Next Article in Journal
Predictors of Surgical Intervention for Pediatric Acute Rhinosinusitis with Periorbital Infection
Previous Article in Journal
Moesin Serves as Scaffold Protein for PD-L1 in Human Uterine Cervical Squamous Carcinoma Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of CMV Infection in Primary Lesions, Development and Clinical Expression of Atherosclerosis

by
Carmen Valentina Cristescu
1,2,*,
Sophie Alain
2 and
Simona Maria Ruță
3,4
1
School of Advanced Studies of the Romanian Academy (SCOSAAR), Romanian Academy, 010071 Bucharest, Romania
2
National Center for Cytomegalovirus Research, UMR 1092, 87042 Limoges, France
3
Virology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
4
Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(13), 3832; https://doi.org/10.3390/jcm11133832
Submission received: 23 May 2022 / Revised: 24 June 2022 / Accepted: 30 June 2022 / Published: 1 July 2022
(This article belongs to the Section Cardiology)

Abstract

:
The number of deaths related to cardiovascular disease is increasing every year, despite all available therapies and the aggressive campaigns for lifestyle modification and prevention of risk factors. Atherosclerosis is a complex process underlying cardiovascular disease. Cytomegalovirus (CMV) is often associated to atherosclerosis and its clinical expression such as coronary heart disease, stroke, or peripheral artery disease. CMV infection may promote acute atherosis within placentas from women with preeclampsia and it may also accelerate atherosclerosis in HIV-infected and organ-transplanted patients. This review focuses on the current scientific evidence for the role of CMV infection in the development of acute atherosis and atherosclerosis from placentation throughout life.

1. Introduction

Atherosclerosis is the pathophysiological phenomenon underlying myocardial infarction, stroke, and peripheral artery disease. It is a progressive inflammatory process that can occur in people of all ages [1,2,3], even in childhood [4]. Atherosclerosis is characterized by the accumulation of lipids and secondary fibrosis of the arterial wall, followed by calcification or plaque instability and eventually plaque rupture with clinical events. Both the innate and adaptive immune responses play an important role in the evolution of the disease [5]. The process of plaque formation may extend through one or more decades or can be very fast, evolving within a few months, depending on the type and onset of the immune response. The mechanism of atherosclerosis is still unclear [3,5], although inflammation plays a crucial role and specific lifestyle-related risk factors have a high impact (smoking, dyslipidemia, diabetes, obesity, level of physical activity, and stress). Various infectious agents have been investigated as potential triggers or cofactors for atherosclerosis [6]. Thus, Helicobacter pylori induces chronic systemic inflammation by molecular mimicry. Periodontal pathogens can cause bacteriemia, leading to direct plaque invasion. SARS-CoV-2 generates a cytokine storm and plaque thrombosis. Human herpes simplex virus produces a proinflammatory state with lipid metabolism impairment. Hepatitis C virus increases the expression of cardiac and inflammatory biomarkers. Streptococcus pneumoniae, Chlamydia pneumoniae, and Mycoplasma pneumoniae may induce persistent pro-inflammatory state and platelet activation with subsequent plaque instability [7]. Among the plethora of bacteria and viruses, CMV was frequently detected in both atherosclerotic plaques and in healthy arteries, and its persistence is related to development overtime of atherosclerosis [8].
CMV is an ubiquitous beta herpesvirus, with a global prevalence in general population estimated at 83% [9] and a regional prevalence correlated with the socio-economic level of each country. During the last decades, CMV was incriminated in plaque formation and cardiovascular disease [10] due to its capacity for infecting endothelial cells, macrophages, dendritic cells, fibroblasts, and even smooth muscle cells, and its lifetime latency in different cellular types including monocytes and hematopoietic stem cells, with multiple reactivations that can trigger a chronic inflammatory status [10,11,12,13].

2. Cytomegalovirus and Cardiovascular Disease

2.1. Evidence of CMV’s Role in Plaque Formation and Cardiovascular Disease

2.1.1. Epidemiological Studies

The first observations of a possible correlation between CMV and atherosclerosis were made almost 30 years ago. The presence of anti-CMV IgG antibodies within the sera and the presence of CMV antigens within vascular smooth muscle cells were associated with different stages of plaque formation in patients undergoing vascular surgery at that time [14,15]. Since then, the ongoing research on the implications of CMV infection in cardiovascular disease generated rather variable results, suggesting the importance of the study design and population sampling (Table 1).
Most CMV serology-based studies in patients at risk for cardiovascular diseases have delivered controversial results, except for a certain population group such as patients with diabetes. High titers of anti-CMV IgG antibodies correlate to atherosclerosis in type 2 diabetes mellitus patients, after adjustment of other common cardiovascular risk factors [16]. Patients with diabetes mellitus have an increased risk and an accelerated rate of development of atherosclerosis [17]. There might be a bi-directional link between CMV infection and diabetes, as patients with diabetes mellitus have an impaired antiviral immune response [18] that might promote CMV reactivation, while chronic CMV infection is associated with glucose regulation [19] and new-onset type 2 diabetes mellitus [20]. Nevertheless, all these complex interactions need a better understanding and more research studies in order to clarify the link between CMV infection and diabetes and cardiovascular disease.
CMV direct detection by DNA or antigen identification strongly associates viral infection with atherosclerosis and cardiovascular disease as an end-point. Recent studies showed a higher viral load in STEMI patients compared to controls [21] and a direct relationship between T cells activation and the number of CMV-DNA copies inside the atherosclerotic plaques in patients with peripheral artery disease [22]. A higher number of CMV-DNA copies were detected in patients with preexistent cardiovascular risk factors and were associated to acute coronary syndrome, suggesting that CMV reactivation may lead to the progression of atherosclerotic lesions, for example, transforming stable angina into unstable angina or myocardial infarction [23]. CMV-DNA was found in aortic plaques, but not in normal artery samples obtained from candidates for coronary artery by-pass graft [24]. Immunohistochemical studies demonstrated the presence of CMV pp65 antigen [25] in plaques obtained from patients with atherosclerosis undergoing vascular surgery [26].
Differences in the worldwide seroprevalence of CMV, as well as in the sample type (blood, atheroma plaques, and vascular wall fragments) and detection technique (serology, immunofluorescence, PCR alone or combined) can account for the sometimes controversial data. Overall, direct detection of viral antigens and/or DNA is a better proof of CMV association with atherosclerosis or cardiovascular disease, compared to serology alone.

