• Open Access
    Review

    Viral infections in cardiometabolic risk and disease between old acquaintances and new enemies

    Cristina Vassalle *

    Explor Cardiol. 2023;1:148–179 DOI: https://doi.org/10.37349/ec.2023.00014

    Received: July 10, 2023 Accepted: September 28, 2023 Published: December 27, 2023

    Academic Editor: Karina Wierzbowska-Drabik, Medical University of Lodz, Poland

    This article belongs to the special issue Environmental Cardiology

    Abstract

    Atherosclerosis is a chronic disease, characterized by chronic inflammation, endothelial dysfunction, and lipid deposition in the vessel. Although many major, well-identified risk factors for atherosclerosis [e.g., hyperlipidemia, hypertension, type 2 diabetes (T2D), smoking habit, and obesity] explain a lot about the risk, there is a considerable number of patients who develop atherosclerotic damage and undergo adverse events without presenting any of these established modifiable risk factors. This observation has stimulated an urgent need to expand knowledge towards the identification of additional, less established risk factors that may help in the assessment of risk and fill the gap of knowledge in the cardiovascular (CV) setting. Among them, the hypothesis of a possible relationship between viral infectious agents and atherosclerosis has risen since the early 1900s. However, there is still a great deal of debate regarding the onset and progression of CV disease in relation to the roles of the pathogens (as active inducers or bystanders), host genomic counterparts, and environmental triggers, affecting both virus abundance and the composition of viral communities. Accordingly, the aim of this review is to discuss the current state of knowledge on infectious agents in the atherosclerotic process, with particular focus on two environmental-related viruses, as examples of familiar (influenza) and unfamiliar [severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)] disease triggers.

    Keywords

    Atherosclerosis, type 2 diabetes, viral infection, severe acute respiratory syndrome coronavirus-2, cardiometabolic risk, cardiometabolic disease, ischemic heart disease, non-traditional cardiovascular risk factors

    Introduction

    Atherosclerosis belongs to the group of chronic degenerative diseases, conditions that affect the subject for a long period or for a lifetime with a progressive deterioration over time, degenerating and causing multi-organ and multi-system damage, which may lead to death [1]. The atherosclerotic process is characterized by chronic inflammation, endothelial dysfunction, and lipid deposition in the vessel [1]. Many major modifiable risk factors for atherosclerosis [e.g., hyperlipidemia, hypertension, type 2 diabetes (T2D), smoking habit, and obesity] explain a lot about the risk, and associated therapies adequately account for the significant reduction in cardiovascular (CV) disability and mortality in high-income countries [2]. Nonetheless, a considerable subset of patients remains, who develop atherosclerotic damage and undergo adverse events, exhibiting distinct risk profiles that either do not include any of these established modifiable risk factors or are poorly explained by traditional risk factors [3, 4]. This observation has stimulated an urgent need to expand our knowledge of less established risk determinants, whose identification may supplement the burden of the risk factors and/or evidence of special causal relevance in specific patient subsets [5, 6]. Among them, a possible relationship between viral infectious agents and atherosclerosis has been proposed since the early 1900s, when Osler included acute infection among the main factors that may cause atherosclerosis [acute infection, stress, intoxication (as T2D, obesity, or smoking habit)], while Frothingham identified the sclerosis of aging as a sum of lesions induced by infectious and metabolic toxins [7, 8]. However, it was in the late 1970s that the first convincing proof-of-concept was obtained, demonstrating that the Marek virus (an avian herpes virus) is able to induce fibroproliferative, lipid-laden atherosclerotic damage, and to generate a thrombus in normocholesterolemic chickens [9]. Since then, several seroepidemiological studies have shown that lasting infections (bacteria or viruses) may be linked to the onset and development of atherosclerotic lesion [10].

    In general, the identification of direct and indirect mechanisms that microorganisms use to elicit chronic inflammation, the presence of an infectious agent in human atherosclerotic tissue, as well as the evidence of atherosclerosis quickening following infection in animal models of atherosclerosis, and the number of infectious agents found associated with CV disease (CVD; e.g., Chlamydia pneumoniae [11], periodontal disease infections [12], human immunodeficiency virus (HIV) [13], cytomegalovirus (CMV) [14], hepatitis C virus (HCV) [15], Helicobacter pylori [16], herpes simplex virus (HSV) types 1 and 2 [17] hepatitis A virus [18], hepatitis B virus (HBV) [19] and gut microbiota [20]) all suggest biological plausibility.

    However, there is still a great debate regarding the onset and progression of CVD on the roles of the pathogens as active inducers or bystanders, host genomic counterparts, and environmental triggers affecting virus abundance and the composition of viral communities. Accordingly, this review aims to address the current state of knowledge about infectious agents and their role in atherosclerosis, including some examples and a special emphasis on two environmental-related viruses as examples of old acquaintances (influenza) and new enemies [severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)].

    Action modes of infectious agents: differences and similarities

    Microorganisms, in general, use a number of common direct or indirect strategies to trigger immunological and oxidative/inflammatory responses [e.g., increased expression of pro-inflammatory and prothrombotic cytokines including tumor necrosis factor (TNF)-α, interferon (INF)-γ, interleukin (IL)-6, IL-1β, proatherogenic factors as TLR2, TLR4, adhesion molecules, lectin-like oxidized low-density lipoprotein (oxLDL) receptor-1 (LOX-1), heat shock protein 60 (Hsp60) and monocyte chemoattractant protein-1 (MCP-1)], generate macrophage-derived foam cell generation, and increase proliferation while inhibiting apoptosis of smooth muscle cells (SMC) [21]. A direct effect of an infectious agent is represented by its ability to infect vascular cells, the detection of the organism within the plaque, and the acceleration of lesion formation following infection in experimental atherosclerosis models, as well as the demonstration of the efficacy of anti-infective therapies either in the reduction of atherosclerotic lesions or in the improvement of CV outcomes [21]. In contrast, an indirect action of infectious agents caused by infection and inflammation at a non-vascular site can accelerate atherosclerosis. Compliance with these criteria defines the degree of the individual infectious agent’s function in the atherosclerotic process, ranging from stronger to weaker [21].

    Moreover, each specific infectious agent may possess specific peculiar mechanisms for acting on the progression of atherosclerotic lesions. For example, Helicobacter pylori induced gastritis (with increased gastric juice pH and/or decreased ascorbic acid) and folate inadequate status [22], which in turn inhibits the methionine synthase with elevation of homocysteine, causing adverse effects on endothelial function and promoting atherosclerosis [22]. Specific inflammatory/atherogenic gene expression pathways in the aortic tissue of apolipoprotein E (apoE)−/− mice were induced by exposure to different pathogens, such as C. pneumoniae and P. gingivalis [23]. However, some differences were observed: P. gingivalis may reduce mitochondrial expression, glucose metabolism, and peroxisome proliferator-activated receptor (PPAR) pathways, whereas C. pneumoniae may, on the other hand, increase mitochondria expression, lipid metabolism, carbohydrate and aminoacid metabolism, and PPAR pathways [23].

    Some examples of action modalities (direct and/or indirect effects and identification of the agent in the atherosclerotic plaque) of common viruses, which have been related to atherosclerosis onset and development, are discussed below and reported in Table 1.

    Main common and specific virus mechanisms and complications related to the increased risk of atherosclerosis

    Infectious agentMain mechanisms and pathways involvedMain CV complications
    HIV

    - Presence in the atherosclerotic lesion

    - Systemic inflammation/oxidative stress

    - Immunity (CD4+ cell depletion)

    - Opportunistic infections

    - AMI

    - High prevalence of traditional CVD risk factors

    - Carotid atherosclerosis

    - Carotid stiffness

    - HF

    - Myocardial fibrosis

    - T2D

    - cART-related adverse cardiometabolic effects

    HSV-1 and -2

    - Presence in the atherosclerotic lesion

    - Systemic inflammation/oxidative stress

    - Endothelial dysfunction

    - Apoptosis

    - Procoagulant effects

    - CV mortality

    - CAD

    - AMI

    - Stroke

    - Carotid atherosclerosis

    CMV

    - Presence in the atherosclerotic lesion

    - Systemic inflammation/oxidative stress

    - Host immune response

    - Endothelial dysfunction

    - Lipid dysregulation and lipid deposition, vascular SMC proliferation and migration

    - Procoagulant effects and increased prothrombotic risk

    - Carotid atherosclerosis

    - CAD

    - AMI

    - Stroke

    - T2D

    HCV

    - Presence in the endothelial cells and atherosclerotic lesion

    - Systemic and local arterial inflammation/oxidative stress

    - Host immune response

    - Liver iron deposition

    - Metabolic disarrangement

    - Cryoglobulinemia

    - CV mortality

    - Carotid atherosclerosis

    - CAD

    - Dysrhythmias

    - HF

    - AMI

    - Cerebrovascular disease

    - Myocarditis and Cardiomyopathies

    - T2D

    Influenza

    - Presence in the endothelial cells and atherosclerotic lesions

    - Systemic and local arterial inflammation/oxidative stress

    - Hypercoagulability

    - Fever, tachycardia, hypoxia-induced injury

    - CV mortality

    - AMI

    - T2D

    SARS-CoV-2

    - Direct viral entry through ACE2 receptor (e.g., cardiomyocytes, fibroblasts, endothelial cells)

    - Systemic inflammation/cytokine storm

    - Hypercoagulability, thrombosis

    - Local inflammation (myocardial inflammation-myocarditis, cardiac fibrosis, myocyte apoptosis)

    - Plaque destabilization

    - Stress, anxiety, psycho-emotional status

    - Hypoxia-induced injury (myocardial oxygen demand/supply mismatch)

    - Endothelial dysfunction

    - Catecholamine stress response

    - HF

    - Ventricular hypertrophy

    - Ventricular dilation

    - AMI

    - Fibrosis

    - Myocarditis

    - T2D

    Display full size

    CAD: coronary artery disease; AMI: acute myocardial infarction; ACE: angiotensin-converting enzyme; cART: combined antiretroviral therapy; HF: heart failure

    HIV

    The HIV was first isolated and identified in the early 1980s. It targets and damages especially the immune system: at the end of 2021, it has been estimated, indeed, that more than 38 million people were infected by HIV, which also induced the deaths of more than 40 million people around the world [24]. Although the development of therapies has contributed to longer life expectancy in patients who have benefited from them, however, there has been an increase in comorbidities, so that atherosclerotic CVD currently represents one of the major causes of morbidity and mortality for people living with HIV (PLWH) [25].

    Among the underlying mechanisms linking HIV and atherosclerosis, it is well known that the activity of monocytes and macrophages, as well as macrophages cholesterol mechanisms, are dysregulated by HIV infection. These cells, which play a critical role in the development of the plaque in the subendothelial layer, support, indeed, HIV replication, and remain chronically infected [26]. Moreover, HIV Nef protein, which promotes viral replication and immune escape, may directly affect endothelial functions and gene expression in human pulmonary artery endothelial cells, reducing endothelium-dependent vasorelaxation in porcine pulmonary arteries [27]. Additionally, in vitro studies reveal a certain permissiveness for HIV by endothelial cells, which thus could serve as a viral reservoir [28].

    Some viral proteins, such as Tat, Nef, Vpr, and glycoprotein 120 (gp120), are involved in indirect HIV effects, such as elevation of pro-inflammatory cytokines and oxidative stress [29, 30]. In particular, Tat, which controls HIV transcription, is able to induce not only oxidative stress [reactive oxygen species (ROS) generation, lipid peroxidation], but also the expression of vascular cell adhesion molecule (VCAM)-1 and intercellular adhesion molecule 1 (ICAM-1), as well as to lower antioxidants (e.g., glutathione-GSH) [3034]. With regards to Nef protein, it increases superoxide release, also by decreasing endothelial nitric oxide (NO) synthase (eNOS) expression and NO release [25, 3537], while Vpr protein promotes cell-cycle arrest, induces DNA damage and apoptosis, and modulates nuclear factor-kappa B (NF-κB) activity; Vpr is also able to reactivate viral production in latently infected cells by releasing ROS and the pro-inflammatory cytokine IL-6 [3840]. Instead, gp120, a surface protein that facilitates HIV entry into the host cell, may promote apoptosis and induce the release of endothelin-1 (ET-1, endothelial vasoconstrictor peptide) [4143]. Recent data also support the role of HIV-1 infection as an activator of the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome, which contributes to the risk of developing atherosclerosis [44]. Moreover, HIV-related immune dysregulation may also be proatherogenic [45], while HIV itself may affect the adipose tissue: adipocytes may, in fact, be involved in the immune response to HIV, act as HIV reservoirs, produce proinflammatory molecules and alter the lipid profile (e.g., enhancing the risk of hypertriglyceridemia); HIV can also be responsible for microbiota dysbiosis and microbial translocation from the intestinal tract into the blood (with chronic immune activation, hypercoagulability, and prothrombotic effects) [4650]. Furthermore, HIV has been identified among diabetogenic viruses, as Nef protein inhibits glucose uptake after exposure in adipocytes [by interfering with glucose transport 4 (GLUT4)] and contributes to insulin resistance (IR) [51]. Although PLWH can nowadays achieve a near-normal life expectancy, mainly thanks to the development of highly efficient cART, these pharmacological tools may, however, contribute to enhancing endothelial dysfunction and CV disease adverse events (e.g., exacerbation of CV risk factors, inflammation, and leptin-mediated reduction of NO bioavailability), although this applies more to the old drugs than to the new ones [5254].

    Subjects affected by HIV show a prevalence of CV risk factors, and higher risk and occurrence of CAD and heart failure (HF) [55]. In particular, AMI is the most common clinical complication in HIV patients, with a higher incidence compared to the general population [56, 57]. The pathogenesis of AMI during HIV infection is multifactorial and primarily influenced by the interaction between host CV risk factors, together with HIV infection and ART [57]. Male gender, viral load of HIV, low CD4 count, higher CD8 count, and types of ART are indeed among the risk factors linked to AMI in HIV patients [57]. Moreover, CV mortality, carotid artery atherosclerosis as well as increased carotid vessel wall stiffness, HF, and myocardial fibrosis have been associated with HIV [58, 59].

    Experimental models also suggest that HIV may accelerate the atherosclerotic process, thus underlying the importance of caspase-1 and monocyte/macrophage activation and IL-18 elevation in HIV atherogenesis [60, 61]. Moreover, studies demonstrated that LOX-1 and VCAM-1 gene expression are increased in aorta as well as soluble ICAM-1(sICAM-1) blood levels in HIV-1 transgenic rats, suggesting HIV infection as a source and promoter of endothelial dysfunction and accelerated atherosclerosis [62].

    HSV (human herpesvirus 1 and 2)

    HSV is responsible for a very common infection affecting the mouth, genital area, or other parts of the body. The relationship between HSV and atherosclerosis has been reported since the beginning of the ‘90s, although it is still greatly debated [63]. Both types of HSV can be either present or absent in atherosclerotic plaques [6466] and HSV-1 was not found in normal artery sites [67]. Moreover, HSV-infected endothelial cells express the adhesion molecule granule membrane protein-140 (GMP140), which may contribute to the recruitment and migration of blood cells to initiate and develop atherosclerotic lesions [68]. HSV infection may increase the expression of LOX-1 and the uptake of ox-LDL hence exacerbating cellular apoptosis [69, 70]. This infection also activates procoagulant changes, alters lipid metabolism, and promotes inflammation [7175]: experimental data indicate that atheroma development is accelerated in infected apoE–/– mice compared to control uninfected apoE–/– mice (24-week period), effect that may be reduced by antiviral drugs [72].

    A meta-analysis (17 studies) also suggests that HSV-1 and HSV-2 infection could increase the risk of atherosclerosis (considering myocardial ischemia, and other types of atherosclerosis as stroke and carotid atherosclerosis) [17], while, in another study based on 14,415 subjects, HSV-2, rather than HSV-1, was associated with premature CVD [76].

    CMV (human CMV, human herpesvirus 5)

    CMV, part of the herpesvirus family, causes a widespread and very common infection, characterized by a wide range of severity (from no symptoms to fever and fatigue to severe complications in the eye, brain, or other organs). This virus was found to be expressed in carotid plaques and it may contribute to the inflammatory response in plaques via enhanced infiltration of CD68 and CD3 cells [77]. CMV infection elicits inflammatory and immune responses [e.g., IL-6, high-sensitivity C-reactive protein (CRP), fibrinogen, and secretory phospholipase A2, increase in memory T-cells]. It also triggers endothelial dysfunction, lipid dysregulation and deposition, proliferation, and migration of vascular SMC (e.g., through modulation of metalloproteinase 9), coagulation, and increased prothrombotic risk (e.g., increasing the production of thrombin and making endothelial cells more responsive to thrombin stimulation), upregulation of adhesion molecules [7885]. Interestingly, murine CMV infection increases aortic expression of proatherosclerotic genes [p38, extracellular signal-regulated kinase1/2 mitogen-activated protein kinase (ERK 1/2 MAPK), VCAM-1, ICAM-1, and MCP-1], whereas inhibition of p38 (SB203580) decreases pro-atherogenic molecules and CMV viral load in aortas of infected mice [85]. The virus has also been associated with endothelial dysfunction, AMI, coronary and peripheral atherosclerosis, and stroke [8688].

