V, Campana S, Piccaluga E, et al. Neuropsychological Testing in Interventional Cardiology Staff after Long-Term Exposure to Ionizing Radiation. J Int Neuropsychol Soc. 2015;21:6706. [DOI] [PubMed]
    Schratt GM, Tuebing F, Nigh EA, Kane CG, Sabatini ME, Kiebler M, et al. A brain-specific microRNA regulates dendritic spine development. Nature. 2006;439:2839. [DOI] [PubMed]
    Baby N, Alagappan N, Dheen ST, Sajikumar S. MicroRNA-134-5p inhibition rescues long-term plasticity and synaptic tagging/capture in an Aβ(1–42)-induced model of Alzheimer's disease. Aging Cell. 2020;19:e13046. [DOI] [PubMed] [PMC]
    Loganovsky KN, Vasilenko ZL. Depression and ionizing radiation. Probl Radiac Med Radiobiol. 201320019. [PubMed]
    Zonenberg A, Leoniak M, Zarzycki W. The effect of Chernobyl accident on the development of non malignant diseases. Endokrynol Pol. 2006;57:3844. [PubMed]
    Ventriglio A, Bellomo A, Gioia Id, Sabatino DD, Favale D, Berardis DD, et al. Environmental pollution and mental health: a narrative review of literature. CNS Spectr. 2021;26:5161. [DOI] [PubMed]
    Dickerson AS, Wu AC, Liew Z, Weisskopf M. A Scoping Review of Non-Occupational Exposures to Environmental Pollutants and Adult Depression, Anxiety, and Suicide. Curr Environ Health Rep. 2020;7:25671. [DOI] [PubMed] [PMC]
    Loganovsky KN, Bomko MO, Abramenko IV, Kuts KV, Belous NI, Masiuk SV, et al. NEUROPSYCHOBIOLOGICAL MECHANISMS OF AFFECTIVE AND COGNITIVE DISORDERS IN THE CHORNOBYL CLEAN-UP WORKERS TAKING INTO ACCOUNT THE SPECIFIC GENE POLYMORPHISMS. Probl Radiac Med Radiobiol. 2018;23:373409. Ukrainian. [DOI] [PubMed]
    Loganovsky KN, Masiuk SV, Buzunov VA, Marazziti D, Voychulene YS. Radiation Risk Analysis of Neuropsychiatric Disorders in Ukrainian Chornobyl Catastrophe Liquidators. Front Psychiatry. 2020;11:553420. [DOI] [PubMed] [PMC]
    Xu L, Huang H, Liu T, Yang T, Yi X. Exposure to X-rays Causes Depression-like Behaviors in Mice via HMGB1-mediated Pyroptosis. Neuroscience. 2022;481:99110. [DOI] [PubMed]
    Yen PN, Lin I, Chang WP, Wang J, Chang T, Kuo K, et al. Risk factors of depression after prolonged low-dose rate environmental radiation exposure. Int J Radiat Biol. 2014;90:85966. [DOI] [PubMed]
    Andreassi MG, Piccaluga E, Guagliumi G, Greco MD, Gaita F, Picano E. Occupational Health Risks in Cardiac Catheterization Laboratory Workers. Circ Cardiovasc Interv. 2016;9:e003273. [DOI] [PubMed]
    Correia MJ, Hellies A, Andreassi MG, Ghelarducci B, Picano E. Lack of radiological awareness among physicians working in a tertiary-care cardiological centre. Int J Cardiol. 2005;103:30711. [DOI] [PubMed]
    Giaccardi M, Anselmino M, Greco MD, Mascia G, Perini AP, Mascia P, et al. Radiation awareness in an Italian multispecialist sample assessed with a web-based survey. Acta Cardiol. 2021;76:30711. [DOI] [PubMed]
    Doyen B, Maurel B, Hertault A, Vlerick P, Mastracci T, Herzeele IV, et al. Radiation Safety Performance is More than Simply Measuring Doses! Development of a Radiation Safety Rating Scale. Cardiovasc Intervent Radiol. 2020;43:133141. [DOI] [PubMed]
    Jr JWH, Ferrari VA, Bengel FM, Bergersen L, Chambers CE, Einstein AJ, et al. 2018 ACC/HRS/NASCI/SCAI/SCCT Expert Consensus Document on Optimal Use of Ionizing Radiation in Cardiovascular Imaging—Best Practices for Safety and Effectiveness, Part 2: Radiological Equipment Operation, Dose-Sparing Methodologies, Patient and Medical Personnel Protection: A Report of the American College of Cardiology Task Force on Expert Consensus Decision Pathways. J Am Coll Cardiol. 2018;71:282955. [DOI] [PubMed]
    Sarkozy A, Potter TD, Heidbuchel H, Ernst S, Kosiuk J, Vano E, et al.; ESC Scientific Document Group. Occupational radiation exposure in the electrophysiology laboratory with a focus on personnel with reproductive potential and during pregnancy: A European Heart Rhythm Association (EHRA) consensus document endorsed by the Heart Rhythm Society (HRS). Europace. 2017;19:190922. [DOI] [PubMed]
    Jr JWH, Balter S, Brinker JA, Kern MJ, Klein LW, Lindsay BD, et al. ACCF/AHA/HRS/SCAI clinical competence statement on physician knowledge to optimize patient safety and image quality in fluoroscopically guided invasive cardiovascular procedures: a report of the American College of Cardiology Foundation/American Heart Association/American College of Physicians Task Force on Clinical Competence and Training. Circulation. 2005;111:51132. [DOI] [PubMed]