2.1.2. HIV Associated CMV Infection and Atherosclerosis

Asymptomatic CMV replication can trigger immune system activation and perpetuate an inflammatory environment that favors the early development of atherosclerosis. This is more evident in immunosuppressed individuals, either transplant recipients or people living with HIV (PLWH). Previous studies in HIV-infected individuals have suggested a link between CMV reactivations and an increased intima-media thickness, demonstrated by higher levels of specific anti-CMV antibodies and CMV-specific T-cell responses [31,32]. Probably, the two pathogens act synergistically to promote the complex process of plaque formation [33]. Macrophages and monocytes involved in the initial “traditional” atherosclerotic lesions are reservoirs for both HIV and CMV and the activation of these cells, with foamy cells formation, represent the main mechanism in plaque initiation. Early stages of atherosclerosis have been detected by coronary computed tomography angiography (CCTA) in HIV individuals, who are at high risk of thrombosis and plaques rupture, leading to HIV-associated acute coronary syndrome [34]. Noncalcified, inflammatory plaques that can lead to myocardial infarction and stroke have been also detected in PLWH [35]. Cardiovascular disease remains an important cause of non-AIDS-related morbidity and mortality during HIV infection even under continuous cART treatment [36]. PLWH successfully treated with cART, with low or undetectable HIV replication, still express regulatory viral proteins (Tat and Nef) that may alter monocyte/macrophages cell function [35], possibly triggering subclinical episodes of CMV reactivation. These, in turn, will maintain or amplify the status of chronic inflammation, T cell activation, and immune dysregulation already present in HIV-infected patients, even in those under long-term suppressive cART or in elite controllers. Supporting this hypothesis, a small randomized placebo controlled trial on the effect of valgancyclovir, a potent antiviral drug, showed a significant decrease in both CMV DNA and the level of CD8 T cell activation in immunosupressed HIV-infected patients under supressive cART [37]. Moreover, chronic CMV infection is characterized by an unusual expansion of specific memory T cells and is a potent trigger for a particular phenotype of CD4 cells with cytotoxic activity that migrate toward the vascular endothelium, playing an important role in the initial vascular lesions and in the progression of atherosclerosis [38]. This process might be accelerated in HIV-infected patients who have a high level of circulating CD8 T cells with increased expression of CX3CR1, the receptor of vascular-endothelium homing chemokine that can be attracted to endothelial cells inducing persistent activation and dysfunction [39].

2.1.3. Cytomegalovirus and Plaque Formation in Pregnancy

Atherosclerosis may start during placentation, increasing the lifelong risk of atherosclerotic disease in women developing preeclampsia, a hypertensive pregnancy disorder [40,41,42]. CMV is the leading cause of neonatal congenital infections worldwide and is strongly associated with neurological sequelae in newborns and possibly associated with arterial hypertension, a mechanical blood flow condition that damages endothelial cells leading to preatherosclerotic lesions [43]. Based on the above-mentioned observations, we searched for studies that investigated the possible role of CMV in preeclampsia. Several studies looked at the impact of CMV infection on pregnancy complications associated with hypertension, but the results are quite controversial. Interestingly, some studies suggest that the medium-term risk of cardiovascular events in women that develop a hypertensive pregnancy disorder and their offspings is double compared to controls [40,44]. A significant association was found between CMV IgG sero-positivity and innate immune response in early-onset preeclampsia and the presence of HELLP (H: hemolysis, EL: elevated liver enzymes, LP: low platelet count) syndrome [45]. On the contrary, a small case-control study on hypertensive pregnancies, as well as a meta-analysis that included 2734 women with preeclampsia and 3424 healthy controls concluded that there is no significant relation between CMV infection (evaluated by both PCR and serology) and the onset of preeclampsia [46]. Nevertheless, it is interesting that acute atherosis occurs more often in women with preeclampsia [47] compared to normal pregnancies and other pregnancy complications like growth restriction, spontaneous preterm labor, or fetal death.
The hypothesis of a possible impact of CMV in vascular remodeling and acute atherosis lesions with maternal and fetal long-term impact on cardiovascular disease is very interesting and opens a new research direction on non-conventional risk factors for cardiovascular disease in young and very young patients.

3. CMV Mechanisms of Action in Atherosclerosis

Ever since it was used for the first time in 1755, the term atheroma describes a complex process involving both chronic inflammation and lipid accumulation, associated with vascular smooth muscle cells proliferation within the intimal layer of the blood vessel [48]. The potential mechanism by which CMV might promote atherogenesis remains poorly understood. However, CMV has a concerted impact on the main pathways involved in atherogenesis (Figure 1).

3.1. Oxidative Stress, Lipidogenesis and Endothelial Injury

Oxidized low-density lipoproteins (ox-LDL), which play a crucial role in the early events of atherogenesis, are preferentially recognized by scavenger receptors on macrophages and vascular cells. During CMV infection, the expression of type-B scavenger receptor on the surface of macrophages is upregulated [49]. Reactivation of the latent CMV infection in endothelial cells recruits macrophages and neutrophils by secreting chemoattractant and adhesion factors (MCP-1, VCAM-1, ICAM-1, and CXC), promoting internalization of oxLDL and foamy cells formation. CMV seems to upregulate several important pro-atherogenic molecules responsible for LDL and VLDL cellular uptake and cholesterol synthesis (NPC1L1, HMGCS1, HMGCR, LRP10, 11, 12, and SCARB) and downregulates anti-atherogenic proteins (ApoA1, ApoM, and ApoH) [50]. CMV-infected endothelial cells inhibit angiogenesis and promote abnormal vessel formation, a very important phenomenon already demonstrated in congenital CMV infection [51,52,53]. The expression of the receptor beta for PDGF (platelet-derived growth factor), an essential factor for the development and the plasticity of the vascular system, is upregulated by HCMV infection, stimulating atherogenesis [54]. CMV UL122 and US28-derived protein are homologous to an 11-aminoacid sequence of HSP60 that seems to produce endothelial cells apoptosis as an early event in atherogenesis [55]. The homology of pUS28 to HSP60 can also determine smooth muscle cells’ migration within vascular intima [55]. In vitro, CMV infects vascular smooth muscle cells, resulting in important alterations in the expression of lipid metabolism genes with intracellular cholesterol accumulation [56], possibly by downregulation of SSBP1, which is an important protein involved in wound repair [57].

3.2. Vascular Remodeling

A series of CMV-encoded proteins stimulate inflammation and vascular remodeling (Table 2).
Antibodies against CMV UL94, a region that encodes the viral tegument protein, are involved in the modulation of thousands of endothelial cells transcripts like adhesion molecules, growth factors, colony-stimulating factors, chemokines involved in leukocytes attraction, angiogenesis, and fibrosis [68]. CMV protein UL7 presents high structural and functional homology to CEACAM1, suggesting a direct involvement of CMV in vasculogenesis [58]. A stable domain in the structure of HCMV UL7 gene is responsible for STAT3/ERK1 MAP signal pathway activation. CMV can bind platelets using TLR2, generating the synthesis of proinflammatory CD40L, IL 1B, and proangiogenic VEGF, and further activating platelets, a cascade that can precipitate atherogenesis [69]. A recent in-vitro study published in 2020 suggests that chronic CMV infection downregulates endothelin 1 (ET-1) expression in endothelial and smooth muscle cells, thus providing additional evidence for the role of this pathogen in vascular impairment [70].
CMV itself can express viral cytokines and chemokines that might play a pivotal role in the angiogenesis and promotion of an inflammatory environment. An extensive homology between CMV and endothelial cell proteins were described and many immune cross-reactions caused by this molecular mimicry were incriminated in endothelial cells apoptosis, in the up-regulated expression of different cytokines and in the pathogeny of vascular damage in autoimmune diseases [68,71].