    Different evidence showed increased antibody positivity in patients at risk of atherosclerosis, as well as the number of plasma virus-DNA copies in patients with acute coronary syndrome compared to controls [8992]. Moreover, the presence of CMV-DNA in atherosclerotic plaques has been commonly found [93, 94]. However, negative data are also available, showing the lack of association between antibody titers and atherosclerosis, and highlighting the presence of the virus in non-atherosclerotic sites [65, 9597].

    HCV

    HCV remains one of the major causes of chronic liver disease, with 58 million HCV infections worldwide, about 1.5 million new HCV infections/year, and 290,000 deaths from HCV infection in 2019 [World Health Organization (WHO) data] [98]. HCV is also associated with several extrahepatic conditions, so the definition of liver-localized disease is superseded by the concept of systemic disease, which can involve a variety of extrahepatic complications [99]. In particular, emerging results highlight the relationship between HCV infection and CV risk factors, subclinical and clinical atherosclerosis (e.g., T2D, alterations of the lipid profile, carotid atherosclerosis, cerebrovascular events, stable coronary heart disease, and acute coronary events), as well as the biological plausibility of this association (especially when considering inflammatory and immune-related responses and metabolic alterations elicited by the virus) [100]. However, data are often contradictory and still widely discussed [101] as reported in the following paragraphs.

    Potential mechanisms linking HCV to atherosclerosis

    Endothelial cells are permissive to HCV infection in culture, an event that may lead to endothelial dysfunction and trigger the early atherogenesis process [102]. As a result, HCV clearance improved not only liver function, but also endothelial dysfunction and subclinical atherosclerosis (improvement of the ankle-brachial index, decrease in VCAM, e-selectin, endothelial and platelet apoptotic microparticles, and cell-free DNA) [103]. In fact, in addition to the identification of HCV in the carotid lesion, an indicator of the direct replication of the virus within the arterial wall, HCV+ was associated with intima-media thickness (IMT), strengthening the concept of a local pro-atherogenic effect of the virus at the atherosclerotic lesion level [104].

    Actually, the virus may contribute to atherosclerosis through different mechanisms, including the enhancement of inflammation and oxidative stress (e.g., by increasing TNF-α and IL-6, activation of toll-like receptors associated with pro-inflammatory cytokines, endoplasmic reticulum stress), both at liver and systemic levels [105110]. Chronic HCV infection may significantly trigger alterations in biomarkers of inflammation (e.g., CRP, IL-6, and TNF-α) and endothelial function (e.g., sICAM-1, sVCAM-1), but also cardiac dysfunction [e.g., on the N-terminal pro-B-type natriuretic peptide (NT-proBNP) and troponins]; HCV therapy may improve some of these alterations [111].

    Furthermore, HCV has been related to IR and T2D, mainly through mechanisms that include direct viral effects, elevation of proinflammatory cytokines, and immune-mediated responses [112]. In particular, HCV core protein expression induces hepatic IR by altering the signaling pathway of the insulin receptor substrate (IRS)-1 and -2 through direct and indirect effects (respectively, proteasomal degradation and increased levels of proinflammatory cytokines, such as TNF-α) [113]. Moreover, in HCV patients, IR is correlated with inflammatory markers like ferritin [114]. Increased levels of hepatic iron and copper, which are associated with chronic HCV, are hepatotoxic and contribute to increased oxidative stress, while zinc reduction (as zinc inhibits viral replication) may, in turn, increase the systemic inflammatory response [115]. In this context, different data demonstrate that Zn, thanks to its several properties (e.g., immune, antioxidant, anti-inflammatory, and antiviral capacities), may be beneficial for HCV patients [115119].

    HCV core protein induces changes in the cellular redox state [decreasing the oxidized and reduced forms of nicotinamide adenine dinucleotide (NAD+/NADH ratio)] and affects the sirtuin-1/AMP-activated protein kinase (SIRT1/AMPK) pathway, thus modifying the expression of glucose and lipid metabolism-related genes, and, as a consequence, inducing metabolic dysregulation at liver level [120]. In particular, this lipids disarrangement [e.g., increase in lipid biosynthesis, hepatic levels of triglycerides, cholesterol esters, and sphingolipids, but also reduction of mitochondrial oxidation, lipid degradation, and apolipoproteins export, in particular very-low-density lipoproteins (VLDL)] lead to circulatory hypo-cholesterolemia and hypo-lipoproteinemia, steatosis and lipotoxicity [121, 122]. Steatosis is in turns a well recognized CV risk factor, as it promotes IR, adiponectin elevation, metabolic syndrome, oxidative stress, hyperhomocysteinemia, and TNF-α [122125]. Clearly, alteration in the lipid asset may also be secondary to worsened liver function induced by HCV, in addition to direct HCV effects.

    Cryoglobulinemia (reversible precipitation of mixed-immunoglobulin at body temperatures lower than 37℃) has been strongly associated with HCV infection, and may exacerbate systemic inflammation and organ damage at different levels (e.g., kidney, liver, vasculitis) [126].

    Carotid atherosclerosis, CAD, cerebrovascular disease, myocarditis, and cardiomyopathies related to HCV

    In the early 2000s, HCV-seropositivity was identified to be correlated to an increased risk of carotid plaque and carotid intima-media thickening (4,784 subjects enrolled, 2.2% HCV+) providing the first evidence of the atherogenic potential of this virus [127]. The same authors also demonstrated that circulating HCV core protein is a strong, independent predictor of carotid plaques in a large general population (1,992 subjects) [128]: since then, several attempts have also been performed in other large populations, but with results that alternately support or debunk the link between HCV infection and CVD, indicating a higher, null, or even lower risk in HCV patients [129131]. In this context, a meta-analysis was performed to better define this relationship: the obtained results showed a higher risk of carotid plaques in HCV-infected patients compared to controls (OR, 2.27; 95% CI, 1.76–2.94; P < 0.001), without significant heterogeneity between studies [15]. Similar results were obtained when IMT, instead of carotid plaques, was considered as the outcome [15]. Interestingly, the significance of HCV infection on the presence of carotid plaques was influenced by smoking habits and was particularly strong in groups with a high prevalence of smokers [15]. A different meta-analysis study (5 studies) shows that the risk of developing carotid intimal media thickening or a carotid plaque in a subject with chronic hepatitis C is approximately four times higher than in an uninfected person [132]. Another recent meta-analysis (consisting of 341,739 people with HCV infection included in 36 studies from 51 countries) showed that HCV infection is associated with an increased risk of myocardial infarction, stroke, and vascular mortality, with values corresponding to 1.13 [95% confidence intervals (CI) 1.00–1.28], 1.38 (1.19–1.60), and 1.39 (1.24–1.55), respectively [133]. The authors also showed that the global burden of CVD associated with HCV infection is responsible for 1.5 million disability-adjusted life-year (DALYs; due to ischaemic heart disease and stroke), with the highest burden in low- and middle-income countries (regions in South Asia, Eastern Europe, North Africa, and the Middle East account for two-thirds of all HCV-associated CV DALYs) [133]. In fact, beyond carotids, many studies suggest that HCV infection can also increase CAD (as well as cerebrovascular disease and stroke). This effect was hypothesized when HCV+ was found to be associated with the presence of CAD, which remained significant even after correction for cardiometabolic risk factors, and which was associated with the severity of disease (number of diseased vessels) [134]. Many further investigations have confirmed this link, whereas others have found no significant relationship [135137]. As a result, the balance of these data ranges widely from no correlation to a favorable relationship. However, when analyzed in aggregate form in a meta-analysis (297,613 HCV patients, 557,814 uninfected controls), these results indicate an increased risk of CAD associated with HCV infection (20 studies, OR: 1.382; 95% CI: 1.103, 1.732) [138]. Similar results were confirmed in a meta-analysis of 10 studies, which showed a tripled risk of developing coronary atherosclerosis for HCV-positive patients when compared to non-infected subjects [139].

    Notably, HCV-related damage to the kidney may impact CV diseases and outcomes, as an increased risk of kidney disease has been observed in various groups of HCV+ patients [140, 141]. Several studies have also considered the potential association between HCV infection and cerebrovascular events, finding contradictory results, ranging from HCV infection as a risk factor to HCV infection as a protective factor [142144]. Despite the heterogeneity found among the studies included in a meta-analysis (made up of six studies, five retrospective and one prospective) published in 2013, results suggested that HCV infection increases the risk of stroke; with a random-effects model, the outcome was 1.58 (0.86, 2.30), which was improved after excluding the study that caused the heterogeneity, with a pooled odds ratio (OR) with a 95% CI equating to 1.97 (1.64, 2.30) [145]. A second meta-analysis, which included 8 studies, found that HCV-infected subjects kept an increased risk of cerebro-CV events (OR, 1.30; 95% CI, 1.10–1.55; P < 0.01) than uninfected subjects, with a stronger impact in those with higher CV risk (e.g., high incidence of T2D or hypertension) [15]. This trend was confirmed in a further meta-analysis (13 studies) where a higher risk of cerebrovascular disease was reported in HCV patients than in uninfected controls (OR: 1.485; 95% CI: 1.079, 2.044) [138]. Actually, a link between chronic HCV infection and altered cerebrovascular reactivity, measured by transcranial color Doppler for measurement of blood flow velocity, has been found, which may have adverse consequences on cerebrovascular hemodynamics and may also lead to increased risk of cerebrovascular diseases [146]. Interestingly, recent data have shown that HCV infection increases the risk of developing CAD or cerebrovascular disease in patients with hepatocellular carcinoma, pointing to the importance of atherosclerosis prevention in this specific group of patients [147].

    Myocarditis and subsequent cardiomyopathy can be triggered by several viruses, including HCV, which has been associated with both dilated cardiomyopathy (DCM; characterized by ventricular dilation and impaired contraction) and hypertrophic cardiomyopathy (HCM; characterized by increased ventricular wall mass not due to volume overload) [100]. Indeed, the virus genome has been detected within heart tissue biopsies from patients with myocarditis and DCM or HCM, suggesting a causal link [148, 149]. Among the possible mechanisms underlying this relationship, immune system activation, cardiac inflammation, and viraemia have been identified as the main HCV-related indirect factors associated with inflammatory myocarditis [150].

    CV mortality and HCV

    Interestingly, a higher mortality due to CV diseases was observed in more than 10,000 HCV-positive blood donors [151]. However, this relationship was not confirmed by other studies, such as in the analysis of a very large Australian population (30,000 subjects; although the particular enrolled population, which consisted of subjects with opioid dependence) [152]. Thus, these controversial results still make the association of HCV with mortality widely discussed and not sure yet. However, when the data are considered as aggregate in meta-analysis approaches, evidence of increased CV mortality due to HCV infection emerges. In fact, a meta-analysis including three cohort studies (68,365 patients, 735 deaths) showed an increased risks of CV-related mortality (OR: 1.65, 95% CI: 1.07–2.56; P = 0.02) in HCV infected patients, despite showing significant heterogeneity (I2 = 76%; P < 0.05) and the lack of consideration of important covariates (e.g., CV risk factors) [15]. Moreover, another meta-analysis including 574,081 patients on long-term dialysis confirmed an increased risk of CV mortality in HCV+ individuals compared with HCV controls [153].

    Interestingly, chronic HCV patients treated with direct-acting antivirals (DAA) experienced lower rates of CV events and all-cause mortality than those without treatment [154]. This finding is of particular interest, especially in view of the development of new DAA, which has profoundly simplified and improved HCV treatment for many more patients, and may lower CVD morbidity and mortality in chronic HCV patients.

    HCV relevance in specific subset of patients: the case of thalassemia major

    HCV infection may be more critical in subsets of patients at high risk of infection, such as thalassemic patients (due to frequent transfusions), where the virus may represent a strong accelerator of thalassemia major (TM) complications [155]. In fact, even though donor screening control reduces the likelihood of new infections, especially in Western nations where the risk of new cases is actually minimized, the elderly remain more vulnerable to the long-term consequences of previous chronic infection [156]. Nonetheless, in many other less-developed countries, patients with TM continue to contract HCV, often as children [156158]. Similar to other patient populations, chronic HCV infection is associated with a significantly higher risk of T2D and CV complications in TM patients, where it should be managed as a systemic disease in which extrahepatic complications exacerbate the weight of its pathological burden [159]. At the CV level, HCV infection can be involved in the pathogenesis of myocardial fibrosis through both myocarditis directly, and the pancreatic and liver damage with the development of diabetes indirectly in TM patients [160]. In this context, HCV infection represents one additional risk factor for low bone mass and reduced osteocalcin blood levels in TM patients [161].

    Interestingly, YKL-4 (inflammatory glycoprotein and a marker of endothelial dysfunction, name based on the one letter code for the first three N-terminal amino acids, tyrosine (Y), lysine (K), and leucine (L) as well as the molecular weight of YKL-40) is higher in patients with CVD or HCV, and it is also linked to liver stiffness and hepatic fibrosis degree in TM patients with liver disease [162]. Moreover, the fact that YKL-40 was also positively associated with the transfusion index, alanine aminotransferase, lactate dehydrogenase, ferritin, and liver iron concentrations (LIC), and negatively correlated with cardiac magnetic resonance imaging (T2*; biomarker to assess iron deposition and guide chelation therapy indicating properties of the tissue, and encoded in the pixels of the map) identified this protein as one potential key factor in the relationship between liver disease and CV complications [162].

    Coinfection

    In nature, viral coinfection is as common as viral infection alone, worsening things. In fact, coinfection may affect mutual viral pathogenicity, synergistically attack of host defense, and mix-up clinical symptoms, making diagnosis and treatment more difficult [163].

    The study of coinfection dynamics still represents an emerging field in virology, which may indeed improve diagnoses, the development of vaccines, and antiviral therapy in the future. HCV and HIV coinfection patients may be of particular interest because they show a higher risk of subclinical atherosclerosis (prevalence of carotid plaque) than those with HIV mono-infection, presenting an elevated blood proinflammatory milieu [164, 165]. Moreover, a meta-analysis including 33,723 participants evidenced a pooled adjusted HRs for the association between HIV/HCV coinfection and CVD (CAD, congestive HF, and stroke) of 1.24 (95% CI: 1.07–1.40) when compared with HIV mono-infection [166]. Co-infection of HIV and HCV also increases the risk of significant QT prolongation in HIV patients because the viruses may independently facilitate the development of torsade-de-pointes arrhythmia and death [167].

    Respiratory viruses

    Many viral infections show a seasonal trend, generally related to colder seasons (e.g., influenza), although some respiratory virus infections peak in the spring or summer (e.g., some parainfluenza virus), events whose underlying factors are not fully defined yet [168]. However, it is known that temperature and humidity are important factors affecting respiratory virus stability and transmission rates, as well as the modulation of host immune responses to viral respiratory infections [168]. In particular, it is known that influenza seasonality is significantly affected by temperature and humidity, with cool and dry conditions enhancing the virus’s survival and transmissibility in temperate climates [169]. These meteorological variables were found to be important factors also in the transmissibility and mortality of coronavirus disease 2019 (COVID-19), similar to what was observed for influenza [170174]. Nonetheless, other studies have shown that changes in meteorological variables (increase in temperature and humidity) may not necessarily lead to a decline in COVID-19 cases [175, 176]. Thus, to establish this association with greater certainty, future studies should evaluate other exposure measurements and important cofactors, such as immunological and socio-economic factors and inter-human contacts, population density, pollution, appropriate lag times, and non-linear associations [174, 177181].

    In view of their common dependency on environmental factors, in many temperate areas of the world a substantial overlap between human seasonal coronaviruses (e.g., alpha-coronaviruses NL63 and 229E and beta-coronaviruses OC43 and HKU1) and influenza virus has been observed, a fact that may increase misdiagnosis and represent a substantial additional burden on health systems [182]. As a result, SARS-CoV-2 and influenza coinfection are quite common (taking into account the underestimation due to the rate of those not detected, especially in those cases with milder symptoms that do not require healthcare services), in particular in patients with comorbidities (e.g., obesity, T2D, hypertension, cancer, and CAD), where exacerbation of symptoms (e.g., fever, cough, dyspnea, diarrhea, and fatigue), and complications (e.g., acute ischemic cardiac disease, kidney injury, and acute HF) can occur [183]. Thus, considering that SARS-CoV-2 and influenza viruses infect a common subgroup of high-risk patients, more knowledge is needed to assess how coinfection may affect a patient’s clinical response and prognosis.