    Bahar AR, Khanal R, Hamza M, Goru RK, Shafiq A, Haider MZ, et al. Assessing the Efficacy of RADPAD Protection Drape in Reducing Radiation Exposure to Operators in the Cardiac Catheterization Laboratory: A Systematic Review and Meta-Analysis. Cureus. 2024;16:e59215. [DOI] [PubMed] [PMC]
    Valuckiene Z, Jurenas M, Cibulskaite I. Ionizing radiation exposure in interventional cardiology: current radiation protection practice of invasive cardiology operators in Lithuania. J Radiol Prot. 2016;36:695708. [DOI] [PubMed]
    Kidoń J, Polaczek-Grelik K, Żurek P, Wojakowski W, Ochala A. Advances in Interventional Cardiology/Postępy w Kardiologii Interwencyjnej. Postepy Kardiol Interwencyjnej. 2021;17:298304. [DOI] [PubMed] [PMC]
    Kuon E, Birkel J, Schmitt M, Dahm JB. Radiation exposure benefit of a lead cap in invasive cardiology. Heart. 2003;89:120510. [DOI] [PubMed] [PMC]
    Qazi E, Ursani A, Patel N, Kennedy SA, Bassett P, Jaberi A, et al. Operator Intracranial Dose Protection During Fluoroscopic-Guided Interventions. Cardiovasc Intervent Radiol. 2023;46:94352. [DOI] [PubMed]
    Kirkwood ML, Arbique GM, Guild JB, Zeng K, Xi Y, Rectenwald J, et al. Radiation brain dose to vascular surgeons during fluoroscopically guided interventions is not effectively reduced by wearing lead equivalent surgical caps. J Vasc Surg. 2018;68:56771. [DOI] [PubMed]
    Sattar Y, Sengodan PM, Cheema MS, Javed N, Ashraf S, Fakhra S, et al. Lead Cap Use in Interventional Cardiology: Time to Protect Our Head in the Cardiac Catheterisation Laboratory? Interv Cardiol. 2023;18:e18. [DOI] [PubMed] [PMC]
    Larsson MEV, Jonasson PI, Apell PS, Kearney PP, Lundh CJ. Evaluation of novel radiation protection devices during radiologically guided interventions. CVIR Endovasc. 2024;7:18. [DOI] [PubMed] [PMC]
    Ramanan B, Pizano A, Solano A, Gonugunta AS, Timaran CH, Siah M, et al. The addition of a leaded arm sleeve to leaded aprons further decreases operator upper outer quadrant chest wall radiation dose during fluoroscopically guided interventions. J Vasc Surg. 2024;79:94853. [DOI] [PubMed]
    Ramos-Avasola S, Ponce L, Leon K, Cuellar-Fritis C, Querales M. Efficacy of radiation attenuating caps in reducing radiation doses received at the cerebral level in interventional physicians: a systematic review. J Radiol Prot. 2024;44:031001. [DOI] [PubMed]
    Gutierrez-Barrios A, Angulo-Pain E, Noval-Morillas I, Cañadas-Pruaño D, Lastra IAdl, Gheorghe L, et al. The radioprotective effect of the Cathpax® AIR cabin during interventional cardiology procedures. Catheter Cardiovasc Interv. 2021;98:E52330. [DOI] [PubMed]
    Domienik-Andrzejewska J, Mirowski M, Jastrzębski M, Górnik T, Masiarek K, Warchoł I, et al. Occupational exposure to physicians working with a Zero-Gravity™ protection system in haemodynamic and electrophysiology labs and the assessment of its performance against a standard ceiling suspended shield. Radiat Environ Biophys. 2022;61:293300. [DOI] [PubMed] [PMC]
    Debreceni D, Janosi K, Bocz B, Turcsan M, Lukacs R, Simor T, et al. Zero fluoroscopy catheter ablation for atrial fibrillation: a systematic review and meta-analysis. Front Cardiovasc Med. 2023;10:1178783. [DOI] [PubMed] [PMC]
    Enriquez A, Sadek M, Hanson M, Yang J, Matos CD, Neira V, et al. Feasibility, Efficacy, and Safety of Fluoroless Ablation of VT in Patients With Structural Heart Disease. JACC Clin Electrophysiol. 2024;10:1287300. [DOI] [PubMed]
    Lehar F, Szegedi N, Hejc J, Jez J, Soucek F, Kulik T, et al. Randomized comparison of atrioventricular node re-entry tachycardia and atrial flutter catheter ablation with and without fluoroscopic guidance: ZeroFluoro study. Europace. 2022;24:163644. [DOI] [PubMed]
    Anselmino M, Sillano D, Casolati D, Ferraris F, Scaglione M, Gaita F. A new electrophysiology era: zero fluoroscopy. J Cardiovasc Med (Hagerstown). 2013;14:2217. [DOI] [PubMed]
    Madder RD, Seth M, Frazier K, Dixon S, Karve M, Collins J, et al. Statewide Initiative to Reduce Patient Radiation Doses During Percutaneous Coronary Intervention. Circ Cardiovasc Interv. 2024;17:e013502. [DOI] [PubMed]
    Shimoda T, D'Oria M, Kuno T, Heindel P, Lepidi S, Hussain MA, et al. Comparative Effectiveness of Intravascular Ultrasound Versus Angiography in Abdominal and Thoracic Endovascular Aortic Repair: Systematic Review and Meta-Analysis. Am J Cardiol. 2024;223:8191. [DOI] [PubMed]
    Stevenson A, Kirresh A, Ahmad M, Candilio L. Robotic-Assisted PCI: The Future of Coronary Intervention? Cardiovasc Revasc Med. 2022;35:1618. [DOI] [PubMed]