3.3. miRNA Regulation

During the last decade, small, highly-conserved, noncoding single-stranded RNA fragments called micro RNA (miR) were described as promoters and potential biomarkers of atherosclerotic lesions [72]. miRs were widely investigated and determined to be regulating different cell functions in atherosclerosis. They play a crucial role in all stages of atherosclerotic process, from the initial endothelial injury and lipid accumulation to angiogenesis, calcification, and thrombosis [72,73]. miR-21, for example, increases NOS phosphorylation and inhibits PPARa and Bcl2, leading to apoptosis and endothelial cells injury [73]. Elevated levels of miR-338-3p were identified in patients with atherosclerosis and were linked to ox-LD-induced apoptosis within endothelial cells [50]. Likewise, miR-126 and miR-127 are related to cell apoptosis by increasing vascular cell adhesion protein1 (VCAM1) and NAD-dependent deacetylase sirtuin-1 [73]. An impressive number of CMV-encoded miR were also identified [74] and it was postulated that they can also participate in vascular remodeling and endothelial injury [50]. A summary of some pro-atherosclerotic CMV-encoded miRs according to cell type function regulation is described in Table 3.

3.4. Animal Model Studies

Due to its species specificity, human CMV in-vivo studies are quite limited and animal models are frequently used to mimic genes/proteins and the secretome of human CMV. Of all animal models, rat and murine models have the highest homology to human CMV [81,82]. Rat and murine infection models produce acute as well as latent infection, with an important symptomatology in immunosuppressed animals. In murine models of atherosclerosis, CMV promotes a rapid course of transplant vascular sclerosis and induces upregulation of a series of genes involved in both angiogenesis and wound repair [83] and promotes an increased cytokine expression in the infected aortic samples [84]. CMV viral transcripts can modulate the host immune response and apoptosis [71], promoting endothelial damage and thus, atherosclerosis. For example, UL122 expressed on endothelial cells during early stages of infection show homology to connexin 45 and integrin alpha 3 and 6, while US28 also expressed on endothelial cells surface shows homology to integrin alpha 6 (CD49f) and seems to be involved in CMV reactivation from latency [68]. CMV IE2 gene delays smooth muscle cells apoptosis by upregulating the expression of antiapoptotic proteins Mcl-1 and Bcl-2 and was linked to a myocardin-induced transcriptional program responsible for aortic smooth muscle cells proliferation in rats’ aortas [85]. Furthermore, the role of CMV in cardiovascular disease seems to extend from a possible risk factor for atherosclerosis to direct myocardial pathology, as a recent animal model study demonstrates direct murine CMV infection of myocardial cells, with tachycardia and hypertrophy, suggesting CMV reactivation within the heart and vasculature [86]. The same murine model study showed viral DNA persistence within myocardial cells up to 100 days post-acute infection, but with a viral gpB gene expression of only 35 days post-infection, suggesting that CMV latency may start within the heart after acute infection [86]. Murine CMV has been associated with myocardial fibrosis, synus tachicardia and ventricular hypertrophy in chronically infected mice [87]. The results of these animal studies are additional proof for a possible role of CMV infection in atherosclerosis, cardiovascular disease, and direct myocardial injury.

4. Conclusions

According to the World Health Organization, cardiovascular disease is the leading cause of death worldwide. Despite all efforts in preventing the conventional risk factors, the number of deaths related to myocardial infarction, stroke, peripheral artery disease, or hypertensive pregnancy disorders is increasing. The involvement of infectious agents in the mechanisms of atherosclerosis has been studied for more than two decades. The association between CMV, atherosclerosis, and cardiovascular diseases is still controversial and needs to be clarified. Due to its ability to infect almost all cell types and its secretome largely based on proinflammatory cytokines, chemoattractant, and vascular growth factors, CMV triggers plaque formation by a complex mechanism of endothelial injury, alteration of lipid metabolism with lipid deposition, vascular smooth muscle cells proliferation and migration, as well as disruption of coagulation mechanism and thrombosis.
This review highlights that CMV infection may be associated with acute atherosis, atherosclerosis, and/or cardiovascular disease in almost all population groups. It may produce vascular injuries eversince placentation and then continue to maintain a chronic inflammation status with subsequent vascular impairment and cardiovascular events later in life, in both healthy and immunosuppressed patients. Direct detection of CMV’s DNA or antigens in atherosclerotic patients is the best proof for a possible role of this ubiquitous virus in plaque development. In order to answer if there is a role as an independet risk factor for CMV in atherosclerosis and cardiovascular disease, complex multicentric cohort studies enrolling newborns, children, adolescents, young adults, adults, and elderly should be considered. This would allow a better understanding of the role played by chronic CMV infection and reactivation in atherogenesis and cardiac disease, in a very exciting era, in which new therapeutic and prophylactic strategies including vaccination are already in progress for this infectious agent.