    Atherosclerosis and influenza (between the old acquaintances)

    Influenza is a very common infection, responsible for an estimated 1 billion cases, and 3 to 5 million severe cases, with up to 650,000 leading to influenza-related respiratory deaths each year worldwide [184, 185].

    The close epidemiological link between atherosclerosis, CVD, and influenza infection has been recognized since the beginning of the 20th century, coinciding with the great flu epidemic of 1918–1920 [186].

    Pathogenic mechanisms

    It has been shown that the virus can infect and localize directly in the atherosclerotic arteries and that infection is associated with the pro-inflammatory response at both systemic and arterial levels [187]. As a result, endothelial cells are permissive to virus infection, which generates a strong increase of cytokines and adhesion molecules locally [188]. Moreover, the virus can induce apoptosis of epithelial cells and increase endothelial permeability through the hyperactivation of cytokines and chemokines, inducing endothelial dysfunction [189]. Another possible mechanism linking the virus to atherosclerosis is the effect observed on the high-density lipoprotein (HDL) antiinflammatory capacity by increasing arterial macrophage traffic in mice, an event that appeared preventable by the administration of D-4F, an apolipoprotein A-I mimetic peptide [190].

    Furthermore, influenza virus infection and AMI resulted in being closely cross-linked, as influenza virus infection can destabilize atherosclerotic plaques and induce platelet activation and inflammatory responses, hence causing a thrombogenic milieu [191195]. One of the main mechanisms the virus can use to exert its effects is the induction of inflammatory cytokines, which may take part in the development of atherosclerosis and trigger the occurrence of AMI [196]. Moreover, influenza A virus infection increases p38 MAPK-mediated matrix metalloproteinase-13 (MMP-13) expression, which may lead to destabilization of vulnerable atherosclerotic plaques in the artery [197]. Type 2 AMI may also indirectly occur due to the increased metabolic demands of myocardial tissue for tachycardia, fever, and hypoxia [198].

    Clinical evidences

    Available data on the relationship between influenza and CAD reported controversial results, ranging from a positive to null relationship [198201]. Nonetheless, a temporal relationship between influenza infection and AMI has also been found using patients self-reported data to classify respiratory symptoms, finding a significant risk due to respiratory tract infections in the week prior to the onset of AMI, and suggesting an inflammatory response associated with the precipitation of the acute CV event [202, 203]. A more recent study that used laboratory-confirmed influenza infection data corroborated these findings by showing a six-fold incidence ratio for AMI hospitalization during the risk interval (one week after respiratory specimen collection); the incidence of AMI was also high after infection with non-influenza respiratory viruses (although to a lesser extent than influenza, corresponding to 3.51 for respiratory syncytial virus; 95% CI: 1.11–11.12, and 2.77 for other viruses; 95% CI: 1.23–6.24) [204]. AMI patients with concomitant influenza infection also exhibited higher rates of in-hospital mortality, 30-day readmission, and in-hospital complications, as well as higher health-system resource utilization compared to those without influenza [205].

    Influenza vaccine

    An interesting experimental study evaluated the effect of vaccination (45 μg/0.5 mL Vaxigrip®, the same dose used to immunize human adults against influenza) on atherosclerotic plaque development in hyperlipidemic mice [apoE–/– mice] [206]. Results showed that vaccinated animals developed smaller lesions with lower lipid but higher SMC content and collagen deposition than control animals, in parallel with decreased levels of pro-inflammatory cytokines (IFN-γ, TNF-α, IL10) and increased levels of the anti-inflammatory cytokine IL-4 [206]. Thus, influenza vaccination seems to exert a beneficial effect by promoting smaller and stable atherosclerotic plaques and eliciting atheroprotective immune responses.

    Moreover, as effective vaccination prophylaxis is possible for influenza, several clinical studies have focused on the effects of influenza vaccination on CV events, overall supporting a correlation between influenza vaccination and a reduction in CV events, although some studies remain controversial [207209]. Notably, the Influenza Vaccination in Prevention From Acute Coronary Events in Coronary Artery Disease study (FLUCAD) study, carried out in CAD patients, reported a lack of significance for the composite outcome estimated at 12-month event rate (including CV death, myocardial infarction, coronary revascularization), and slight significance for the composite outcome (including the previous endpoints and rehospitalization for myocardial ischaemia) [209]. A meta-analysis of case-control studies (8 on influenza vaccination, 10 on influenza infection and AMI) found a significant association between recent respiratory infection and AMI, with a pooled OR 2.01 (95% CI 1.47 to 2.76) [210]. Moreover, influenza vaccination was found to be significantly associated with protection from AMI, with an estimated vaccine effectiveness of 29% (95% CI 9% to 44%) [210]. Recent results from the Influenza Vaccination After Myocardial Infarction (IAMI) study also showed that early influenza vaccination after an AMI (within 72 h of hospitalization) result in a significant 28% reduction in major adverse CV events (MACE; as AMI, or stent thrombosis) and a 41% reduction in CV mortality, with no excess in serious adverse complications [211]. The updated meta-analysis of 8 randomized clinical trials, including IAMI and the recent influenza vaccine to prevent adverse vascular events trials (4,420 patients), showed that influenza vaccine, compared with control/placebo, was associated with a significantly lower risk of MACE at follow-up [risk ratio (RR) 0.75, 95% CI 0.57–0.97] [212]. Moreover, a very recent meta-analysis (5 randomized trials, 4,187 patients, 2 studies included patients with acute coronary syndrome, 3 patients with stable CAD and acute coronary syndrome) reported that influenza vaccine may represent a cheap and reliable tool to reduce the risk for all-cause mortality (by by 44%), CV mortality (by 46%), major acute CV events (by 34%), and acute coronary syndrome among CAD patients (by 36%) [213]. Another very recent meta-analysis (22,634,643 hospitalizations) assessed the role of influenza vaccination in protecting the CV system, revealing that those patients vaccinated against influenza were associated with a lower incidence of AMI (RR = 0.84, 95% CI: 0.82–0.87, P < 0.001), transient ischemic attack (TIA; RR = 0.93, 95% CI: 0.9–0.96, P < 0.001), cardiac arrest (RR = 0.36, 95% CI: 0.33–0.39, P < 0.001), stroke (RR = 0.94, 95% CI: 0.91, 0.97, P < 0.001), and mortality (RR = 0.38, 95% CI: 0.36–0.4, P < 0.001) [214].

    Taken altogether, these results support the hypothesis that the influenza vaccine may be a cheap and effective intervention to reduce the risk for all-cause mortality, CV mortality, major acute CV events, and acute coronary syndrome. This information may be of critical public health significance, because of the number of unvaccinated subjects, the high number of CV complications associated with respiratory tract infection, and the beneficial effect of vaccination in terms of risk reduction on patients with high CV risk. Accordingly, the 2019 European Society of Cardiology-ESC guidelines for the diagnosis and management of chronic coronary syndromes recommended influenza vaccination (class I, level of evidence B) [215]. However, these recommendations have not yet been fully implemented in the cardiology clinical practice. As a matter of fact, a US survey (2016–2019) reported that only 50% of patients with CVD had received influenza vaccination, pointing to socio-economic disparities as a critical determinant associated with the inadequate delivery of this important preventive tool [216].

    Atherosclerosis and SARS-CoV-2: a new enemy?

    Since its first appearance at the end of 2019, SARS-CoV-2 virus, responsible for COVID-19, has infected more than 766,440,796 people and caused about 6,932,591 deaths until now [184]. Its link to CVD has emerged since the beginning of the pandemic, with an increased incidence of myocarditis, pericarditis, arrhythmias, HF, and thromboembolism, not only in the acute phase but also beyond the first 30 days of the infection [217, 218]. Moreover, preexisting CVD risk factors (such as obesity, hypertension, and T2D) increase susceptibility to COVID-19 [219]. In particular, hypertension was found to be closely related to disease severity and higher mortality [220]. To date, COVID-19 represents one of the most critical health and economic issues, as this infection is posing new diagnostic, clinical, and therapeutic challenges as well as long-lasting direct and indirect effects whose recognition and containment are critical to addressing the negative collateral consequences, which are likely to be seen in currently infected patients in the near future but also for decades to come.

    Pathogenic mechanisms

    The ability of SARS-CoV-2 to bind to ACE2 receptors for entry into cells allows direct access to the circulatory and CV systems, disseminating the virus towards major organs and leading to major health complications; SARS-CoV-2 may directly enter endothelial cells (triggering the inflammatory response and causing apoptosis and disruption of intercellular junctions) and cardiac cells (causing myocarditis through direct cardiac damage) [221].

    Although every viral infection triggers an increase in certain inflammatory biomarkers, SARS-CoV-2 is mainly associated with the activation of an even greater number of different cytokines (e.g., TNF-α, IL-1β, IL-2, IL-6, CRP, and ferritin) at very high levels, which likely contribute to the more severe symptoms and health complications observed in many COVID-19 patients compared with other respiratory infections [222, 223]. In addition, SARS-CoV-2 also works as a complement activator (C3, C5), exacerbating symptoms and inducing acute respiratory distress syndrome [224]. The hypercoagulable state associated with COVID-19 is reflected in the increased concentration of coagulation and inflammatory biomarkers (e.g., CRP, D-dimer, fibrinogen, and fibrin degradation products) [225]. Moreover, other mechanisms, including hypoxia-induced injury and endothelial dysfunction, may play a pathogenetic role in the SARS-CoV-2-related CV manifestations [226, 227].

    T2D and COVID have already shown a bidirectional relationship; in fact, if T2D is a risk factor for the development of severe and critical forms of COVID-19, the virus can also induce new-onset T2D in nondiabetic patients. Mechanisms likely involve ACE2 (entry factor) and ketoacidosis precipitation in infected patients (characterized by decreased insulin blood levels, decreased glucose utilization, and uncontrolled lipolysis, but also an excessive increase in ketone bodies and acidosis) [228, 229]. Moreover, the virus may increase IR through enhanced expression of the RE1-silencing transcription factor (REST), which in turn modulates the expression of metabolic factors (e.g., myeloperoxidase, apelin, and myostatin), thus altering glucose and lipid metabolism [229]. The reduction of adiponectin, a hormone involved in glycemic and lipid homeostasis, may be an additional mechanism linking SARS-CoV-2 to IR [230].

    An interesting recent development in this field concerns sphingolipids, bioactive molecules that play important roles in many crucial cellular pathways, including inflammatory and oxidative stress, as well as in all processes related to membrane dynamics [231, 232]. The properties of these molecule make them potential regulators of the life cycle of several viruses (e.g., HCV, HIV, influenza, and SARS-CoV-2) and, indeed, evidence of the interaction has been found at different steps of viral replication cycle (entry into the plasma membrane or endosomal membranes, relationship leading to sphingolipid-mediated signal transduction, relationship with internal membranes and lipids during replication, virus assembly and budding) [233]. Remarkably, recent data evaluated the antiviral activity of two specific inhibitors of uracil-diphosphate glucose (UDP)-glucose:ceramide glucosyl-transferase [glucosylceramide synthase (GCS)], which catalyzes the biosynthesis of glucosylceramide (GlcCer; backbone of more than 300 structurally different glycosphingolipids), blocking the conversion of ceramide to GlcCer [234]. Both inhibitors hamper the replication of SARS-CoV-2 and influenza virus, suggesting that synthesis of glycosphingolipids is needed to sustain viral life cycles and that GCS inhibitors might be used as antiviral tools, likely effective also in case of coinfections [234].

    Thus, as some sphingolipids inhibitors are already in use or being studied in different clinical settings, targeted modulation of sphingolipid metabolism may really open new possibilities for additive pharmacological strategies to reinforce current available antiviral therapies [235].

    Clinical evidences

    High levels of cardiac biomarkers (e.g., NT-proBNP, troponin), common in COVID-19 patients, are indicative of myocardial injury and can be used to predict short- and long-term outcome of COVID-19 infection [236]. In this regard, a meta-analysis (3,044 confirmed COVID-19 cases from 12 studies) reported that the most common CV complications in COVID-19 patients were myocardial injury (21.2%, 95% CI 12.3–30.0%), arrhythmia (15.3%, 95% CI 8.4–22.3%), HF (14.4%, 95% CI 5.7–23.1%), and acute coronary syndrome (1.0%, 95% CI 0.5–1.5%) [237]. Moreover, the pooled incidence of HF, arrhythmia, and myocardial injury in non-survivors was 47.8% (95% CI 41.4–54.2%), 40.3% (95% CI 1.6–78.9%) and 61.7% (95% CI 46.8–76.6%), respectively [237]. In another meta-analysis (20,875,843 patients, follow-up of 8.5 months), COVID-19 patients resulted at high risk of AMI (HR: 1.93, 95% CI: 1.65–2.26, P < 0.0001, I2 = 83.5%) [238]. Acute event precipitation in COVID patients is likely triggered by high levels of inflammation and hypercoagulability, as suggested by proteomic analysis [239]. Moreover, several studies reported an increased risk of mortality and complications in COVID-19 patients with preexisting CVD [240, 241].

    Myocarditis may also occur during COVID-19, with variable severity and generally characterized by high lymphocytic inflammatory infiltrates [242]. It has been estimated that the incidence of myocarditis pre-COVID ranged from 1 to 10 cases/100,000 subjects, increasing after the SARS-CoV-2 pandemic from 150 to 4,000 cases/100,000 individuals [243]. Five years after the onset of the pandemic, long-term outcome data indicate a significant burden of CVD after recovery from acute COVID-19 illness [243]. In particular, late complications following acute SARS-CoV-2 infection may occur, including disturbances in vascular hemostasis and blood coagulation, CAD, myocardial fibrosis, AMI, and cardiac hypertrophy [244246]. Interestingly, also asymptomatic SARS-CoV-2 infection may present CV complications, as shown in a study that enrolled 139 healthcare workers with confirmed prior SARS-CoV-2 infection [serological or reverse transcriptase-polymerase chain reaction (RT-PCR)], in which signs of myocarditis were observed on cardiac magnetic resonance in 37% of the participants (median of 10 weeks after infection) [247]. Among the participants, only half had symptoms of COVID-19, suggesting that even asymptomatic patients may present important CV complications quite sometime after SARS-CoV-2 infection. Notably, following SARS-CoV-2 infection and inflammatory burst, autoimmune reactions may complicate post-COVID recovery, contributing to the post-acute complications of COVID-19 (long COVID) [248].

    Some CV drugs [including ACE inhibitors (ACEIs) and angiotensin receptor blockers (ARBs), statins, anticoagulants, and aspirin] seem to have a role in preventing COVID-19 complications, although current recommendations from leading Cardiovascular Scientific Societies are to maintain patient’s current regimens, unless clinically indicated (e.g., hemodynamic instability) [249]. In contrast, pharmacological agents used for COVID-19 (e.g., remdesivir, ribavirin, lopinavir, chloroquine, methylprednisolone, and tocilizumab) may have significant CV side effects and, by interacting with CV agents, cause potential adverse effects on the CV system [250].

    Although vaccination against SARS-CoV-2 is generally safe and effective, a certain rate of complications is observed. In particular, compared to the influenza vaccine, COVID-19 mRNA vaccines showed a significantly higher risk for hypertensive crisis (adjusted OR 12.72; 95% CI 2.47–65.54), and supraventricular tachycardia (7.94; 2.62–24.00) [251]. The overall risk of myopericarditis after receiving the COVID-19 vaccine is low; however, cases of myocarditis and pericarditis may occur, with an overall incidence of about 10/100,000, a range that may increase when considering the subgroup of young males (about 50/100,000; most cases occurring some days after the second dose) [244, 252255]. Autopsies performed on 25 people who died unexpectedly and within 20 days after anti-SARS-CoV-2 vaccination revealed acute (epi-)myocarditis in four patients without detection of any other significant disease that may have caused an unexpected death [256]. Histological analysis showed patchy interstitial myocardial T-lymphocytic infiltration (predominantly of the CD4-positive subset), associated with mild myocyte damage. Taken together, autopsy findings suggested death due to acute arrhythmogenic cardiac failure, and indicated myocarditis as a life-threatening complication following mRNA-based anti-SARS-CoV-2 vaccination. One origin of this damage is that, despite differences in their composition (amount of lipid nanoparticles and excipients), all vaccines elicit a strong release of pro-inflammatory cytokines (e.g., IL-18), and NFκB activation, which may increase immune-mediated responses, cardiotoxicity, and myocarditis risk [257]. Moreover, spike protein impairs mitochondrial function in human cardiomyocytes and alters endothelial function, immune response, and renin-angiotensin-system balance, with negative consequences for the pathophysiology of the CV system [258, 259]. A “hyper-catecholaminergic” status has also been identified as one of the key triggers of SARS-CoV-2 mRNA vaccine-induced myocarditis and related outcomes (whether triggered by SARS-CoV-2 mRNA, SARS-CoV-2 spike protein, or both remains unclear) [260]. Noteworthy, the psycho-emotional repercussions and decreased well-being perception in Italian adolescents in response to the COVID-19 pandemic are significant, with gender-related differences (concerning psychological and physical well-being, mood/emotion, and self-perception), whereas combined stress (psychological and physical) could exacerbate CV responses in young males, contributing to adverse consequences after vaccination in this particular population segment [261, 262].