Author Contributions

Conceptualization, C.V.C. and S.M.R.; investigation, C.V.C.; writing—original draft preparation, C.V.C. and S.M.R.; writing—review and editing, C.V.C., S.M.R. and S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was partially supported by Romanian Ministry of Research, Innovation and Digitalisation, grant number PFE_33/2021.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fowkes, F.G.R.; Rudan, D.; Rudan, I.; Aboyans, V.; Denenberg, J.O.; McDermott, M.M.; Norman, P.E.; Sampson, U.K.A.; Williams, L.J.; Mensah, G.A.; et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: A systematic review and analysis. Lancet 2013, 382, 1329–1340. [Google Scholar] [CrossRef]
  2. Blum, A.; Blum, N. Coronary artery disease: Are men and women created equal? Gend. Med. 2009, 6, 410–418. [Google Scholar] [CrossRef]
  3. William, H.; Ben, L.; Paul, S.; Jane, A.; Sarah, L. Epidemiology of Atherosclerosis and the Potential to Reduce the Global Burden of Atherothrombotic Disease. Circ. Res. 2016, 118, 535–546. [Google Scholar]
  4. Abrignani, M.G.; Lucà, F.; Favilli, S.; Benvenuto, M.; Rao, C.M.; Di Fusco, S.A.; Gabrielli, D.; Gulizia, M.M. Lifestyles and Cardiovascular Prevention in Childhood and Adolescence. Pediatr. Cardiol. 2019, 40, 1113–1125. [Google Scholar] [CrossRef] [PubMed]
  5. Fatkhullina, A.R.; Peshkova, I.O.; Koltsova, E.K. The Role of Cytokines in the Development of Atherosclerosis. Biochememistry 2016, 81, 1358–1370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Campbell, L.A.; Rosenfeld, M.E. Infection and Atherosclerosis Development. Arch. Med. Res. 2015, 46, 339–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Szwed, P.; Gąsecka, A.; Zawadka, M.; Eyileten, C.; Postuła, M.; Mazurek, T.; Szarpak, Ł.; Filipiak, K. Infections as Novel Risk Factors of Atherosclerotic Cardiovascular Diseases: Pathophysiological Links and Therapeutic Implications. J. Clin. Med. 2021, 10, 2539. [Google Scholar] [CrossRef] [PubMed]
  8. Clifford, A.; Hoffman, G.S. Evidence for a vascular microbiome and its role in vessel health and disease. Curr. Opin. Rheumatol. 2015, 27, 397–405. [Google Scholar] [CrossRef] [PubMed]
  9. Zuhair, M.; Smit, G.S.A.; Wallis, G.; Jabbar, F.; Smith, C.; Devleesschauwer, B.; Griffiths, P. Estimation of the worldwide seroprevalence of cytomegalovirus: A systematic review and meta-analysis. Rev. Med. Virol. 2019, 29, e2034. [Google Scholar] [CrossRef] [Green Version]
  10. Du, Y.; Zhang, G.; Liu, Z. Human cytomegalovirus infection and coronary heart disease: A systematic review. Virol. J. 2018, 15, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Britt, W. Manifestations of human cytomegalovirus infection: Proposed mechanisms of acute and chronic disease. Curr. Top. Microbiol. Immunol. 2008, 325, 417–470. [Google Scholar]
  12. Capron, L.; Wyplosz, B. The infection theory in atherosclerosis. Arch. Mal. Coeur Vaiss. 1998, 91, 21–26. [Google Scholar]
  13. Chen, R.; Xiong, S.; Yang, Y.; Fu, W.; Wang, Y.; Ge, J. The relationship between human cytomegalovirus infection and atherosclerosis development. Mol. Cell Biochem. 2003, 249, 91–96. [Google Scholar] [CrossRef]
  14. Adam, E.; Melnick, J.L.; Probtsfield, J.L.; Petrie, B.L.; Burek, J.; Bailey, K.R.; Debakey, M.E. High levels of cytomegalovirus antibody in patients requiring vascular surgery for atherosclerosis. Lancet 1987, 2, 291–293. [Google Scholar] [CrossRef]
  15. Melnick, J.L.; Petrie, B.L.; Dreesman, G.R.; Burek, J.; McCollum, C.H.; DeBakey, M.E. Cytomegalovirus antigen within human arterial smooth muscle cells. Lancet 1983, 2, 644–647. [Google Scholar] [CrossRef]
  16. Zhang, J.; Liu, Y.Y.; Sun, H.L.; Li, S.; Xiong, H.R.; Yang, Z.Q.; Jiang, X.J. High Human Cytomegalovirus IgG Level is Associated with Increased Incidence of Diabetic Atherosclerosis in Type 2 Diabetes Mellitus Patients. Med. Sci. Monit. 2015, 21, 4102–4110. [Google Scholar] [CrossRef] [Green Version]
  17. Poznyak, A.; Grechko, A.V.; Poggio, P.; Myasoedova, V.A.; Alfieri, V.; Orekhov, A.N. The Diabetes Mellitus—Atherosclerosis Connection: The Role of Lipid and Glucose Metabolism and Chronic Inflammation. Int. J. Mol. Sci. 2020, 21, 1835. [Google Scholar] [CrossRef] [Green Version]
  18. Turk Wensveen, T.; Gašparini, D.; Rahelić, D.; Wensveen, F.M. Type 2 diabetes and viral infection; cause and effect of disease. Diabetes Res. Clin. Pract. 2021, 172, 108637. [Google Scholar] [CrossRef]
  19. Chen, S.; de Craen, A.J.M.; Raz, Y.; Derhovanessian, E.; Vossen, A.C.T.M.; Westendorp, R.G.J.; Pawelec, G.; Maier, A.B. Cytomegalovirus seropositivity is associated with glucose regulation in the oldest old. Results from the Leiden 85-plus Study. Immun. Ageing. 2012, 9, 18. [Google Scholar] [CrossRef] [Green Version]
  20. Yoo, S.G.; Han, K.D.; Lee, K.H.; La, Y.; Kwon, D.E.; Han, S.H. Impact of Cytomegalovirus Disease on New-Onset Type 2 Diabetes Mellitus: Population-Based Matched Case-Control Cohort Study. Diabetes Metab. J. 2019, 43, 815–829. [Google Scholar] [CrossRef]
  21. Lebedeva, A.; Maryukhnich, E.; Grivel, J.-C.; Vasilieva, E.; Margolis, L.; Shpektor, A. Productive Cytomegalovirus Infection Is Associated with Impaired Endothelial Function in ST-Elevation Myocardial Infarction. Am. J. Med. 2019, 133, 133–142. [Google Scholar] [CrossRef] [Green Version]
  22. Nikitskaya, E.; Lebedeva, A.; Ivanova, O.; Maryukhnich, E.; Shpektor, A.; Grivel, J.; Margolis, L.; Vasilieva, E. Cytomegalovirus‐Productive Infection Is Associated with Acute Coronary Syndrome. J. Am. Hear. Assoc. 2016, 5, e003759. [Google Scholar] [CrossRef] [Green Version]
  23. Nikitskaya, E.A.; Grivel, J.C.; Maryukhnich, E.V.; Lebedeva, A.M.; Ivanova, O.I.; Savvinova, P.P.; Shpektor, A.V.; Margolis, L.B.; Vasilieva, E.Y. Cytomegalovirus in Plasma of Acute Coronary Syndrome Patients. Acta Nat. 2016, 8, 102–107. [Google Scholar] [CrossRef]
  24. Heybar, H.; Alavi, S.M.; Nejad, M.F.; Latifi, M. Cytomegalovirus Infection and Atherosclerosis in Candidate of Coronary Artery Bypass Graft. Jundishapur J. Microbiol. 2015, 8, e15476. [Google Scholar] [CrossRef] [Green Version]
  25. Wang, Z.; Cai, J.; Zhang, M.; Wang, X.; Chi, H.; Feng, H.; Yang, X. Positive Expression of Human Cytomegalovirus Phosphoprotein 65 in Atherosclerosis. BioMed Res. Int. 2016, 2016, 4067685. [Google Scholar] [CrossRef]
  26. Ibrahim, A.I.; Obeid, M.T.; Jouma, M.J.; Moasis, G.A.; Al-Richane, W.L.; Kindermann, I.; Boehm, M.; Roemer, K.; Mueller-Lantzsch, N.; Gärtner, B.C. Detection of herpes simplex virus, cytomegalovirus and Epstein-Barr virus DNA in atherosclerotic plaques and in unaffected bypass grafts. J. Clin. Virol. 2005, 32, 29–32. [Google Scholar] [CrossRef]
  27. Jia, Y.-J.; Liu, J.; Han, F.-F.; Wan, Z.-R.; Gong, L.-L.; Liu, H.; Zhang, W.; Wardell, T.; Lv, Y.-L.; Liu, L.-H. Cytomegalovirus infection and atherosclerosis risk: A meta-analysis. J. Med. Virol. 2017, 89, 2196–2206. [Google Scholar] [CrossRef]
  28. Courivaud, C.; Bamoulid, J.; Chalopin, J.-M.; Gaiffe, E.; Tiberghien, P.; Saas, P.; Ducloux, D. Cytomegalovirus Exposure and Cardiovascular Disease in Kidney Transplant Recipients. J. Infect. Dis. 2013, 207, 1569–1575. [Google Scholar] [CrossRef] [Green Version]
  29. Betjes, M.G.H.; Litjens, N.H.R.; Zietse, R. Seropositivity for cytomegalovirus in patients with end-stage renal disease is strongly associated with atherosclerotic disease. Nephrol. Dial. Transplant. 2007, 22, 3298–3303. [Google Scholar] [CrossRef] [Green Version]
  30. Hamilton, E.M.; E Allen, N.; Mentzer, A.J.; Littlejohns, T.J. Human Cytomegalovirus and Risk of Incident Cardiovascular Disease in United Kingdom Biobank. J. Infect. Dis. 2021, 225, 1179–1188. [Google Scholar] [CrossRef]
  31. Knudsen, A.; Kristoffersen, U.S.; Panum, I.; Hansen, Y.B.; Skottrup, P.D.; Hasbak, P.; Kjaer, A.; Lebech, A.-M. Coronary artery calcium and intima-media thickness are associated with level of cytomegalovirus immunoglobulin G in HIV-infected patients. HIV Med. 2019, 20, 60–62. [Google Scholar] [CrossRef] [PubMed]
  32. Hsue, P.Y.; Hunt, P.W.; Sinclair, E.; Bredt, B.; Franklin, A.; Killian, M.; Hoh, R.; Martin, J.N.; McCune, J.M.; Waters, D.D.; et al. Increased carotid intima-media thickness in HIV patients is associated with increased cytomegalovirus-specific T-cell responses. AIDS 2006, 20, 2275–2283. [Google Scholar] [CrossRef] [PubMed]
  33. Bourgi, K.; Wanjalla, C.; Koethe, J.R. Inflammation and Metabolic Complications in HIV. Curr. HIV AIDS Rep. 2018, 15, 371–381. [Google Scholar] [CrossRef]
  34. Augustemak de Lima, L.R.; Petroski, E.L.; Moreno, Y.M.F.; Silva, D.A.S.; de Trindade, E.B.M.S.; de Carvalho, A.P.; de Carlos, I. BackDyslipidemia, chronic inflammation, and subclinical atherosclerosis in children and adolescents infected with HIV: The PositHIVe Health Study. PLoS ONE 2018, 13, e0190785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kearns, A.; Gordon, J.; Burdo, T.H.; Qin, X. HIV-1-Associated Atherosclerosis: Unraveling the Missing Link. J. Am. Coll. Cardiol. 2017, 69, 3084–3098. [Google Scholar] [CrossRef] [PubMed]
  36. Strategies for Management of Antiretroviral Therapy (SMART) Study Group; El-Sadr, W.M.; Lundgren, J.; Neaton, J.D.; Gordin, F.; Abrams, D.; Arduino, R.C.; Babiker, A.; Burman, W.; Clumeck, N.; et al. CD4+ count-guided interruption of antiretroviral treatment. N. Engl. J. Med 2006, 355, 2283–2296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Hunt, P.W.; Martin, J.N.; Sinclair, E.; Epling, L.; Teague, J.; Jacobson, M.A.; Tracy, R.P.; Corey, L.; Deeks, S.G. Valganciclovir Reduces T Cell Activation in HIV-infected Individuals with Incomplete CD4+ T Cell Recovery on Antiretroviral Therapy. J. Infect. Dis. 2011, 203, 1474–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Ducloux, D.; Bamoulid, J.; Crepin, T.; Rebibou, J.M.; Courivaud, C.; Saas, P. Posttransplant Immune Activation: Innocent Bystander or Insidious Culprit of Posttransplant Accelerated Atherosclerosis. Cell Transplant. 2017, 26, 1601–1609. [Google Scholar] [CrossRef] [Green Version]
  39. Panigrahi, S.; Chen, B.; Fang, M.; Potashnikova, D.; Komissarov, A.A.; Lebedeva, A.; Michaelson, G.M.; Wyrick, J.M.; Morris, S.R.; Sieg, S.F.; et al. CX3CL1 and IL-15 Promote CD8 T cell chemoattraction in HIV and in atherosclerosis. PLoS Pathog. 2020, 16, e1008885. [Google Scholar] [CrossRef]
  40. Amaral, L.M.; Cunningham, M.W.; Cornelius, D.C.; LaMarca, B. Preeclampsia: Long-term consequences for vascular health. Vasc. Health Risk Manag. 2015, 11, 403–415. [Google Scholar]
  41. Brosens, I.; Benagiano, M.; Puttemans, P.; D’Elios, M.M.; Benagiano, G. The placental bed vascular pathology revisited: A risk indicator for cardiovascular disease. J. Matern. Fetal Neonatal Med. 2019, 32, 1556–1564. [Google Scholar] [CrossRef]
  42. Staff, A.C.; Johnsen, G.M.; Dechend, R.; Redman, C.W.G. Preeclampsia and uteroplacental acute atherosis: Immune and inflammatory factors. J. Reprod. Immunol. 2014, 101–102, 120–126. [Google Scholar] [CrossRef]
  43. Cheng, J.; Ke, Q.; Jin, Z.; Wang, H.; Kocher, O.; Morgan, J.P.; Zhang, J.; Crumpacker, C.S. Cytomegalovirus infection causes an increase of arterial blood pressure. PLoS Pathog. 2009, 5, e1000427. [Google Scholar] [CrossRef] [Green Version]