    The primary concern on the difficulties associated with managing the negative consequences of the COVID-19 pandemic, which has strained the health care system globally, has decreased attention towards psychological effects in the general population and people with chronic diseases. In particular, a significant drop in AMI hospitalizations was observed worldwide during the COVID-19 outbreak. One possible explanation for this outcome is that the pandemic may have generated fear and adverse psychological consequences in AMI patients, delaying hospital access [263]. Hence, there was a significant delay in the elapsed time “from symptom onset to first medical contact” when considering “total ischemic time” (definition denoting the elapsed time from the onset of chest pain to the first medical contact, arrival at the hospital, and balloon inflation during primary percutaneous coronary intervention) [263, 264]. This behavior denotes patients’ hesitation to contact healthcare personnel and go to the hospital or even not seek care at all, which is also confirmed by the higher levels of emotional and symptomatic fear expressions [fear of COVID-19 scale (FCV-19S) questionnaire] found in AMI patients during the pandemic compared to the general population [265, 266]. Noteworthy, “door-to-hospital-arrival-time” and “hospital-arrival-to-insufflation-time” did not differ significantly during the pandemic with respect to the pre-COVID period, suggesting an effective organization of the healthcare system and leaving a major role to the patient’s fear and reluctance [263, 264].

    Moreover, health workers, potentially exposed to the pathogen and subjected to high workload and job stress, generally did not receive mental health assistance during the pandemic (e.g., mindfulness-based stress reduction courses to reduce distress during emergency periods), and this may indirectly affect care quality [267]. Thus, it is crucial to focus on the psycho-emotional aspects of the current sanitary crisis and to understand how people relate to the pandemic, providing assistance also for these aspects and giving correct information about the pandemic course and the risks of delayed access to the hospital in case of acute events, supporting at the same time caregivers as well as patients and the general population with appropriate and targeted coping strategies when needed.

    Discussion

    It is still very difficult to establish whether infection and CV risk and disease are actually related through direct or indirect underlying pathogenic mechanisms, or whether they simply coexist in an association, as both diseases are common occurrences that can be observed in a significant portion of the general population. Moreover, the available results are often controversial and limited by different design and patient cohorts, the definition of CV outcomes, confounding factors, and small sample size, thus lacking adequate statistical power to demonstrate significant differences.

    Despite extensive efforts, there is still weak proof of infections of some viruses in the atherosclerotic lesions [65, 268]. Even the presence of a virus in an atherosclerotic artery doesn’t constitute a conclusive finding since it might be a bystander rather than an active inducer of the illness, which has to be verified by additional evidence at the cellular and molecular levels. Moreover, a diseased vessel may simply be more vulnerable to an infectious agent. Noteworthy, an extensive reviewed literature places greater correlations in chronic infections, in agreement with several physiopathogenetic mechanisms. In particular, it appears that chronic infections may induce structural and proinflammatory modification related to the development of the atherosclerotic lesion, as in the case of latent and persistent infection from CMV [269]. However, things in this area are much more complex; in fact, influenza A infection appears to accelerate the early stage of atherosclerosis [270, 271]. Thus, a specific virus would play a more important role in specific steps of the atherosclerotic process (e.g., at the beginning of the injury vs. following phases during the development of the plaque) acting more in specific subgroups of patients (e.g., patients with subclinical atherosclerosis vs. those with an established disease). Moreover, the presence of infected individuals without atherosclerosis, together with others who develop extensive damage in the context of a multifactorial disease such as CVD, implies that a single factor cannot be considered a necessary and sufficient causal factor, and that the contribution of other factors (e.g., host genomics and environmental factors) is also required. Anyway, for example considering influenza, the effect of meteorological variables is still under evaluation, due to the complex mechanisms underlying disease dynamics (e.g., peculiarities of the population, demographic dispersion, spatial diffusion, and climatic characteristics) [272, 273].

    Viruses are not studied in the same way since they act through some common mechanisms but also present peculiarities; in the future, it will be important to define the strength of each agent in terms of the onset and development of atherosclerosis, also considering the role of coinfection and the interaction of other factors (e.g., diseases such as steatosis and T2D; biomarkers like CRP, TNF-α, and ILs; host susceptibility, and host genetic asset). In addition, as more evidence has reported the presence of more than one pathogen in atherosclerotic plaques [274, 275], the study of coinfection, which is now an emerging discipline in virology, will be important to define subgroups of patients at higher risk (as simultaneous infection with multiple agents seems to increase the risk of CVD and its complications), where combined anti-infectious and anti-inflammatory strategies might be useful and more efficient [166, 276, 277]. Nonetheless, antiviral pharmacological agents may also alternatively benefit or exacerbate CVD risk, as in the case of antiretroviral therapy, which may reduce inflammation and other markers of CV risk but induce lipid abnormalities (e.g., hypertriglyceridemia) [50].

    For it concerns vaccinations, data from different meta-analyses testify for a protective role of influenza vaccination on the CV system [213, 214], although the fact remains that influenza vaccination as a preventive measure for CV disease remains low and is not yet considered an additional and reliable CV preventive strategy in clinical settings, although recommended by many guidelines for different subgroups of patients at risk [208, 215, 278, 279]. In particular, the 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes recommended annual influenza vaccination in patients with CAD, especially in the elderly [215]. Similar advice from the AHA/ACC for patients with coronary and other atherosclerotic vascular disease (class I, level B) [278]. Awareness of the general population, patients, and cardiologists surely needs to be improved with targeted campaigns, and alternatives to vaccination must be applied in patients with contraindication to vaccine (e.g., severe allergic reaction to any vaccine component or to a previous dose of vaccine, immune compromisation) recommending protective behaviors, always considering that key barriers to influenza vaccination (e.g., socioeconomic status, racial disparities) are not completely eliminated, and not all the causes have been completely identified, thus needing further careful evaluation in future [278, 280].

    In contrast, COVID-19 mRNA vaccines may induce a range of CV complications (e.g., thrombosis, stroke, myocarditis, myocardial infarction, pulmonary embolism, and arrhythmias) [281]. Moreover, the long-term outcomes following CV complications (e.g., myocarditis) related to SARS-CoV-2 vaccination are yet to be clarified. Therefore, it may be crucial to monitor more, and with appropriate diagnostic and therapeutic tools, subgroups of vaccinated patients at risk to minimize serious adverse events.

    The COVID-19 pandemic has led to new indirect effects on atherosclerosis for the general population, patients, and healthcare workers, including delayed presentation of acute illness (e.g., AMI) and the burden of social distancing and quarantine on socialization, decreased psycho-emotional well-being, and reduced physical activity. Hence, it will be particularly important to assess the role of social and psychological issues and adopt measures to assist vulnerable and CV high-risk patients, as well as distressed healthcare professionals.

    Conclusions

    Despite the amount of evidence and strong biological plausibility (particularly in terms of inducing inflammatory and immunological responses), the causal relationship between viral infection and atherosclerosis has not been confirmed without any doubt, due to the complexity in the relation between viruses and atherosclerosis, and the persistence of many aspects still unsolved. At present, the importance of viral infection as risk factors has not been fully recognized in clinical practice. Nonetheless, monitoring, targeting, and treating conditions that predispose to CVD in infected subjects as well as considering infection as a risk factor for cardiometabolic disease are concerns gaining more and more attention among cardiologists. So, further research in this field, increased information for patients and physicians together with the development of improved tools (e.g., vaccine strategies and therapeutic agents reliable to prevent and/or treat the acute and long-term effects of viruses in terms of atherosclerosis) may have important implications for CV patient health, especially for those belonging to more vulnerable subgroups.

    Key points requiring further deepening in the future:

    • Although the amount of evidence and biological plausibility strongly suggest a pathogenic relationship between virus infection and CVD, a definitive proof of evidence has not yet been clearly reached.

    • Not all viruses have been studied equally, and even for the most studied heterogeneity between studies exists.

    • Coinfection may exacerbate the pro-atherogenic effects.

    • The relationship between viruses and atherosclerosis is likely to be more important in subgroups of patients with specific risk profiles.

    • Viruses may have peculiar mechanisms of action, in addition to common effects (such as the enhancement of inflammation and oxidative stress), which may affect their acute and long-term effects on the atherosclerotic process.

    • Anti-/pro-atherogenic effects of antiviral drugs may be taken into consideration.

    • SARS-CoV-2 has evidenced new additive mechanisms related to fear and social reactions, which may involve the social and psycho-emotional status of the individual and affect management and prognosis in the CV setting.

    Abbreviations

    ACE:

    angiotensin-converting enzyme

    AMI:

    acute myocardial infarction

    apoE:

    apolipoprotein E

    CAD:

    coronary artery disease

    COVID-19:

    coronavirus disease 2019

    CMV:

    cytomegalovirus

    CRP:

    C-reactive protein

    CV:

    cardiovascular

    CVD:

    cardiovascular disease

    CI:

    confidence intervals

    HCV:

    hepatitis C virus

    HIV:

    human immunodeficiency virus

    HSV:

    herpes simplex virus

    HF:

    heart failure

    ICAM-1:

    intercellular adhesion molecule 1

    IL:

    interleukin

    IMT:

    intima-media thickness

    IR:

    insulin resistance

    LOX-1:

    lectin-like oxidized low-density lipoprotein receptor-1

    NO:

    nitric oxide

    OR:

    odds ratio

    RR:

    risk ratio

    SARS-CoV-2:

    severe acute respiratory syndrome coronavirus-2

    SMC:

    smooth muscle cells

    T2D:

    type 2 diabetes

    TM:

    thalassemia major

    TNF:

    tumor necrosis factor

    VCAM:

    vascular cell adhesion molecule

    Declarations

    Acknowledgments

    The author deserves special thanks to Dr. Simona Fenizia for her valuable contribution to English-style editing.

    Author contributions

    CV: Conceptualization, writing—original draft, writing—review & editing.

    Conflicts of interest

    The author declares that he has no conflicts of interest.

    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) 2023.