  44. Ahmed, R.; Dunford, J.; Mehran, R.; Robson, S.; Kunadian, V. Pre-eclampsia and future cardiovascular risk among women: A review. J. Am. Coll. Cardiol. 2014, 63, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
  45. Xie, F.; Hu, Y.; Magee, L.A.; Money, D.M.; Patrick, D.M.; Krajden, M.; Thomas, E.; von Dadelszen, P. An association between cytomegalovirus infection and pre-eclampsia: A case-control study and data synthesis. Acta Obstet. Gynecol. Scand. 2010, 89, 1162–1167. [Google Scholar] [CrossRef]
  46. Geraili, Z.; Riahi, S.M.; Khani, S.; Rostami, A.; Bayani, M.; Hajian-Tilaki, K.; Shiadeh, M.N. Cytomegalovirus infection and risk of preeclampsia: A meta-analysis of observational studies. Casp. J. Intern. Med. 2018, 9, 211–219. [Google Scholar]
  47. Kim, Y.M.; Chaemsaithong, P.; Romero, R.; Shaman, M.; Kim, C.J.; Kim, J.S.; Qureshi, F.; Jacques, S.M.; Ahmed, A.I.; Chaiworapongsa, T.; et al. The frequency of acute atherosis in normal pregnancy and preterm labor, preeclampsia, small-for-gestational age, fetal death and midtrimester spontaneous abortion. J. Matern. Fetal Neonatal Med. 2015, 28, 2001–2009. [Google Scholar] [CrossRef] [Green Version]
  48. Tedgui, A.; Mallat, Z. Cytokines in atherosclerosis: Pathogenic and regulatory pathways. Physiol. Rev. 2006, 86, 515–581. [Google Scholar] [CrossRef] [Green Version]
  49. Carlquist, J.F.; Muhlestein, J.B.; Horne, B.D.; Hart, N.I.; Lim, T.; Habashi, J.; Anderson, J.G. Cytomegalovirus stimulated mRNA accumulation and cell surface expression of the oxidized LDL scavenger receptor, CD36. Atherosclerosis 2004, 177, 53–59. [Google Scholar] [CrossRef]
  50. Zhu, W.; Liu, S. The role of human cytomegalovirus in atherosclerosis: A systematic review. Acta Biochim. Biophys. Sin. 2020, 52, 339–353. [Google Scholar] [CrossRef]
  51. Jeffery, H.C.; Söderberg-Naucler, C.; Butler, L.M. Human cytomegalovirus induces a biphasic inflammatory response in primary endothelial cells. J. Virol. 2013, 87, 6530–6535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Grundy, J.E.; Lawson, K.M.; MacCormac, L.P.; Fletcher, J.M.; Yong, K.L. Cytomegalovirus-infected endothelial cells recruit neutrophils by the secretion of C-X-C chemokines and transmit virus by direct neutrophil-endothelial cell contact and during neutrophil transendothelial migration. J. Infect. Dis. 1998, 177, 1465–1474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. DuRose, J.B.; Li, J.; Chien, S.; Spector, D.H. Infection of vascular endothelial cells with human cytomegalovirus under fluid shear stress reveals preferential entry and spread of virus in flow conditions simulating atheroprone regions of the artery. J. Virol. 2012, 86, 13745–13755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Reinhardt, B.; Mertens, T.; Mayr-Beyrle, U.; Frank, H.; Lüske, A.; Schierling, K.; Waltenberger, J. HCMV infection of human vascular smooth muscle cells leads to enhanced expression of functionally intact PDGF beta-receptor. Cardiovasc. Res. 2005, 67, 151–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Bason, C.; Corrocher, R.; Lunardi, C.; Puccetti, P.; Olivieri, O.; Girelli, D.; Navone, R.; Beri, R.; Millo, E.; Margonato, A.; et al. Interaction of antibodies against cytomegalovirus with heat-shock protein 60 in pathogenesis of atherosclerosis. Lancet 2003, 362, 1971–1977. [Google Scholar] [CrossRef]
  56. Li, L.; Li, Y.; Dai, Z.; Liu, M.; Wang, B.; Liu, S.; Wang, L.; Chen, L.; Tan, Y.; Wu, G. Lipid Metabolism in Vascular Smooth Muscle Cells Infuenced by HCMV Infection. Cell. Physiol. Biochem. 2016, 39, 1804–1812. [Google Scholar] [CrossRef] [Green Version]
  57. Guo, N.; Zhang, N.; Yan, L.; Cao, X.; Lv, F.; Wang, J.; Wang, Y.; Cong, H. Down-regulation of single-stranded DNA-binding protein 1 expression induced by HCMV infection promotes lipid accumulation in cells. Braz. J. Med. Biol. Res. 2017, 50, e6389. [Google Scholar] [CrossRef] [Green Version]
  58. MacManiman, J.D.; Meuser, A.; Botto, S.; Smith, P.P.; Liu, F.; Jarvis, M.A.; Nelson, J.A.; Caposio, P. Human cytomegalovirus-encoded pUL7 is a novel CEACAM1-like molecule responsible for promotion of angiogenesis. mBio 2014, 5, e02035. [Google Scholar] [CrossRef] [Green Version]
  59. Dumortier, J.; Streblow, D.N.; Moses, A.V.; Jacobs, J.M.; Kreklywich, C.N.; Camp, D.; Smith, R.D.; Orloff, S.L.; Nelson, J.A. Human Cytomegalovirus Secretome Contains Factors That Induce Angiogenesis and Wound Healing. J. Virol. 2008, 82, 6524–6535. [Google Scholar] [CrossRef] [Green Version]
  60. Costa, H.; Nascimento, R.; Sinclair, J.; Parkhouse, R.M.E. Human cytomegalovirus gene UL76 induces IL-8 expression through activation of the DNA damage response. PLoS Pathog. 2013, 9, e1003609. [Google Scholar] [CrossRef] [Green Version]
  61. Wang, S.K.; Duh, C.Y.; Wu, C.W. Human Cytomegalovirus UL76 Encodes a Novel Virion-Associated Protein That Is Able To Inhibit Viral Replication. J. Virol. 2004, 78, 9750–9762. [Google Scholar] [CrossRef] [Green Version]
  62. Tu, C.C.; Arnolds, K.L.; O’Connor, C.M.; Spencer, J.V. Human Cytomegalovirus UL111A and US27 Gene Products Enhance the CXCL12/CXCR4 Signaling Axis via Distinct Mechanisms. J. Virol. 2018, 92, e01981-17. [Google Scholar] [CrossRef] [Green Version]
  63. Harris, S.M.; Bullock, B.; Westgard, E.; Zhu, H.; Stenberg, R.M.; Kerry, J.A. Functional Properties of the Human Cytomegalovirus IE86 Protein Required for Transcriptional Regulation and Virus Replication. J. Virol. 2010, 84, 8839–8848. [Google Scholar] [CrossRef] [Green Version]
  64. Taylor, R.T.; Bresnahan, W.A. Human cytomegalovirus immediate-early 2 protein IE86 blocks virus-induced chemokine expression. J. Virol. 2006, 80, 920–928. [Google Scholar] [CrossRef] [Green Version]
  65. Casarosa, P.; Bakker, R.A.; Verzijl, D.; Navis, M.; Timmerman, H.; Leurs, R.; Smit, M.J. Constitutive signaling of the human cytomegalovirus-encoded chemokine receptor US28. J. Biol. Chem. 2001, 276, 1133–1137. [Google Scholar] [CrossRef] [Green Version]
  66. Straschewski, S.; Patrone, M.; Walther, P.; Gallina, A.; Mertens, T.; Frascaroli, G. Protein pUL128 of Human Cytomegalovirus Is Necessary for Monocyte Infection and Blocking of Migration. J. Virol. 2011, 85, 5150–5158. [Google Scholar] [CrossRef] [Green Version]
  67. Zheng, Q.; Tao, R.; Gao, H.; Xu, J.; Shang, S.; Zhao, N. HCMV-encoded UL128 enhances TNF-α and IL-6 expression and promotes PBMC proliferation through the MAPK/ERK pathway in vitro. Viral Immunol. 2012, 25, 98–105. [Google Scholar] [CrossRef] [Green Version]
  68. Dolcino, M.; Puccetti, A.; Barbieri, A.; Bason, C.; Tinazzi, E.; Ottria, A.; Patuzzo, G.; Martinelli, N.; Lunardi, C. Infections and autoimmunity: Role of human cytomegalovirus in autoimmune endothelial cell damage. Lupus 2015, 24, 419–432. [Google Scholar] [CrossRef]
  69. Assinger, A.; Kral, J.B.; Yaiw, K.C.; Schrottmaier, W.C.; Kurzejamska, E.; Wang, Y.; Mohammad, A.-A.; Religa, P.; Rahbar, A.; Schabbauer, G.; et al. Human cytomegalovirus-platelet interaction triggers toll-like receptor 2-dependent proinflammatory and proangiogenic responses. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 801–809. [Google Scholar] [CrossRef] [Green Version]
  70. Yaiw, K.C.; Mohammad, A.A.; Taher, C.; Cui, H.L.; Costa, H.; Kostopoulou, O.N.; Jung, M.; Assinger, A.; Wilhelmi, V.; Yang, J.; et al. Human Cytomegalovirus Reduces Endothelin-1 Expression in Both Endothelial and Vascular Smooth Muscle Cells. Microorganisms 2021, 9, 1137. [Google Scholar] [CrossRef]
  71. Van Damme, E.; van Loock, M. Functional Annotation of Human Cytomegalovirus Gene Products: An Update. Front. Microbiol. 2014, 5, 218. [Google Scholar] [CrossRef] [PubMed]
  72. Churov, A.; Summerhill, V.; Grechko, A.; Orekhova, V.; Orekhov, A. MicroRNAs as Potential Biomarkers in Atherosclerosis. Int. J. Mol. Sci. 2019, 20, 5547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hosen, M.R.; Goody, P.R.; Zietzer, A.; Nickenig, G.; Jansen, F. MicroRNAs as Master Regulators of Atherosclerosis: From Pathogenesis to Novel Therapeutic Options. Antioxid. Redox Signal. 2020, 33, 621–644. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, L.; Yu, J.; Liu, Z. MicroRNAs expressed by human cytomegalovirus. Virol. J. 2020, 17, 34. [Google Scholar] [CrossRef]
  75. Zhang, S.; Liu, L.; Wang, R.; Tuo, H.; Guo, Y.; Yi, L.; Wang, D.; Wang, J. MicroRNA-217 Promotes Angiogenesis of Human Cytomegalovirus-Infected Endothelial Cells through Downregulation of SIRT1 and FOXO3A. PLoS ONE 2013, 8, e83620. [Google Scholar] [CrossRef] [Green Version]
  76. Fan, J.; Zhang, W.; Liu, Q. Human cytomegalovirus-encoded miR-US25-1 aggravates the oxidised low density lipoprotein-induced apoptosis of endothelial cells. Biomed. Res. Int. 2014, 2014, 531979. [Google Scholar] [CrossRef] [Green Version]
  77. Shen, K.; Xu, L.; Chen, D.; Tang, W.; Huang, Y. Human cytomegalovirus-encoded miR-UL112 contributes to HCMV-mediated vascular diseases by inducing vascular endothelial cell dysfunction. Virus Genes 2018, 54, 172–181. [Google Scholar] [CrossRef]
  78. Landais, I.; Pelton, C.; Streblow, D.; DeFilippis, V.; McWeeney, S.; Nelson, J.A. Human Cytomegalovirus miR-UL112-3p Targets TLR2 and Modulates the TLR2/IRAK1/NFκB Signaling Pathway. PLoS Pathog. 2015, 11, e1004881. [Google Scholar] [CrossRef]
  79. Shao, Y.; Qi, Y.; Huang, Y.; Liu, Z.; Ma, Y.; Guo, X.; Jiang, S.; Sun, Z.; Ruan, Q. Human cytomegalovirus miR-US4-5p promotes apoptosis via downregulation of p21-activated kinase 2 in cultured cells. Mol. Med. Rep. 2017, 16, 4171–4178. [Google Scholar] [CrossRef] [Green Version]
  80. Dong, J.; Li, S.; Lu, Z.; Du, P.; Liu, G.; Li, M.; Ma, C.; Zhou, J.; Bao, J. HCMV-miR-US33-5p promotes apoptosis of aortic vascular smooth muscle cells by targeting EPAS1/SLC3A2 pathway. Cell. Mol. Biol. Lett. 2022, 27, 40. [Google Scholar] [CrossRef]
  81. Rawlinson, W.D.; Farrell, H.E.; Barrell, B.G. Analysis of the complete DNA sequence of murine cytomegalovirus. J. Virol. 1996, 70, 8833–8849. [Google Scholar] [CrossRef] [Green Version]
  82. Dogra, P.; Sparer, T.E. What we have learned from animal models of HCMV. Methods Mol. Biol. 2014, 1119, 267–288. [Google Scholar]
  83. Streblow, D.N.; Kreklywich, C.N.; Andoh, T.; Moses, A.V.; Dumortier, J.; Smith, P.P.; DeFilippis, V.; Fruh, K.; Nelson, J.A.; Orloff, S.L. The Role of Angiogenic and Wound Repair Factors During CMV-Accelerated Transplant Vascular Sclerosis in Rat Cardiac Transplants. Am. J. Transplant. 2008, 8, 277–287. [Google Scholar] [CrossRef]
  84. Vliegen, I.; Duijvestijn, A.; Grauls, G.; Herngreen, S.; Bruggeman, C.; Stassen, F. Cytomegalovirus infection aggravates atherogenesis in apoE knockout mice by both local and systemic immune activation. Microbes Infect. 2004, 6, 17–24. [Google Scholar] [CrossRef] [PubMed]
  85. Akiyama, H.; Gummuluru, S. HIV-1 Persistence and Chronic Induction of Innate Immune Responses in Macrophages. Viruses 2020, 12, 711. [Google Scholar] [CrossRef]
  86. Bonavita, C.M.; Cardin, R.D. Don’t Go Breaking My Heart: MCMV as a Model for HCMV-Associated Cardiovascular Diseases. Pathogens 2021, 10, 619. [Google Scholar] [CrossRef]