    References

    Ross R. Atherosclerosis—an inflammatory disease. N Engl J Med. 1999;340:11526. [DOI] [PubMed]
    Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S. Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease. Circ Res. 2016;118:53546. [DOI] [PubMed]
    Vernon ST, Coffey S, Bhindi R, Soo Hoo SY, Nelson GI, Ward MR, et al. Increasing proportion of ST elevation myocardial infarction patients with coronary atherosclerosis poorly explained by standard modifiable risk factors. Eur J Prev Cardiol. 2017;24:182430. [DOI] [PubMed]
    Khot UN, Khot MB, Bajzer CT, Sapp SK, Ohman EM, Brener SJ, et al. Prevalence of conventional risk factors in patients with coronary heart disease. JAMA. 2003;290:898904. [DOI] [PubMed]
    Traghella I, Mastorci F, Pepe A, Pingitore A, Vassalle C. Nontraditional cardiovascular biomarkers and risk factors: rationale and future perspectives. Biomolecules. 2018;8:40. [DOI] [PubMed] [PMC]
    Gaggini M, Gorini F, Vassalle C. Lipids in atherosclerosis: pathophysiology and the role of calculated lipid indices in assessing cardiovascular risk in patients with hyperlipidemia. Int J Mol Sci. 2023;24:75. [DOI] [PubMed] [PMC]
    Osler W. Diseases of the arteries. In: Modern medicine: its practice and theory. Washington: Lea & Febiger; 1908. pp. 26–47.
    Frothingham C. The relation between acute infectious diseases and arterial lesions. Arch Intern Med. 1911;8:15362. [DOI]
    Fabricant CG, Fabricant J, Litrenta MM, Minick CR. Virus-induced atherosclerosis. J Exp Med. 1978;148:33540. [DOI] [PubMed] [PMC]
    Rosenfeld ME, Campbell LA. Pathogens and atherosclerosis: update on the potential contribution of multiple infectious organisms to the pathogenesis of atherosclerosis. Thromb Haemost. 2011;106:85867. [DOI] [PubMed]
    Filardo S, Di Pietro M, Farcomeni A, Schiavoni G, Sessa R. Chlamydia pneumoniae-mediated inflammation in atherosclerosis: a meta-analysis. Mediators Inflamm. 2015;2015:378658. [DOI] [PubMed] [PMC]
    Liccardo D, Cannavo A, Spagnuolo G, Ferrara N, Cittadini A, Rengo C, et al. Periodontal disease: a risk factor for diabetes and cardiovascular disease. Int J Mol Sci. 2019;20:1414. [DOI] [PubMed] [PMC]
    Barbaro G. Cardiovascular manifestations of HIV infection. Circulation. 2002;106:14205. [DOI] [PubMed] [PMC]
    Lebedeva AM, Shpektor AV, Vasilieva EY, Margolis LB. Cytomegalovirus infection in cardiovascular diseases. Biochemistry (Mosc). 2018;83:143747. [DOI] [PubMed]
    Petta S, Maida M, Macaluso FS, Barbara M, Licata A, Craxì A, et al. Hepatitis C virus infection is associated with increased cardiovascular mortality: a meta-analysis of observational studies. Gastroenterology. 2016;150:14555. [DOI] [PubMed]
    Wang B, Yu M, Zhang R, Chen S, Xi Y, Duan G. A meta-analysis of the association between Helicobacter pylori infection and risk of atherosclerotic cardiovascular disease. Helicobacter. 2020;25:12761. [DOI] [PubMed]
    Wu YP, Sun DD, Wang Y, Liu W, Yang J. Herpes simplex virus type 1 and type 2 infection increases atherosclerosis risk: evidence based on a meta-analysis. Biomed Res Int. 2016;2016:2630865. [DOI] [PubMed] [PMC]
    Zhu J, Quyyumi AA, Norman JE, Costello R, Csako G, Epstein SE. The possible role of hepatitis A virus in the pathogenesis of atherosclerosis. J Infect Dis. 2000;182:15837. [DOI] [PubMed]
    Ishizaka N, Ishizaka Y, Takahashi E, Toda E, Hashimoto H, Ohno M, et al. Increased prevalence of carotid atherosclerosis in hepatitis B virus carriers. Circulation. 2002;105:102830. [DOI] [PubMed]
    Choroszy M, Litwinowicz K, Bednarz R, Roleder T, Lerman A, Toya T, et al. Human gut microbiota in coronary artery disease: a systematic review and meta-analysis. Metabolites. 2022;12:1165. [DOI] [PubMed] [PMC]
    Campbell LA, Rosenfeld ME. Infection and atherosclerosis development. Arch Med Res. 2015;46:33950. [DOI] [PubMed] [PMC]
    Markle HV. Coronary artery disease associated with Helicobacter pylori infection is at least partially due to inadequate folate status. Med Hypotheses. 1997;49:28992. [DOI] [PubMed]
    Kramer CD, Weinberg EO, Gower AC, He X, Mekasha S, Slocum C, et al. Distinct gene signatures in aortic tissue from ApoE-/- mice exposed to pathogens or Western diet. BMC Genomics. 2014;15:1176. [DOI] [PubMed] [PMC]
    HIV and AIDS [Internet]. Geneva: World Health Organization; c2023 [cited 2023 Dec 21]. Available from: https://www.who.int/news-room/fact-sheets/detail/hiv-aids
    Hsue PY, Waters DD. HIV infection and coronary heart disease: mechanisms and management. Nat Rev Cardiol. 2019;16:74559. [DOI] [PubMed] [PMC]
    Kearns A, Gordon J, Burdo TH, Qin X. HIV-1-associated atherosclerosis: unraveling the missing link. J Am Coll Cardiol. 2017;69:308498. [DOI] [PubMed] [PMC]
    Duffy P, Wang X, Lin PH, Yao Q, Chen C. HIV Nef protein causes endothelial dysfunction in porcine pulmonary arteries and human pulmonary artery endothelial cells. J Surg Res. 2009;156:25764. [DOI] [PubMed] [PMC]
    Lafon ME, Gendrault JL, Royer C, Jaeck D, Kirn A, Steffan AM. Human endothelial cells isolated from the hepatic sinusoids and the umbilical vein display a different permissiveness for HIV1. Res Virol. 1993;144:99104. [DOI] [PubMed]
    Di Yacovo S, Saumoy M, Sánchez-Quesada JL, Navarro A, Sviridov D, Javaloyas M, et al. Lipids, biomarkers, and subclinical atherosclerosis in treatment-naive HIV patients starting or not starting antiretroviral therapy: comparison with a healthy control group in a 2-year prospective study. PLoS One. 2020;15:e0237739. [DOI] [PubMed] [PMC]
    Islam RK, Donnelly E, Islam KN. Circulating hydrogen sulfide (H2S) and nitric oxide (NO) levels are significantly reduced in HIV patients concomitant with increased oxidative stress biomarkers. J Clin Med. 2021;10:4460. [DOI] [PubMed] [PMC]
    Kannan M, Sil S, Oladapo A, Thangaraj A, Periyasamy P, Buch S. HIV-1 Tat-mediated microglial ferroptosis involves the miR-204-ACSL4 signaling axis. Redox Biol. 2023;62:102689. [DOI] [PubMed] [PMC]
    Toborek M, Lee YW, Pu H, Malecki A, Flora G, Garrido R, et al. HIV-Tat protein induces oxidative and inflammatory pathways in brain endothelium. J Neurochem. 2003;84:16979. [DOI] [PubMed]
    Song HY, Ju SM, Seo WY, Goh AR, Lee JK, Bae YS, et al. Nox2-based NADPH oxidase mediates HIV-1 Tat-induced up-regulation of VCAM-1/ICAM-1 and subsequent monocyte adhesion in human astrocytes. Free Radic Biol Med. 2011;50:57684. [DOI] [PubMed]
    El-Amine R, Germini D, Zakharova VV, Tsfasman T, Sheval EV, Louzada RAN, et al. HIV-1 Tat protein induces DNA damage in human peripheral blood B-lymphocytes via mitochondrial ROS production. Redox Biol. 2018;15:97108. [DOI] [PubMed] [PMC]
    Ramirez PW, Vollbrecht T, Acosta FM, Suarez M, Angerstein AO, Wallace J, et al. Nef enhances HIV-1 replication and infectivity independently of SERINC5 in CEM T cells. Virology. 2023;578:15462. [DOI] [PubMed] [PMC]
    Duette G, Cronin S, Kelleher AD, Palmer S. Viral competition assay to assess the role of HIV-1 proteins in immune evasion. STAR Protoc. 2023;4:102025. [DOI] [PubMed] [PMC]
    Terán-Ángel G, Montes-Berrueta D, Camilo Valencia-Molina J, Carlos Gabaldon-Figueira J, Alejandro Bastidas-Azuaje M, Peterson DL, et al. Identification of Nef-HIV-1 domains involved in p22-phox interaction and superoxide production. Invest Clin. 2016;57:30417. [PubMed]
    Andersen JL, Le Rouzic E, Planelles V. HIV-1 Vpr: mechanisms of G2 arrest and apoptosis. Exp Mol Pathol. 2008;85:210. [DOI] [PubMed] [PMC]
    Li D, Lopez A, Sandoval C, Nichols Doyle R, Fregoso OI. HIV Vpr modulates the host DNA damage response at two independent steps to damage DNA and repress double-strand DNA break repair. mBio. 2020;11:e00940-20. [DOI] [PubMed] [PMC]
    Hoshino S, Konishi M, Mori M, Shimura M, Nishitani C, Kuroki Y, et al. HIV-1 Vpr induces TLR4/MyD88-mediated IL-6 production and reactivates viral production from latency. J Leukoc Biol. 2010;87:113343. [DOI] [PubMed]
    Shao J, Liu G, Lv G. Mutation in the D1 domain of CD4 receptor modulates the binding affinity to HIV-1 gp120. RSC Adv. 2023;13:207080. [DOI] [PubMed] [PMC]
    Gao R, Fang Q, Zhang X, Xu Q, Ye H, Guo W, et al. R5 HIV-1 gp120 activates p38 MAPK to induce rat cardiomyocyte injury by the CCR5 coreceptor. Pathobiology. 2019;86:27484. [DOI] [PubMed]
    Kanmogne GD, Primeaux C, Grammas P. Induction of apoptosis and endothelin-1 secretion in primary human lung endothelial cells by HIV-1 gp120 proteins. Biochem Biophys Res Commun. 2005;333:110715. [DOI] [PubMed]
    Yin Y, Zhou Z, Liu W, Chang Q, Sun G, Dai Y. Vascular endothelial cells senescence is associated with NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation via reactive oxygen species (ROS)/thioredoxin-interacting protein (TXNIP) pathway. Int J Biochem Cell Biol. 2017;84:22-34. [DOI] [PubMed]
    Perkins MV, Joseph SB, Dittmer DP, Mackman N. Cardiovascular disease and thrombosis in HIV infection. Arterioscler Thromb Vasc Biol. 2023;43:17591. [DOI] [PubMed]
    Wanjalla CN, McDonnell WJ, Barnett L, Simmons JD, Furch BD, Lima MC, et al. Adipose tissue in persons with HIV is enriched for CD4+ T effector memory and T effector memory RA+ cells, which show Higher CD69 Expression and CD57, CX3CR1, GPR56 Co-expression With Increasing Glucose Intolerance. Front Immunol. 2019;10:408. [DOI] [PubMed] [PMC]
    Couturier J, Lewis DE. HIV persistence in adipose tissue reservoirs. Curr HIV/AIDS Rep. 2018;15:6071. [DOI] [PubMed] [PMC]
    Roever L, Resende ES, Diniz AL, Penha-Silva N, O’Connell JL, Gomes PFS, et al. Statins in adult patients with HIV: protocol for a systematic review and network meta-analysis. Medicine (Baltimore). 2018;97:e0116. [DOI] [PubMed] [PMC]
    Wang Z, Peters BA, Usyk M, Xing J, Hanna DB, Wang T, et al. Gut microbiota, plasma metabolomic profiles, and carotid artery atherosclerosis in HIV infection. Arterioscler Thromb Vasc Biol. 2022;42:108193. [DOI] [PubMed] [PMC]
    Hsue PY. Mechanisms of cardiovascular disease in the setting of HIV infection. Can J Cardiol. 2019;35:23848. [DOI] [PubMed]
    Cheney L, Hou JC, Morrison S, Pessin J, Steigbigel RT. Nef inhibits glucose uptake in adipocytes and contributes to insulin resistance in human immunodeficiency virus type I infection. J Infect Dis. 2011;203:182431. [DOI] [PubMed] [PMC]
    Bruder-Nascimento T, Kress TC, Kennard S, Belin de Chantemèle EJ. HIV protease inhibitor ritonavir impairs endothelial function via reduction in adipose mass and endothelial leptin receptor-dependent increases in NADPH oxidase 1 (Nox1), C-C chemokine receptor type 5 (CCR5), and inflammation. J Am Heart Assoc. 2020;9:e018074. [DOI] [PubMed] [PMC]
    Nolan D, Watts GF, Herrmann SE, French MA, John M, Mallal S. Endothelial function in HIV-infected patients receiving protease inhibitor therapy: does immune competence affect cardiovascular risk? QJM. 2003;96:82532. [DOI] [PubMed]
    Vos AG, Venter WDF. Cardiovascular toxicity of contemporary antiretroviral therapy. Curr Opin HIV AIDS. 2021;16:28691. [DOI] [PubMed]
    Feinstein MJ, Bogorodskaya M, Bloomfield GS, Vedanthan R, Siedner MJ, Kwan GF, et al. Cardiovascular complications of HIV in endemic countries. Curr Cardiol Rep. 2016;18:113. [DOI] [PubMed] [PMC]
    Fanari Z, Hammami S, Hammami MB, Weintraub WS, Qureshi WA. Acute coronary syndrome in HIV naïve patient with low CD4 count and no other significant risk factors: case report and literature review. Open J Clin Med Case Rep. 2015;1:1009. [PubMed] [PMC]
    Alsheikh MM, Alsheikh AM. Risk of myocardial infarction in HIV patients: a systematic review. Cureus. 2022;14:e31825. [DOI] [PubMed] [PMC]
    Feinstein MJ, Bahiru E, Achenbach C, Longenecker CT, Hsue P, So-Armah K, et al. Patterns of cardiovascular mortality for HIV-infected adults in the United States: 1999 to 2013. Am J Cardiol. 2016;117:21420. [DOI] [PubMed] [PMC]
    Ntsekhe M, Baker JV. Cardiovascular disease among persons living with HIV: new insights into pathogenesis and clinical manifestations in a global context. Circulation. 2023;147:83100. [DOI] [PubMed]
    Kearns AC, Liu F, Dai S, Robinson JA, Kiernan E, Tesfaye Cheru L, et al. Caspase-1 activation is related with HIV-associated atherosclerosis in an HIV transgenic mouse model and HIV patient cohort. Arterioscler Thromb Vasc Biol. 2019;39:176275. [DOI] [PubMed] [PMC]
    Yearley JH, Xia D, Pearson CB, Carville A, Shannon RP, Mansfield KG. Interleukin-18 predicts atherosclerosis progression in SIV-infected and uninfected rhesus monkeys (Macaca mulatta) on a high-fat/high-cholesterol diet. Lab Invest. 2009;89:65767. [DOI] [PubMed] [PMC]
    Hag AMF, Kristoffersen US, Pedersen SF, Gutte H, Lebech AM, Kjaer A. Regional gene expression of LOX-1, VCAM-1, and ICAM-1 in aorta of HIV-1 transgenic rats. PLoS One. 2009;4:e8170. [DOI] [PubMed] [PMC]
    Visser MR, Vercellotti GM. Herpes simplex virus and atherosclerosis. Eur Heart J. 1993;14:3942. [PubMed]
    Chiu B. Multiple infections in carotid atherosclerotic plaques. Am Heart J. 1999;138:S5346. [DOI] [PubMed]
    Mazzaccaro D, Dolci M, Perego F, Delbue S, Giannetta M, Cardani R, et al. Viral agents and systemic levels of inflammatory cytokines in vulnerable and stable atherosclerotic carotid plaques. Ann Vasc Surg. 2022;82:32533. [DOI] [PubMed]
    Raza-Ahmad A, Klassen GA, Murphy DA, Sullivan JA, Kinley CE, Landymore RW, et al. Evidence of type 2 herpes simplex infection in human coronary arteries at the time of coronary artery bypass surgery. Can J Cardiol. 1995;11:10259. [PubMed]
    Nagel MA, Choe A, Khmeleva N, Overton L, Rempel A, Wyborny A, et al. Search for varicella zoster virus and herpes simplex virus-1 in normal human cerebral arteries. J Neurovirol. 2013;19:1815. [DOI] [PubMed] [PMC]
    Etingin OR, Silverstein RL, Hajjar DP. Identification of a monocyte receptor on herpesvirus-infected endothelial cells. Proc Natl Acad Sci U S A. 1991;88:72003. [DOI] [PubMed] [PMC]
    Chirathaworn C, Pongpanich A, Poovorawan Y. Herpes simplex virus 1 induced LOX-1 expression in an endothelial cell line, ECV 304. Viral Immunol. 2004;17:30814. [DOI] [PubMed]
    Zhang X, Tang Q, Xu L. Herpes simplex virus 2 infects human endothelial ECV304 cells and induces cell apoptosis synergistically with ox-LDL. J Toxicol Sci. 2014;39:90917. [DOI] [PubMed]
    Gershom ES, Sutherland MR, Lollar P, Pryzdial ELG. Involvement of the contact phase and intrinsic pathway in herpes simplex virus-initiated plasma coagulation. J Thromb Haemost. 2010;8:103743. [DOI] [PubMed]
    Alber DG, Powell KL, Vallance P, Goodwin DA, Grahame-Clarke C. Herpesvirus infection accelerates atherosclerosis in the apolipoprotein E-deficient mouse. Circulation. 2000;102:77985. [DOI] [PubMed]
    Tudorache IF, Trusca VG, Gafencu AV. Apolipoprotein E - a multifunctional protein with implications in various pathologies as a result of its structural features. Comput Struct Biotechnol J. 2017;15:35965. [DOI] [PubMed] [PMC]
    Wuest TR, Carr DJJ. The role of chemokines during herpes simplex virus-1 infection. Front Biosci. 2008;13:486272. [DOI] [PubMed] [PMC]
    Van de Walle GR, Jarosinski KW, Osterrieder N. Alphaherpesviruses and chemokines: pas de deux not yet brought to perfection. J Virol. 2008;82:60907. [DOI] [PubMed] [PMC]
    Mendy A, Vieira ER, Gasana J. Seropositivity to herpes simplex virus type 2, but not type 1 is associated with premature cardiovascular diseases: a population-based cross-sectional study. Atherosclerosis. 2013;231:1821. [DOI] [PubMed]
    Yaiw KC, Ovchinnikova O, Taher C, Mohammad AA, Davoudi B, Shlyakhto E, et al. High prevalence of human cytomegalovirus in carotid atherosclerotic plaques obtained from Russian patients undergoing carotid endarterectomy. Herpesviridae. 2013;4:3. [DOI] [PubMed] [PMC]
    Mundkur LA, Rao VS, Hebbagudi S, Shanker J, Shivanandan H, Nagaraj RK, et al. Pathogen burden, cytomegalovirus infection and inflammatory markers in the risk of premature coronary artery disease in individuals of Indian origin. Exp Clin Cardiol. 2012;17:638. [PubMed] [PMC]
    Kirkham F, Pera A, Simanek AM, Bano A, Morrow G, Reus B, et al. Cytomegalovirus infection is associated with an increase in aortic stiffness in older men which may be mediated in part by CD4 memory T-cells. Theranostics. 2021;11:572841. [DOI] [PubMed] [PMC]
    Lv YL, Jia Y, Wan Z, An ZL, Yang S, Han FF, et al. Curcumin inhibits the formation of atherosclerosis in ApoE-/- mice by suppressing cytomegalovirus activity in endothelial cells. Life Sci. 2020;257:117658. [DOI] [PubMed]
    He H, Tan Y, Tang Z, Wang L, Liu S, Wu G. ADAM9: a regulator between HCMV infection and function of smooth muscle cells. J Med Virol. 2023;95:e28352. [DOI] [PubMed]
    Yonemitsu Y, Kaneda Y, Komori K, Hirai K, Sugimachi K, Sueishi K. The immediate early gene of human cytomegalovirus stimulates vascular smooth muscle cell proliferation in vitro and in vivo. Biochem Biophys Res Commun. 1997;231:44751. [DOI] [PubMed]
    Popović M, Paskas S, Zivković M, Burysek L, Laumonnier Y. Human cytomegalovirus increases HUVEC sensitivity to thrombin and modulates expression of thrombin receptors. J Thromb Thrombolysis. 2010;30:16471. [DOI] [PubMed]
    Bouwman JJM, Visseren FLJ, Bosch MC, Bouter KP, Diepersloot RJA. Procoagulant and inflammatory response of virus-infected monocytes. Eur J Clin Invest. 2002;32:75966. [DOI] [PubMed]
    Tang-Feldman YJ, Lochhead SR, Lochhead GR, Yu C, George M, Villablanca AC, et al. Murine cytomegalovirus (MCMV) infection upregulates P38 MAP kinase in aortas of Apo E KO mice: a molecular mechanism for MCMV-induced acceleration of atherosclerosis. J Cardiovasc Transl Res. 2013;6:5464. [DOI] [PubMed] [PMC]
    Lebedeva A, Maryukhnich E, Grivel JC, Vasilieva E, Margolis L, Shpektor A. Productive cytomegalovirus infection is associated with impaired endothelial function in ST-elevation myocardial infarction. Am J Med. 2020;133:13342. [DOI] [PubMed] [PMC]
    Hamilton EM, E Allen N, Mentzer AJ, Littlejohns TJ. Human cytomegalovirus and risk of incident cardiovascular disease in United Kingdom Biobank. J Infect Dis. 2022;225:117988. [DOI] [PubMed] [PMC]
    Cristescu CV, Alain S, Ruță SM. The role of CMV infection in primary lesions, development and clinical expression of atherosclerosis. J Clin Med. 2022;11:3832. [DOI] [PubMed] [PMC]
    Wahlin B, Fasth AER, Karp K, Lejon K, Malmström V, Rahbar A, et al. Atherosclerosis in rheumatoid arthritis: associations between anti-cytomegalovirus IgG antibodies, CD4+CD28null T-cells, CD8+CD28null T-cells and intima-media thickness. Clin Exp Rheumatol. 2021;39:57886. [DOI] [PubMed]
    Jia YJ, Liu J, Han FF, Wan ZR, Gong LL, Liu H, et al. Cytomegalovirus infection and atherosclerosis risk: a meta-analysis. J Med Virol. 2017;89:2196206. [DOI] [PubMed]
    Nikitskaya EA, Grivel JC, Maryukhnich EV, Lebedeva AM, Ivanova OI, Savvinova PP, et al. Cytomegalovirus in plasma of acute coronary syndrome patients. Acta Naturae. 2016;8:1027. [PubMed] [PMC]
    Nikitskaya E, Lebedeva A, Ivanova O, Maryukhnich E, Shpektor A, Grivel JC, et al. Cytomegalovirus-productive infection is associated with acute coronary syndrome. J Am Heart Assoc. 2016;5:e003759. [DOI] [PubMed] [PMC]
    Beyaz MO, Ugurlucan M, Oztas DM, Meric M, Conkbayir C, Agacfidan A, et al. Evaluation of the relationship between plaque formation leading to symptomatic carotid artery stenosis and cytomegalovirus by investigating the virus DNA. Arch Med Sci Atheroscler Dis. 2019;4:e1924. [DOI] [PubMed] [PMC]
    Izadi M, Zamani MM, Sabetkish N, Abolhassani H, Saadat SH, Taheri S, et al. The probable role of cytomegalovirus in acute myocardial infarction. Jundishapur J Microbiol. 2014;7:e9253. [DOI] [PubMed] [PMC]
    Hagiwara N, Toyoda K, Inoue T, Shimada H, Ibayashi S, Iida M, et al. Lack of association between infectious burden and carotid atherosclerosis in Japanese patients. J Stroke Cerebrovasc Dis. 2007;16:14552. [DOI] [PubMed]
    Khairy P, Rinfret S, Tardif JC, Marchand R, Shapiro S, Brophy J, et al. Absence of association between infectious agents and endothelial function in healthy young men. Circulation. 2003;107:196671. [DOI] [PubMed]
    Xenaki E, Hassoulas J, Apostolakis S, Sourvinos G, Spandidos DA. Detection of cytomegalovirus in atherosclerotic plaques and nonatherosclerotic arteries. Angiology. 2009;60:5048. [DOI] [PubMed]
    Hepatitis C [Internet]. HIV and AIDS. Geneva: World Health Organization; c2023 [cited 2022 May 15]. Available from: https://www.who.int/news-room/fact-sheets/detail/hepatitis-c
    Negro F, Forton D, Craxı̀ A, Sulkowski MS, Feld JJ, Manns MP. Extrahepatic morbidity and mortality of chronic hepatitis C. Gastroenterology. 2015;149:134560. [DOI] [PubMed]
    Haykal M, Matsumori A, Saleh A, Fayez M, Negm H, Shalaby M, et al. Diagnosis and treatment of HCV heart diseases. Expert Rev Cardiovasc Ther. 2021;19:4939. [DOI] [PubMed]
    Goossens N, Negro F. Cardiovascular manifestations of hepatitis C virus. Clin Liver Dis. 2017;21:46573. [DOI] [PubMed]
    Fletcher NF, Wilson GK, Murray J, Hu K, Lewis A, Reynolds GM, et al. Hepatitis C virus infects the endothelial cells of the blood-brain barrier. Gastroenterology. 2012;142:63443. [DOI] [PubMed] [PMC]
    Muñoz-Hernández R, Ampuero J, Millán R, Gil-Gómez A, Rojas Á, Macher HC, et al. Hepatitis C virus clearance by direct-acting antivirals agents improves endothelial dysfunction and subclinical atherosclerosis: HEPCAR study. Clin Transl Gastroenterol. 2020;11:e00203. [DOI] [PubMed] [PMC]
    Boddi M, Abbate R, Chellini B, Giusti B, Solazzo V, Soft F, et al. HCV infection facilitates asymptomatic carotid atherosclerosis: preliminary report of HCV RNA localization in human carotid plaques. Dig Liver Dis. 2007;39:S5560. [DOI] [PubMed]
    Abouelasrar Salama S, Lavie M, De Buck M, Van Damme J, Struyf S. Cytokines and serum amyloid A in the pathogenesis of hepatitis C virus infection. Cytokine Growth Factor Rev. 2019;50:2942. [DOI] [PubMed]
    Ramachandran A, Kumar B, Waris G, Everly D. Deubiquitination and activation of the NLRP3 inflammasome by UCHL5 in HCV-infected cells. Microbiol Spectr. 2021;9:e0075521. [DOI] [PubMed] [PMC]
    Che Noh I, Avoi R, Abdullah Nurul A, Ahmad I, Abu Bakar R. Analysis of serum and gene expression profile of cytokines (IL-6, TNF-α and TGF-β1) in chronic hepatitis C virus infection. PeerJ. 2022;10:e13330. [DOI] [PubMed] [PMC]
    Cheng PN, Sun HY, Feng IC, Wang ST, Chiu YC, Chiu HC, et al. Reversibility of some oxidative stress markers in chronic hepatitis C patients after receiving direct-acting antiviral agents. J Virus Erad. 2023;9:100318. [DOI] [PubMed] [PMC]
    Schank M, Zhao J, Wang L, Nguyen LNT, Cao D, Dang X, et al. Oxidative stress induces mitochondrial compromise in CD4 T cells from chronically HCV-infected individuals. Front Immunol. 2021;12:760707. [DOI] [PubMed] [PMC]
    Lin D, Chen Y, Koksal AR, Dash S, Aydin Y. Targeting ER stress/PKA/GSK-3β/β-catenin pathway as a potential novel strategy for hepatitis C virus-infected patients. Cell Commun Signal. 2023;21:102. [DOI] [PubMed] [PMC]
    Babiker A, Hassan M, Muhammed S, Taylor G, Poonia B, Shah A, et al. Inflammatory and cardiovascular diseases biomarkers in chronic hepatitis C virus infection: a review. Clin Cardiol. 2020;43:22234. [DOI] [PubMed] [PMC]
    Alzahrani N. Hepatitis C virus, insulin resistance, and diabetes: a review. Microbiol Immunol. 2022;66:4539. [DOI] [PubMed]
    Parvaiz F, Manzoor S, Tariq H, Javed F, Fatima K, Qadri I. Hepatitis C virus infection: molecular pathways to insulin resistance. Virol J. 2011;8:474. [DOI] [PubMed] [PMC]
    Mishra PR, Bharti A, Arora R, Mir IA, Punia VPS. Increased insulin resistance in hepatitis-C infection—association with altered hepatic function testing. Pathophysiology. 2022;29:32632. [DOI] [PubMed] [PMC]
    Gupta S, Read SA, Shackel NA, Hebbard L, George J, Ahlenstiel G. The role of micronutrients in the infection and subsequent response to hepatitis C virus. Cells. 2019;8:603. [DOI] [PubMed] [PMC]
    Hosui A, Tanimoto T, Okahara T, Ashida M, Ohnishi K, Wakahara Y, et al. Oral zinc supplementation decreases the risk of HCC development in patients with HCV eradicated by DAA. Hepatol Commun. 2021;5:20018. [DOI] [PubMed] [PMC]
    Attallah AM, Omran D, Abdelrazek MA, Hassany M, Saif S, Farid A, et al. IL28B rs12979860 polymorphism and zinc supplementation affect treatment outcome and liver fibrosis after direct-acting antiviral hepatitis C therapy. J Genet Eng Biotechnol. 2021;19:150. [DOI] [PubMed] [PMC]
    Suda T, Okawa O, Shirahashi R, Tokutomi N, Tamano M. Changes in serum zinc levels in hepatitis C patients before and after treatment with direct-acting antiviral agents. Hepatol Res. 2019;49:13536. [DOI] [PubMed]
    Himoto T, Hosomi N, Nakai S, Deguchi A, Kinekawa F, Matsuki M, et al. Efficacy of zinc administration in patients with hepatitis C virus-related chronic liver disease. Scand J Gastroenterol. 2007;42:107887. [DOI] [PubMed]
    Yu JW, Sun LJ, Liu W, Zhao YH, Kang P, Yan BZ. Hepatitis C virus core protein induces hepatic metabolism disorders through down-regulation of the SIRT1-AMPK signaling pathway. Int J Infect Dis. 2013;17:e53945. [DOI] [PubMed]
    Miyazaki T, Honda A, Ikegami T, Saitoh Y, Hirayama T, Hara T, et al. Hepatitis C virus infection causes hypolipidemia regardless of hepatic damage or nutritional state: an epidemiological survey of a large Japanese cohort. Hepatol Res. 2011;41:53041. [DOI] [PubMed]
    Grammatikos G, Ferreiros N, Bon D, Schwalm S, Dietz J, Berkowski C, et al. Variations in serum sphingolipid levels associate with liver fibrosis progression and poor treatment outcome in hepatitis C virus but not hepatitis B virus infection. Hepatology. 2015;61:81222. [DOI] [PubMed]
    Gomes D, Sobolewski C, Conzelmann S, Schaer T, Lefai E, Alfaiate D, et al. ANGPTL4 is a potential driver of HCV-induced peripheral insulin resistance. Sci Rep. 2023;13:6767. [DOI] [PubMed] [PMC]
    Mustafa M, Hussain S, Qureshi S, Malik SA, Kazmi AR, Naeem M. Study of the effect of antiviral therapy on homocysteinemia in hepatitis C virus- infected patients. BMC Gastroenterol. 2012;12:117. [DOI] [PubMed] [PMC]
    Canavesi E, Porzio M, Ruscica M, Rametta R, Macchi C, Pelusi S, et al. Increased circulating adiponectin in males with chronic HCV hepatitis. Eur J Intern Med. 2015;26:6359. [DOI] [PubMed]
    Habas E Sr, Farfar KL, Errayes N, Habas AM, Errayes M, Alfitori G, et al. Hepatitis virus C-associated nephropathy: a review and update. Cureus. 2022;14:e27322. [DOI] [PubMed] [PMC]
    Ishizaka N, Ishizaka Y, Takahashi E, Tooda Ei, Hashimoto H, Nagai R, et al. Association between hepatitis C virus seropositivity, carotid-artery plaque, and intima-media thickening. Lancet. 2002;359:1335. [DOI] [PubMed]
    Ishizaka Y, Ishizaka N, Takahashi E, Unuma T, Tooda Ei, Hashimoto H, et al. Association between hepatitis C virus core protein and carotid atherosclerosis. Circ J. 2003;67:2630. [DOI] [PubMed]
    Tomiyama H, Arai T, Hirose K, Hori S, Yamamoto Y, Yamashina A. Hepatitis C virus seropositivity, but not hepatitis B virus carrier or seropositivity, associated with increased pulse wave velocity. Atherosclerosis. 2003;166:4013. [DOI] [PubMed]
    Tien PC, Schneider MF, Cole SR, Cohen MH, Glesby MJ, Lazar J, et al. Association of hepatitis C virus and HIV infection with subclinical atherosclerosis in the women’s interagency HIV study. AIDS. 2009;23:17814. [DOI] [PubMed] [PMC]
    Miyajima I, Kawaguchi T, Fukami A, Nagao Y, Adachi H, Sasaki S, et al. Chronic HCV infection was associated with severe insulin resistance and mild atherosclerosis: a population-based study in an HCV hyperendemic area. J Gastroenterol. 2013;48:93100. [DOI] [PubMed]
    Olubamwo OO, Onyeka IN, Miettola J, Kauhanen J, Tuomainen TP. Hepatitis C as a risk factor for carotid atherosclerosis – a systematic review. Clin Physiol Funct Imaging. 2016;36:24960. [DOI] [PubMed]
    Lee KK, Stelzle D, Bing R, Anwar M, Strachan F, Bashir S, et al. Global burden of atherosclerotic cardiovascular disease in people with hepatitis C virus infection: a systematic review, meta-analysis, and modeling study. Lancet Gastroenterol Hepatol. 2019;4:794804. [DOI] [PubMed] [PMC]
    Vassalle C, Masini S, Bianchi F, Zucchelli GC. Evidence for association between hepatitis C virus seropositivity and coronary artery disease. Heart. 2004;90:5656. [DOI] [PubMed] [PMC]
    Salam RAE, Nabil B, Saber M, AbdelWahab HA, Saber T. Prevalence of hepatitis C virus seropositivity and its impact on coronary artery disease among Egyptian patients referred for coronary angiography. Cardiol Res Pract. 2016;2016:1623197. [DOI] [PubMed] [PMC]
    Wu A, Burrowes S, Zisman E, Brown TT, Bagchi S. Association of hepatitis C infection and acute coronary syndrome: a case-control study. Medicine (Baltimore). 2021;100:e26033. [DOI] [PubMed] [PMC]
    Arcari CM, Nelson KE, Netski DM, Nieto FJ, Gaydos CA. No association between hepatitis C virus seropositivity and acute myocardial infarction. Clin Infect Dis. 2006;43:e536. [DOI] [PubMed]
    Ambrosino P, Lupoli R, Di Minno A, Tarantino L, Spadarella G, Tarantino P, et al. The risk of coronary artery disease and cerebrovascular disease in patients with hepatitis C: a systematic review and meta-analysis. Int J Cardiol. 2016;221:74654. [DOI] [PubMed]
    Olubamwo OO, Aregbesola AO, Miettola J, Kauhanen J, Tuomainen TP, et al. Hepatitis C and risk of coronary atherosclerosis – a systematic review. Public Health. 2016;138:1225. [DOI] [PubMed]
    Fabrizi F, Donato FM, Messa P. Hepatitis C and its metabolic complications in kidney disease. Ann Hepatol. 2017;16:85161. [DOI] [PubMed]
    Wang PC, Wu YF, Lin MS, Lin CL, Chang ML, Chang ST, et al. The impact of hepatitis C virus, metabolic disturbance, and unhealthy behavior on chronic kidney disease: a secondary cross-sectional analysis. Int J Environ Res Public Health. 2022;19:3558. [DOI] [PubMed] [PMC]