  87. Bonavita, C.M.; White, T.M.; Francis, J.; Cardin, R.D. Heart Dysfunction Following Long-Term Murine Cytomegalovirus Infection: Fibrosis, Hypertrophy, and Tachycardia. Viral Immunol. 2020, 33, 237–245. [Google Scholar] [CrossRef] [Green Version]
Figure 1. CMV mechanism in atherosclerosis (Created in BioRender.com).
Figure 1. CMV mechanism in atherosclerosis (Created in BioRender.com).
Jcm 11 03832 g001
Table 1. Associations between CMV, atherosclerosis, and cardiovascular disease.
Table 1. Associations between CMV, atherosclerosis, and cardiovascular disease.
AuthorsReferencePopulationCMV DeterminantOutcomeAssociationStudy Type
Adam et al.[14]157 caucasian male undergoing vascular surgery for atherosclerosisSerologyPADYESCase-control
Melnick et al.[15]132 arterial tissue samples from atheroslerotic plaques of caucasian patients undergoing vascular surgery for atherosclerosisIF pp65PADYESProspective cohort
Jia et al.[27]3328 multiethnic patients but predominantly asians with CAD and PAD with or without surgical indicationSerology
CMV DNA
IF pp65
CAD and PADYESMeta-analysis (control-case or nested control-case)
Nikitskaya et al.[22]71 ACS, 26 SCAD, 22 atheroslerotic plaques (PAD)CMV DNACAD and PADYESCase-control
Zhang et al.[16]222 hospitalized patient with type 2 diabetes mellitusSerology
CMV DNA
T2DM and ultrasound confirmed atherosclerosisYES for latent infection onlyProspective cohort
Courivaud et al.[28]570 renal transplant recipientsSerology
CMV DNA
CAD, PAD, strokeYESProspective cohort
Betjes et al.[29]408 ESRD patientsSerologyCAD and PADYESRetrospective cohort
Lebedeva et al.[21]33 STEMI patientsCMV DNAIHD (STEMI)YESCase-control
Nikitskaya et al.[23]97 CAD patiensCMV DNACADYESCase-control
Heybar et al.[24]55 CABG—normal and atherosclerotic samplesCMV DNACAD—CABGYESCase-control
Wang et al.[25]15 paraffin-embedded peripheral artery specimens from patients with ATSIHCPADYESProspective cohort
Ibrahim et al.[26]48 biopsies from atherosclerotic plaquesCMV DNAPADYESCase-control
Hamilton et al.[30]8531 white ethnic background with no prevalent CVDMultiplex serology panelCVD, IHD, strokeNOProspective cohort
CAD—coronary artery disease, CABG—coronary artery by-pass graft, CVD—cardiovascular disease, ESRD—end-stage renal disease, IHD—ischemic heart disease, IHC—immunohistochemistry, PAD—peripheral artery disease, STEMI—ST-elevated myocardial infarction.
Table 2. CMV encoded proteins and the expression of humoral factors involved in atherosclerosis.
Table 2. CMV encoded proteins and the expression of humoral factors involved in atherosclerosis.
HCMV ProteinFunctionMechanism in Atheromatous Process
UL 7Early-late gene that is cleaved to release glycosilated ectodomain [58]Stimulates inflammation in endothelial cells by IL-6 expression and STAT and MAPK pathway activation [58]
Increased similarity to the first Ig-like domain of the CEACAM protein family playing a key role in vasculogenesis [58,59]
UL 76Unclear—highly conserved protein that may cleave nuclear proteins and modulate viral activation or repression [60,61]Induces IL-8 expression by DNA damage [60]
UL 111AEncodes human homologous IL-10 cmvIL-10 [62]Activates CXCL12/CXCR4 and STAT3 signal pathways in monocytes, epithelial cells and fibroblasts [62]
UL 122IE2 regulatory protein- modulates viral activation and reactivation from latency [55]
A protein isoform IE86 encoded by UL122—DNA binding protein that regulates viral gene expression and recruits chromatin modeling enzymes.
Essential for viral replication by transactivation of vital viral early promoters probably by direct DNA binding [63]
Homology to heat shock protein 60 (HSP60) and increased monocytes adhesion [55]
Supresses the expression of proinflammatory citokines IFN β, RANTES, MIG, MCP2, IE1, GAPDH [64] and may play a protective role in atherosclerosis development
US 28Expressed on cell surface with high homology to CC chemokine receptor CCR1 facilitating cell-cell fusion [65]Homology to heat shock protein 60 (HSP60), determines smooth muscle cells migration [55]
UL 128Envelope protein with immunoregulatory properties, responsible for monocytes and epithelial cells infection [66], thus virus replication and disseminationRecruits monocytes/macrophages cells by chemoattraction and determines high expression of TNF-α and IL-6 [67]
Table 3. CMV encoded miR according to cell type function in atherogenesis.
Table 3. CMV encoded miR according to cell type function in atherogenesis.
CMV Encoded miRCell TypeFunctionReferences
miR-217EndothelialAngiogenesis[50,75]
miR-US25-1EndothelialOx-LDL induced apoptosis[76]
miR-UL112EndothelialEndothelial dysfunction by modulation of multiple signal pathways[77,78]
miR-US4-5pMacrophagesApoptosis[50,79]
miR-US33Vascular smooth muscle cellsApoptosis[80]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cristescu, C.V.; Alain, S.; Ruță, S.M. The Role of CMV Infection in Primary Lesions, Development and Clinical Expression of Atherosclerosis. J. Clin. Med. 2022, 11, 3832. https://doi.org/10.3390/jcm11133832

AMA Style

Cristescu CV, Alain S, Ruță SM. The Role of CMV Infection in Primary Lesions, Development and Clinical Expression of Atherosclerosis. Journal of Clinical Medicine. 2022; 11(13):3832. https://doi.org/10.3390/jcm11133832

Chicago/Turabian Style

Cristescu, Carmen Valentina, Sophie Alain, and Simona Maria Ruță. 2022. "The Role of CMV Infection in Primary Lesions, Development and Clinical Expression of Atherosclerosis" Journal of Clinical Medicine 11, no. 13: 3832. https://doi.org/10.3390/jcm11133832

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

Article Metrics

Back to TopTop