    Lee MH, Yang HI, Wang CH, Jen CL, Yeh SH, Liu CJ, et al. Hepatitis C virus infection and increased risk of cerebrovascular disease. Stroke. 2010;41:2894900. [DOI] [PubMed]
    Younossi ZM, Stepanova M, Nader F, Younossi Z, Elsheikh E. Associations of chronic hepatitis C with metabolic and cardiac outcomes. Aliment Pharmacol Ther. 2013;37:64752. [DOI] [PubMed]
    Butt AA, Khan UA, McGinnis KA, Skanderson M, Kent Kwoh C. Co-morbid medical and psychiatric illness and substance abuse in HCV-infected and uninfected veterans. J Viral Hepat. 2007;14:8906. [DOI] [PubMed]
    Huang H, Kang R, Zhao Z. Hepatitis C virus infection and risk of stroke: a systematic review and meta-analysis. PLoS One. 2013;8:e81305. [DOI] [PubMed] [PMC]
    Pavicic Ivelja M, Ivic I, Dolic K, Mestrovic A, Perkovic N, Jankovic S. Evaluation of cerebrovascular reactivity in chronic hepatitis C patients using transcranial color Doppler. PLoS One. 2019;14:e0218206. [DOI] [PubMed] [PMC]
    Lu MC, Wu YH, Chung CH, Lin HH, Hsieh TY, Chen PJ, et al. Association of hepatitis B and C virus with the risk of coronary artery disease and cerebrovascular disease in patients with hepatocellular carcinoma. J Clin Med. 2023;12:2602. [DOI] [PubMed] [PMC]
    Matsumori A, Ohashi N, Nishio R, Kakio T, Hara M, Furukawa Y, et al. Apical hypertrophic cardiomyopathy and hepatitis C virus infection. Jpn Circ J. 1999;63:4338. [DOI] [PubMed]
    Matsumori A, Yutani C, Ikeda Y, Kawai S, Sasayama S. Hepatitis C virus from the hearts of patients with myocarditis and cardiomyopathy. Lab Invest. 2000;80:113742. [DOI] [PubMed]
    Tschöpe C, Ammirati E, Bozkurt B, Caforio ALP, Cooper LT, Felix SB, et al. Myocarditis and inflammatory cardiomyopathy: current evidence and future directions. Nat Rev Cardiol. 2021;18:16993. [DOI] [PubMed] [PMC]
    Guiltinan AM, Kaidarova Z, Custer B, Orland J, Strollo A, Cyrus S, et al. Increased all-cause, liver, and cardiac mortality among hepatitis C virus-seropositive blood donors. Am J Epidemiol. 2008;167:74350. [DOI] [PubMed] [PMC]
    Vajdic CM, Marashi Pour S, Olivier J, Swart A, O’Connell DL, Falster MO, et al. The impact of blood-borne viruses on cause-specific mortality among opioid dependent people: an Australian population-based cohort study. Drug Alcohol Depend. 2015;152:26471. [DOI] [PubMed]
    Fabrizi F, Dixit V, Messa P. Hepatitis C virus and mortality among patients on dialysis: a systematic review and meta-analysis. Clin Res Hepatol Gastroenterol. 2019;43:24454. [DOI] [PubMed]
    Wu VCC, Huang CH, Wang CL, Lin MH, Kuo TY, Chang CH, et al. Cardiovascular outcomes in hepatitis C virus infected patients treated with direct acting antiviral therapy: a retrospective multi-institutional study. Eur Heart J Cardiovasc Pharmacother. 2023;9:50714. [DOI] [PubMed]
    Vassalle C, Meloni A, Pistoia L, Pepe A. Liver pancreas heart triangle and HCV in Thalassemia: expanding the horizon through biomarker networks. Int J Hematol Ther. 2017;3:16. [DOI]
    Origa R. Hepatitis C and thalassemia: a story with (almost) a happy ending. Pathogens. 2023;12:683. [DOI] [PubMed] [PMC]
    Akhtar S, Nasir JA, Hinde A. The prevalence of hepatitis C virus infection in β-thalassemia patients in Pakistan: a systematic review and meta-analysis. BMC Public Health. 2020;20:587. [DOI] [PubMed] [PMC]
    Yasmeen H, Hasnain S. Epidemiology and risk factors of transfusion transmitted infections in thalassemia major: a multicenter study in Pakistan. Hematol Transfus Cell Ther. 2019;41:31623. [DOI] [PubMed] [PMC]
    Meloni A, Pistoia L, Gamberini MR, Spasiano A, Cuccia L, Allò M, et al. The impact of HCV chronic positivity and clearance on extrahepatic morbidity in thalassemia major patients: an observational study from MIOT Network. Eur J Intern Med. 2023;114:93100. [DOI] [PubMed]
    Pepe A, Meloni A, Borsellino Z, Cuccia L, Borgna-Pignatti C, Maggio A, et al. Myocardial fibrosis by late gadolinium enhancement cardiac magnetic resonance and hepatitis C virus infection in thalassemia major patients. J Cardiovasc Med (Hagerstown). 2015;16:68995. [DOI] [PubMed]
    Meloni A, Pistoia L, Maffei S, Ricchi P, Casini T, Corigliano E, et al. Bone status and HCV infection in thalassemia major patients. Bone. 2023;169:116671. [DOI] [PubMed]
    El-Asrar MA, Elbarbary NS, Ismail EA, Elshenity AM. Serum YKL-40 in young patients with β-thalassemia major: relation to hepatitis C virus infection, liver stiffness by transient elastography and cardiovascular complications. Blood Cells Mol Dis. 2016;56:18. [DOI] [PubMed]
    Du Y, Wang C, Zhang Y. Viral coinfections. Viruses. 2022;14:2645. [DOI] [PubMed] [PMC]
    Sosner P, Wangermez M, Chagneau-Derrode C, Le Moal G, Silvain C. Atherosclerosis risk in HIV-infected patients: the influence of hepatitis C virus co-infection. Atherosclerosis. 2012;222:2747. [DOI] [PubMed]
    Chen X, Liu X, Duan S, Tang R, Zhou S, Ye R, et al. Plasma inflammatory biomarkers associated with advanced liver fibrosis in HIV–HCV-coinfected individuals. Int J Environ Res Public Health. 2020;17:9474. [DOI] [PubMed] [PMC]
    Osibogun O, Ogunmoroti O, Michos ED, Spatz ES, Olubajo B, Nasir K, et al. HIV/HCV coinfection and the risk of cardiovascular disease: a meta-analysis. J Viral Hepat. 2017;24:9981004. [DOI] [PubMed]
    Nordin C, Kohli A, Beca S, Zaharia V, Grant T, Leider J, et al. Importance of hepatitis C coinfection in the development of QT prolongation in HIV-infected patients. J Electrocardiol. 2006;39:199205. [DOI] [PubMed]
    Moriyama M, Hugentobler WJ, Iwasaki A. Seasonality of respiratory viral infections. Annu Rev Virol. 2020;7:83101. [DOI] [PubMed]
    Sooryanarain H, Elankumaran S. Environmental role in influenza virus outbreaks. Annu Rev Anim Biosci. 2015;3:34773. [DOI] [PubMed]
    Ran J, Zhao S, Han L, Liao G, Wang K, Wang MH, et al. A re-analysis in exploring the association between temperature and COVID-19 transmissibility: an ecological study with 154 Chinese cities. Eur Respir J. 2020;56:2001253. [DOI] [PubMed] [PMC]
    Ma Y, Zhao Y, Liu J, He X, Wang B, Fu S, et al. Effects of temperature variation and humidity on the death of COVID-19 in Wuhan, China. Sci Total Environ. 2020;724:138226. [DOI] [PubMed] [PMC]
    Lipsitch M, Viboud C. Influenza seasonality: lifting the fog. Proc Natl Acad Sci U S A. 2009;106:36456. [DOI] [PubMed] [PMC]
    Lowen AC, Mubareka S, Steel J, Palese P. Influenza virus transmission is dependent on relative humidity and temperature. PLoS Pathog. 2007;3:14706. [DOI] [PubMed] [PMC]
    Martins LD, da Silva I, Batista WV, Andrade MF, Freitas ED, Martins JA. How socio-economic and atmospheric variables impact COVID-19 and influenza outbreaks in tropical and subtropical regions of Brazil. Environ Res. 2020;191:110184. [DOI] [PubMed] [PMC]
    Poirier C, Luo W, Majumder MS, Liu D, Mandl KD, Mooring TA, et al. The role of environmental factors on transmission rates of the COVID-19 outbreak: an initial assessment in two spatial scales. Sci Rep. 2020;10:17002. [DOI] [PubMed] [PMC]
    Bukhari Q, Massaro JM, D’Agostino RB Sr, Khan S. Effects of weather on coronavirus pandemic. Int J Environ Res Public Health. 2020;17:5399. [DOI] [PubMed] [PMC]
    Romero Starke K, Mauer R, Karskens E, Pretzsch A, Reissig D, Nienhaus A, et al. The effect of ambient environmental conditions on COVID-19 mortality: a systematic review. Int J Environ Res Public Health. 2021;18:6665. [DOI] [PubMed] [PMC]
    Nichols GL, Gillingham EL, Macintyre HL, Vardoulakis S, Hajat S, Sarran CE, et al. Coronavirus seasonality, respiratory infections and weather. BMC Infect Dis. 2021;21:1101. [DOI] [PubMed] [PMC]
    Haga L, Ruuhela R, Auranen K, Lakkala K, Heikkilä A, Gregow H. Impact of selected meteorological factors on COVID-19 incidence in southern Finland during 2020–2021. Int J Environ Res Public Health. 2022;19:13398. [DOI] [PubMed] [PMC]
    Hosseini V. SARS-CoV-2 virulence: interplay of floating virus-laden particles, climate, and humans. Adv Biosyst. 2020;4:e2000105. [DOI] [PubMed] [PMC]
    Ren SY, Wang WB, Hao YG, Zhang HR, Wang ZC, Chen YL, et al. Stability and infectivity of coronaviruses in inanimate environments. World J Clin Cases. 2020;8:13919. [DOI] [PubMed] [PMC]
    Li Y, Wang X, Nair H. Global seasonality of human seasonal coronaviruses: a clue for postpandemic circulating season of severe acute respiratory syndrome coronavirus 2? J Infect Dis. 2020;222:10907. [DOI] [PubMed] [PMC]
    Maltezou HC, Papanikolopoulou A, Vassiliu S, Theodoridou K, Nikolopoulou G, Sipsas NV. COVID-19 and respiratory virus co-infections: a systematic review of the literature. Viruses. 2023;15:865. [DOI] [PubMed] [PMC]
    Global influenza strategy 2019-2030 [Internet]. Geneva: World Health Organization; c2023 [cited 2022 May 23]. Available from: https://www.who.int/publications/i/item/9789241515320
    Iuliano AD, Roguski KM, Chang HH, Muscatello DJ, Palekar R, Tempia S, et al. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. Lancet. 2018;391:1285300. [DOI] [PubMed] [PMC]
    Collins SD. Excess mortality from causes other than influenza and pneumonia during influenza epidemics. Public Health Rep. 1932;47:2159. [DOI]
    Haidari M, Wyde PR, Litovsky S, Vela D, Ali M, Casscells SW, et al. Influenza virus directly infects, inflames, and resides in the arteries of atherosclerotic and normal mice. Atherosclerosis. 2010;208:906. [DOI] [PubMed]
    Zeng H, Pappas C, Belser JA, Houser KV, Zhong W, Wadford DA, et al. Human pulmonary microvascular endothelial cells support productive replication of highly pathogenic avian influenza viruses: possible involvement in the pathogenesis of human H5N1 virus infection. J Virol. 2012;86:66778. [DOI] [PubMed] [PMC]
    Wang S, Le TQ, Kurihara N, Chida J, Cisse Y, Yano M, et al. Influenza virus—cytokine-protease cycle in the pathogenesis of vascular hyperpermeability in severe influenza. J Infect Dis. 2010;202:9911001. [DOI] [PubMed] [PMC]
    Navab M, Anantharamaiah GM, Reddy ST, Van Lenten BJ, Hough G, Wagner A, et al. Human apolipoprotein AI mimetic peptides for the treatment of atherosclerosis. Curr Opin Investig Drugs. 2003;4:11004. [PubMed]
    Warren-Gash C, Smeeth L, Hayward AC. Influenza as a trigger for acute myocardial infarction or death from cardiovascular disease: a systematic review. Lancet Infect Dis. 2009;9:60110. [DOI] [PubMed]
    Bocale R, Necozione S, Desideri G. The link between influenza and myocardial infarction: vaccination protects. Eur Heart J Suppl. 2022;24:I848. [DOI] [PubMed] [PMC]
    Young-Xu Y, Smith J, Mahmud SM, Van Aalst R, Thommes EW, Neupane N, et al. Laboratory-confirmed influenza infection and acute myocardial infarction among United States senior Veterans. PLoS One. 2020;15:e0243248. [DOI] [PubMed] [PMC]
    McCarthy Z, Xu S, Rahman A, Bragazzi NL, Corrales-Medina VF, Lee J, et al. Modelling the linkage between influenza infection and cardiovascular events via thrombosis. Sci Rep. 2020;10:14264. [DOI] [PubMed] [PMC]
    Rubino R, Imburgia C, Bonura S, Trizzino M, Iaria C, Cascio A. Thromboembolic events in patients with influenza: a scoping review. Viruses. 2022;14:2817. [DOI] [PubMed] [PMC]
    Guan X, Yang W, Sun X, Wang L, Ma B, Li H, et al. Association of influenza virus infection and inflammatory cytokines with acute myocardial infarction. Inflamm Res. 2012;61:5918. [DOI] [PubMed]
    Lee HS, Noh JY, Shin OS, Song JY, Cheong HJ, Kim WJ. Matrix metalloproteinase-13 in atherosclerotic plaque is increased by influenza a virus infection. J Infect Dis. 2020;221:25666. [DOI] [PubMed]
    Muscente F, De Caterina R. Causal relationship between influenza infection and risk of acute myocardial infarction: pathophysiological hypothesis and clinical implications. Eur Heart J Suppl. 2020;22:E6872. [DOI] [PubMed] [PMC]
    Auer J, Leitinger M, Berent R, Prammer W, Weber T, Lassnig E, et al. Influenza A and B IgG seropositivity and coronary atherosclerosis assessed by angiography. Heart Dis. 2002;4:34954. [DOI] [PubMed]
    Caldeira D, Nogueira-Garcia B. Myocardial infarction and viral triggers: what do we know by now? Eur Heart J Suppl. 2023;25:A126. [DOI] [PubMed] [PMC]
    Mohammad MA, Tham J, Koul S, Rylance R, Bergh C, Erlinge D, et al. Association of acute myocardial infarction with influenza: a nationwide observational study. PLoS One. 2020;15:e0236866. [DOI] [PubMed] [PMC]
    Chew DP, Mattschoss S, Horsfall M, Astley C, Vaile JC, Joseph MX. Patterns of inflammatory activation associated with precipitants of acute coronary syndromes: a case-crossover study. Intern Med J. 2012;42:1096103. [DOI] [PubMed]
    Ruane L, Buckley T, Hoo SYS, Hansen PS, McCormack C, Shaw E, et al. Triggering of acute myocardial infarction by respiratory infection. Intern Med J. 2017;47:5229. [DOI] [PubMed]
    Kwong JC, Schwartz KL, Campitelli MA, Chung H, Crowcroft NS, Karnauchow T, et al. Acute myocardial infarction after laboratory-confirmed influenza infection. N Engl J Med. 2018;378:34553. [DOI] [PubMed]
    Tripathi B, Kumar V, Kalra A, Gupta T, Sawant AC, Sharma P, et al. Influence of influenza infection on in-hospital acute myocardial infarction outcomes. Am J Cardiol. 2020;130:714. [DOI] [PubMed]
    Bermúdez-Fajardo A, Oviedo-Orta E. Influenza vaccination promotes stable atherosclerotic plaques in apoE knockout mice. Atherosclerosis. 2011;217:97105. [DOI] [PubMed]
    Udell JA, Zawi R, Bhatt DL, Keshtkar-Jahromi M, Gaughran F, Phrommintikul A, et al. Association between influenza vaccination and cardiovascular outcomes in high-risk patients: a meta-analysis. JAMA. 2013;310:171120. [DOI] [PubMed]
    Hebsur S, Vakil E, Oetgen WJ, Kumar PN, Lazarous DF. Influenza and coronary artery disease: exploring a clinical association with myocardial infarction and analyzing the utility of vaccination in prevention of myocardial infarction. Rev Cardiovasc Med. 2014;15:16875. [DOI] [PubMed]
    Ciszewski A, Bilinska ZT, Brydak LB, Kepka C, Kruk M, Romanowska M, et al. Influenza vaccination in secondary prevention from coronary ischaemic events in coronary artery disease: FLUCAD study. Eur Heart J. 2008;29:13508. [DOI] [PubMed]
    Barnes M, Heywood AE, Mahimbo A, Rahman B, Newall AT, Macintyre CR. Acute myocardial infarction and influenza: a meta-analysis of case–control studies. Heart. 2015;101:173847. [DOI] [PubMed] [PMC]
    Fröbert O, Götberg M, Erlinge D, Akhtar Z, Christiansen EH, MacIntyre CR, et al. Influenza vaccination after myocardial infarction: a randomized, double-blind, placebo-controlled, multicenter trial. Circulation. 2021;144:147684. [DOI] [PubMed]
    Maniar YM, Al-Abdouh A, Michos ED. Influenza vaccination for cardiovascular prevention: further insights from the IAMI trial and an updated meta-analysis. Curr Cardiol Rep. 2022;24:132735. [DOI] [PubMed] [PMC]
    Barbetta LMDS, Correia ETO, Gismondi RAOC, Mesquita ET. Influenza vaccination as prevention therapy for stable coronary artery disease and acute coronary syndrome: a meta-analysis of randomized trials. Am J Med. 2023;136:46675. [DOI] [PubMed]
    Ngwudike CJ, Villalobos A. Correlation between cardiovascular protection and influenza vaccination. Curr Cardiol Rep. 2023;25:5716. [DOI] [PubMed] [PMC]
    Knuuti J, Wijns W, Saraste A, Capodanno D, Barbato E, Funck-Brentano C, et al.; ESC Scientific Document Group. 2019 ESC guidelines for the diagnosis and management of chronic coronary syndromes. Eur Heart J. 2020;41:40777. [DOI] [PubMed]
    Al Rifai M, Khalid U, Misra A, Liu J, Nasir K, Cainzos-Achirica M, et al. Racial and geographic disparities in influenza vaccination in the U.S. among individuals with atherosclerotic cardiovascular disease: renewed importance in the setting of COVID-19. Am J Prev Cardiol. 2021;5:100150. [DOI] [PubMed] [PMC]
    Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323:10619. [DOI] [PubMed] [PMC]
    Kole C, Stefanou Ε, Karvelas N, Schizas D, Toutouzas KP. Acute and post-acute COVID-19 cardiovascular complications: a comprehensive review. Cardiovasc Drugs Ther. 2023:116. [DOI] [PubMed] [PMC]
    Shu H, Wen Z, Li N, Zhang Z, Ceesay BM, Peng Y, et al. COVID-19 and cardiovascular diseases: from cellular mechanisms to clinical manifestations. Aging Dis. 2023. [DOI] [PubMed] [PMC]
    Cook TM. The importance of hypertension as a risk factor for severe illness and mortality in COVID-19. Anaesthesia. 2020;75:9767. [DOI] [PubMed] [PMC]
    Tsai EJ, Čiháková D, Tucker NR. Cell-specific mechanisms in the heart of COVID-19 patients. Circ Res. 2023;132:1290301. [DOI] [PubMed] [PMC]
    Qudus MS, Tian M, Sirajuddin S, Liu S, Afaq U, Wali M, et al. The roles of critical pro-inflammatory cytokines in the drive of cytokine storm during SARS-CoV-2 infection. J Med Virol. 2023;95:e28751. [DOI] [PubMed]
    Szarpak L, Zaczynski A, Kosior D, Bialka S, Ladny JR, Gilis-Malinowska N, et al. Evidence of diagnostic value of ferritin in patients with COVID-19. Cardiol J. 2020;27:8867. [DOI] [PubMed] [PMC]
    Risitano AM, Mastellos DC, Huber-Lang M, Yancopoulou D, Garlanda C, Ciceri F, et al. Complement as a target in COVID-19? Nat Rev Immunol. 2020;20:3434. [DOI] [PubMed] [PMC]
    Teimury A, Khameneh MT, Khaledi EM. Major coagulation disorders and parameters in COVID-19 patients. Eur J Med Res. 2022;27:25. [DOI] [PubMed] [PMC]
    Araújo CRDS, Fernandes J, Caetano DS, Barros AEVDR, de Souza JAF, Machado MDGR, et al. Endothelial function, arterial stiffness and heart rate variability of patients with cardiovascular diseases hospitalized due to COVID-19. Heart Lung. 2023;58:2106. [DOI] [PubMed] [PMC]
    Jahani M, Dokaneheifard S, Mansouri K. Hypoxia: a key feature of COVID-19 launching activation of HIF-1 and cytokine storm. J Inflamm (Lond). 2020;17:33. [DOI] [PubMed] [PMC]
    Wihandani DM, Purwanta MLA, Mulyani WRW, Putra IWAS, Supadmanaba IGP. New-onset diabetes in COVID-19: the molecular pathogenesis. Biomedicine (Taipei). 2023;13:312. [DOI] [PubMed] [PMC]
    Montefusco L, Ben Nasr M, D’Addio F, Loretelli C, Rossi A, Pastore I, et al. Acute and long-term disruption of glycometabolic control after SARS-CoV-2 infection. Nat Metab. 2021;3:77485. [DOI] [PubMed] [PMC]
    Ho G, Ali A, Takamatsu Y, Wada R, Masliah E, Hashimoto M. Diabetes, inflammation, and the adiponectin paradox: therapeutic targets in SARS-CoV-2. Drug Discov Today. 2021;26:203644. [DOI] [PubMed] [PMC]
    Gaggini M, Ndreu R, Michelucci E, Rocchiccioli S, Vassalle C. Ceramides as mediators of oxidative stress and inflammation in cardiometabolic disease. Int J Mol Sci. 2022;23:2719. [DOI] [PubMed] [PMC]
    Gaggini M, Pingitore A, Vassalle C. Plasma ceramides pathophysiology, measurements, challenges, and opportunities. Metabolites. 2021;11:719. [DOI] [PubMed] [PMC]
    Avota E, Bodem J, Chithelen J, Mandasari P, Beyersdorf N, Schneider-Schaulies J. The manifold roles of sphingolipids in viral infections. Front Physiol. 2021;12:715527. [DOI] [PubMed] [PMC]
    Vitner EB, Achdout H, Avraham R, Politi B, Cherry L, Tamir H, et al. Glucosylceramide synthase inhibitors prevent replication of SARS-CoV-2 and influenza virus. J Biol Chem. 2021;296:100470. [DOI] [PubMed] [PMC]
    Fenizia S, Gaggini M, Vassalle C. The sphingolipid-signaling pathway as a modulator of infection by SARS-CoV-2. Curr Issues Mol Biol. 2023;45:795673. [DOI] [PubMed] [PMC]
    Sabanoglu C, Inanc IH, Polat E, Peker SA. Long-term predictive value of cardiac biomarkers in patients with COVID-19 infection. Eur Rev Med Pharmacol Sci. 2022;26:6396403. [DOI] [PubMed]
    Zhao YH, Zhao L, Yang XC, Wang P. Cardiovascular complications of SARS-CoV-2 infection (COVID-19): a systematic review and meta-analysis. Rev Cardiovasc Med. 2021;22:15965. [DOI] [PubMed]
    Zuin M, Rigatelli G, Battisti V, Costola G, Roncon L, Bilato C. Increased risk of acute myocardial infarction after COVID-19 recovery: a systematic review and meta-analysis. Int J Cardiol. 2023;372:13843. [DOI] [PubMed] [PMC]
    Kalinskaya A, Vorobyeva D, Rusakovich G, Maryukhnich E, Anisimova A, Dukhin O, et al. Targeted blood plasma proteomics and hemostasis assessment of post COVID-19 patients with acute myocardial infarction. Int J Mol Sci. 2023;24:6523. [DOI] [PubMed] [PMC]
    Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020;323:123942. [DOI] [PubMed]
    Augustine R, S A, Nayeem A, Salam SA, Augustine P, Dan P, et al. Increased complications of COVID-19 in people with cardiovascular disease: role of the renin–angiotensin-aldosterone system (RAAS) dysregulation. Chem Biol Interact. 2022;351:109738. [DOI] [PubMed] [PMC]
    Fairweather D, Beetler DJ, Di Florio DN, Musigk N, Heidecker B, Cooper LT Jr. COVID-19, myocarditis and pericarditis. Circ Res. 2023;132:130219. [DOI] [PubMed] [PMC]
    Vilaplana-Carnerero C, Giner-Soriano M, Dominguez À, Morros R, Pericas C, Álamo-Junquera D, et al. Atherosclerosis, cardiovascular disease, and COVID-19: a narrative review. Biomedicines. 2023;11:1206. [DOI] [PubMed] [PMC]
    Nicolai L, Kaiser R, Stark K. Thromboinflammation in long COVID—the elusive key to post-infection sequelae? J Thromb Haemost. 2023;21:202031. [DOI] [PubMed] [PMC]
    Morrow AJ, Sykes R, McIntosh A, Kamdar A, Bagot C, Bayes HK, et al. A multisystem, cardio-renal investigation of post-COVID-19 illness. Nat Med. 2022;28:130313. [DOI] [PubMed] [PMC]
    Grosse C, Grosse A, Salzer HJF, Dünser MW, Motz R, Langer R. Analysis of cardiopulmonary findings in COVID-19 fatalities: high incidence of pulmonary artery thrombi and acute suppurative bronchopneumonia. Cardiovasc Pathol. 2020;49:107263. [DOI] [PubMed] [PMC]
    Eiros R, Barreiro-Pérez M, Martín-García A, Almeida J, Villacorta E, Pérez-Pons A, et al.; en representación de los investigadores CCC (cardiac COVID-19 healthcare workers). Afección pericárdica y miocárdica tras infección por SARS-CoV-2: estudio descriptivo transversal en trabajadores sanitarios. Rev Esp Cardiol. 2022;75:73547. Spanish. [DOI] [PubMed] [PMC]
    Fagyas M, Nagy B Jr, Ráduly AP, Mányiné IS, Mártha L, Erdősi G, et al. The majority of severe COVID-19 patients develop anti-cardiac autoantibodies. Geroscience. 2022;44:234760. [DOI] [PubMed] [PMC]
    Bozkurt B, Kovacs R, Harrington B. Joint HFSA/ACC/AHA statement addresses concerns Re: using RAAS antagonists in COVID-19. J Card Fail. 2020;26:370. [DOI] [PubMed] [PMC]
    Long B, Brady WJ, Koyfman A, Gottlieb M. Cardiovascular complications in COVID-19. Am J Emerg Med. 2020;38:15047. [DOI] [PubMed] [PMC]
    Kim MS, Jung SY, Ahn JG, Park SJ, Shoenfeld Y, Kronbichler A, et al. Comparative safety of mRNA COVID-19 vaccines to influenza vaccines: a pharmacovigilance analysis using WHO international database. J Med Virol. 2022;94:108595. [DOI] [PubMed] [PMC]
    Ling RR, Ramanathan K, Tan FL, Tai BC, Somani J, Fisher D, et al. Myopericarditis following COVID-19 vaccination and non-COVID-19 vaccination: a systematic review and meta-analysis. Lancet Respir Med. 2022;10:67988. [DOI] [PubMed] [PMC]
    Massari M, SpilaAlegiani S, Morciano C, Spuri M, Marchione P, Felicetti P, et al. Postmarketing active surveillance of myocarditis and pericarditis following vaccination with COVID-19 mRNA vaccines in persons aged 12 to 39 years in Italy: a multi-database, self-controlled case series study. PLoS Med. 2022;19:e1004056. [DOI] [PubMed] [PMC]
    Wong HL, Hu M, Zhou CK, Lloyd PC, Amend KL, Beachler DC, et al. Risk of myocarditis and pericarditis after the COVID-19 mRNA vaccination in the USA: a cohort study in claims databases. Lancet. 2022;399:21919. [DOI] [PubMed] [PMC]
    Straus W, Urdaneta V, Esposito DB, Mansi JA, Sanz Rodriguez C, Burton P, et al. Analysis of myocarditis among 252 million mRNA-1273 recipients worldwide. Clin Infect Dis. 2023;76:e54452. [DOI] [PubMed] [PMC]
    Schwab C, Domke LM, Hartmann L, Stenzinger A, Longerich T, Schirmacher P. Autopsy-based histopathological characterization of myocarditis after anti-SARS-CoV-2-vaccination. Clin Res Cardiol. 2023;112:43140. [DOI] [PubMed] [PMC]
    Won T, Gilotra NA, Wood MK, Hughes DM, Talor MV, Lovell J, et al. Increased interleukin 18-dependent immune responses are associated with myopericarditis after COVID-19 mRNA vaccination. Front Immunol. 2022;13:851620. [DOI] [PubMed] [PMC]
    Huynh TV, Rethi L, Lee TW, Higa S, Kao YH, Chen YJ. Spike protein impairs mitochondrial function in human cardiomyocytes: mechanisms underlying cardiac injury in COVID-19. Cells. 2023;12:877. [DOI] [PubMed] [PMC]
    Bellavite P, Ferraresi A, Isidoro C. Immune response and molecular mechanisms of cardiovascular adverse effects of spike proteins from SARS-CoV-2 and mRNA vaccines. Biomedicines. 2023;11:451. [DOI] [PubMed] [PMC]
    Cadegiani FA. Catecholamines are the key trigger of COVID-19 mRNA vaccine-induced myocarditis: a compelling hypothesis supported by epidemiological, anatomopathological, molecular, and physiological findings. Cureus. 2022;14:e27883. [DOI] [PubMed] [PMC]
    Mastorci F, Piaggi P, Doveri C, Trivellini G, Casu A, Pozzi M, et al. Health-related quality of life in Italian adolescents during covid-19 outbreak. Front Pediatr. 2021;9:611136. [DOI] [PubMed] [PMC]
    Mastorci F, Bastiani L, Trivellini G, Doveri C, Casu A, Pozzi M, et al. Well-being perception during COVID-19 pandemic in healthy adolescents: from the Avatar study. Int J Environ Res Public Health. 2021;18:6388. [DOI] [PubMed] [PMC]
    Gorini F, Chatzianagnostou K, Mazzone A, Bustaffa E, Esposito A, Berti S, et al. “Acute myocardial infarction in the time of COVID-19”: a review of biological, environmental, and psychosocial contributors. Int J Environ Res Public Health. 2020;17:7371. [DOI] [PubMed] [PMC]
    Tam CCF, Cheung KS, Lam S, Wong A, Yung A, Sze M, et al. Impact of coronavirus disease 2019 (COVID-19) outbreak on ST-segment–elevation myocardial infarction care in Hong Kong, China. Circ Cardiovasc Qual Outcomes. 2020;13:e006631. [DOI] [PubMed] [PMC]
    Marotta M, Gorini F, Parlanti A, Chatzianagnostou K, Mazzone A, Berti S, et al. Fear of COVID-19 in patients with acute myocardial infarction. Int J Environ Res Public Health. 2021;18:9847. [DOI] [PubMed] [PMC]
    Soraci P, Ferrari A, Abbiati FA, Del Fante E, De Pace R, Urso A, et al. Validation and psychometric evaluation of the Italian version of the fear of COVID-19 scale. Int J Ment Health Addict. 2022;20:191322. [DOI] [PubMed] [PMC]
    Marotta M, Gorini F, Parlanti A, Berti S, Vassalle C. Effect of mindfulness-based stress reduction on the well-being, burnout and stress of Italian healthcare professionals during the COVID-19 pandemic. J Clin Med. 2022;11:3136. [DOI] [PubMed] [PMC]
    Lawson JS, Glenn WK, Tran DD, Ngan CC, Duflou JA, Whitaker NJ. Identification of human papilloma viruses in atheromatous coronary artery disease. Front Cardiovasc Med. 2015;2:17. [DOI] [PubMed] [PMC]
    Popović M, Smiljanić K, Dobutović B, Syrovets T, Simmet T, Isenović ER. Human cytomegalovirus infection and atherothrombosis. J Thromb Thrombolysis. 2012;33:16072. [DOI] [PubMed]
    Naghavi M, Wyde P, Litovsky S, Madjid M, Akhtar A, Naguib S, et al. Influenza infection exerts prominent inflammatory and thrombotic effects on the atherosclerotic plaques of apolipoprotein E–deficient mice. Circulation. 2003;107:7628. [DOI] [PubMed]
    Van Lenten BJ, Wagner AC, Anantharamaiah GM, Garber DW, Fishbein MC, Adhikary L, et al. Influenza infection promotes macrophage traffic into arteries of mice that is prevented by D-4F, an apolipoprotein A-I mimetic peptide. Circulation. 2002;106:112732. [DOI] [PubMed]
    Morris SE, Freiesleben de Blasio B, Viboud C, Wesolowski A, Bjørnstad ON, Grenfell BT. Analysis of multi-level spatial data reveals strong synchrony in seasonal influenza epidemics across Norway, Sweden, and Denmark. PLoS One. 2018;13:e0197519. [DOI] [PubMed] [PMC]
    Rittweger J, Gilardi L, Baltruweit M, Dally S, Erbertseder T, Mittag U, et al. Temperature and particulate matter as environmental factors associated with seasonality of influenza incidence – an approach using Earth observation-based modeling in a health insurance cohort study from Baden-Württemberg (Germany). Environ Health. 2022;21:131. [DOI] [PubMed] [PMC]
    Radke PW, Merkelbach-Bruse S, Messmer BJ, vom Dahl J, Dörge H, Naami A, et al. Infectious agents in coronary lesions obtained by endarterectomy: pattern of distribution, coinfection, and clinical findings. Coron Artery Dis. 2001;12:16. [DOI] [PubMed]
    Virok D, Kis Z, Kari L, Barzo P, Sipka R, Burian K, et al. Chlamydophila pneumoniae and human cytomegalovirus in atherosclerotic carotid plaques - combined presence and possible interactions. Acta Microbiol Immunol Hung. 2006;53:3550. [DOI] [PubMed]
    Fernández-Montero JV, Barreiro P, de Mendoza C, Labarga P, Soriano V. Hepatitis C virus coinfection independently increases the risk of cardiovascular disease in HIV-positive patients. J Viral Hepat. 2016;23:4752. [DOI] [PubMed]
    Khan A, El Hosseiny A, Siam R. Assessing and reassessing the association of comorbidities and coinfections in COVID-19 patients. Cureus. 2023;15:e36683. [DOI] [PubMed] [PMC]
    Davis MM, Taubert K, Benin AL, Brown DW, Mensah GA, Baddour LM, et al. Influenza vaccination as secondary prevention for cardiovascular disease: a science advisory from the American Heart Association/American College of Cardiology. J Am Coll Cardiol. 2006;48:1498502. [DOI] [PubMed]
    Elkind MSV. Infectious burden: a new risk factor and treatment target for atherosclerosis. Infect Disord Drug Targets. 2010;10:8490. [DOI] [PubMed] [PMC]
    Cho BH, O’Halloran A, Pike J. Investigation of barriers to county-level seasonal influenza vaccine uptake among Medicare beneficiaries in the United States – 2018–2019 seasonal influenza season. Vaccine X. 2023;14:100326. [DOI] [PubMed] [PMC]
    Yasmin F, Najeeb H, Naeem U, Moeed A, Atif AR, Asghar MS, et al. Adverse events following COVID-19 mRNA vaccines: a systematic review of cardiovascular complication, thrombosis, and thrombocytopenia. Immun Inflamm Dis. 2023;11:e807. [DOI] [PubMed] [PMC]