ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Overview of possible risk factors for cardiovascular disease

Overview of possible risk factors for cardiovascular disease
Literature review current through: Jan 2024.
This topic last updated: Jul 21, 2023.

INTRODUCTION — An overview of the possible emerging cardiovascular risk disease (CVD) risk factors is presented here. An overview of the established risk factors for CVD, data supporting the importance of the individual risk factors (eg, hyperlipidemia, hypertension, smoking), coronary risk factors of particular importance in women and in young patients, and estimation of coronary risk in an individual patient are discussed elsewhere.

(See "Overview of established risk factors for cardiovascular disease".)

(See "Low-density lipoprotein cholesterol-lowering therapy in the primary prevention of cardiovascular disease".)

(See "Overview of hypertension in adults", section on 'Treatment'.)

(See "Overview of atherosclerotic cardiovascular risk factors in females".)

(See "Coronary artery disease and myocardial infarction in young people".)

DEFINITIONS AND BACKGROUND — CVD is common in the general population, affecting the majority of adults past the age of 60 years. As a diagnostic category, CVD includes four major areas:

Coronary heart disease (CHD), manifested by myocardial infarction (MI), angina pectoris, heart failure, and coronary death

Cerebrovascular disease, manifested by stroke and transient ischemic attack

Peripheral artery disease, manifested by intermittent claudication

Aortic atherosclerosis and thoracic or abdominal aortic aneurysm

CHD accounts for approximately one-third to one-half of the total cases of CVD. The lifetime risk of CHD was illustrated in the Framingham Heart Study of 7733 participants, ages 40 to 94, who were initially free of CHD [1]. The lifetime risk for individuals at age 40 was 49 percent in men and 32 percent in women. Even those who were free from CHD at age 70 had a non-trivial lifetime risk of developing CHD (35 and 24 percent in males and females, respectively).

Death rates from CVD, CHD, and stroke have declined in the United States since 1975, and evidence from 2000 to 2008 shows a decline in CHD mortality (figure 1). CVD and its related complications remain highly prevalent and expensive to treat [2]. CVD is the leading cause of death in most developed countries, with approximately one million Americans dying annually from CVD [3]. The prevalence of CVD is rapidly increasing in resource-limited countries as well.

Many risk factors for CVD are modifiable by specific preventive measures. In the worldwide INTERHEART study of patients from 52 countries, nine potentially modifiable factors accounted for over 90 percent of the population-attributable risk of a first MI: smoking, dyslipidemia, hypertension, diabetes, abdominal obesity, psychosocial factors, lack of daily consumption of fruits and vegetables, regular alcohol consumption, and lack of regular physical activity [4].

Most patients with CVD have at least one established or borderline risk factor other than age and sex [5-7]. However, it is clear that there are other risk factors, not all treatable, that may be important. The additive value of screening for these risk factors has not been firmly established, and there is only limited evidence that targeted therapeutic intervention will reduce the risk associated with these factors [8].

EVALUATION OF PUTATIVE CVD RISK FACTORS — Prospective observational studies are typically used to assess whether a risk factor contributes to risk. The starting point in the analyses are often the variables used to predict the CVD outcome of interest. For instance, for three-point major adverse cardiovascular events, the factors are typically age, sex, blood pressure, cholesterol (or low-density lipoprotein [LDL] cholesterol), high-density lipoprotein (HDL) cholesterol, diabetes mellitus, and cigarette smoking. Robust analyses include use of the key factors in a Cox proportional hazards model followed by assessment of the effects of adding the newer factor(s) to the prediction, with evaluation of the incremental effect on the C-statistic or on reclassification. Analyses that do not include this strategy and simply assess the impact of adding a new putative risk factor to national risk factor prediction algorithms such as the pooled cohort equations published in 2014 are generally not a satisfactory assessment to establish utility for a potential new CVD risk factor [9].

CARDIOVASCULAR FACTORS

Arterial intima-media thickness — Carotid artery intima-media thickness (IMT) is linked to the atherosclerotic process because of its association with known cardiovascular risk factors [10-13]. A more extensive discussion of carotid IMT with CVD risk is presented separately. (See "Carotid intima-media thickness".)

Arterial stiffness — Arterial stiffness, measured as the aortic pulse wave velocity (PWV) between the carotid and femoral arteries, is a predictor of cardiovascular events [14,15]. This was demonstrated in a meta-analysis of 17 studies that included over 15,000 patients in whom aortic PWV had been correlated to clinical outcome [14]. The pooled relative risks for total cardiovascular events, cardiovascular mortality, and all-cause mortality were significantly increased comparing high versus low aortic PWV groups: 2.26 (95% CI 1.89-2.70), 2.02 (95% CI 1.68-2.42), and 1.90 (95% CI 1.61-2.24) respectively.

Arterial calcification — Calcium deposits in extracoronary arteries, particularly the aortic arch and abdominal aorta, may be a marker for CVD and an increase in cardiovascular events and overall mortality [16]. However, it is not certain if the predictive value of calcification is independent of other coronary risk factors [17].

Aortic arch – Calcification of the aortic arch is associated with a greater risk of CVD and total mortality.

In one study of 139,849 members of a health maintenance organization followed for a median of 28 years, aortic arch calcification identified on a chest radiograph was independently associated with an increased risk of coronary heart disease (CHD) in men (relative risk [RR] 1.3) and women (RR 1.2), as well as a 1.5-fold increased risk of ischemic stroke in women [18].

In a subset of 2408 patients from the Rotterdam Study who underwent computed tomography (CT) to quantify arterial calcification in the coronary arteries, intracranial and extracranial carotid arteries, and the aortic arch, increasing amounts of calcification in any vascular territory were associated with increases in mortality [16]. The largest associated increase in both total and cardiovascular mortality was seen with calcification in the aortic arch.

Ischemic strokes in such patients may reflect embolic as well as thrombotic events. (See "Embolism from atherosclerotic plaque: Atheroembolism (cholesterol crystal embolism)" and "Thromboembolism from aortic plaque".)

Abdominal aorta – In addition to thoracic aortic calcification, calcification of the abdominal aorta is also associated with an increased risk of CVD. In a meta-analysis of 10 studies (22,170 patients), calcification of the abdominal aorta was associated with significantly higher risks of four major endpoints [19]:

CHD events (five studies with 11,250 patients; RR 1.81, 95% CI 1.54-2.14)

Cerebrovascular disease events (four studies with 9736 patients; RR 1.37, 95% CI 1.22-3.54)

Combined CVD events (four studies with 4960 patients; RR 1.64, 95% CI 1.24-2.17)

CVD death (three studies with 4986 patients; RR 1.72, 95% CI 1.03-2.86)

Another method for establishing the presence and severity of aortic calcification and atherosclerotic plaque burden is measurement of aortic elastic properties, based on the biomechanics of pulse wave-vessel wall interaction determined by transesophageal echocardiography [20]. The specific elastance is of incremental value to other clinical variables.

Renal arteries – Calcification of the renal arteries has also been reported to be associated with greater overall mortality. Among a cohort of 4450 persons without known CVD who underwent whole-body CT, 622 (14 percent) were found to have calcification of one or both renal arteries [21]. Following adjustment for age, sex, and traditional cardiac risk factors, those with renal artery calcification had a significantly greater overall mortality (risk-adjusted hazard ratio [HR] 1.63, 95% CI 1.17-2.29).

Breast arteries – Breast arterial calcification (BAC) can be an incidental finding on mammography. in one cohort study, over 5000 women aged 60-79 years were recruited after attending mammography screening [22]. Over 6.5 years of followup, BAC presence was associated with increased incidence of atherosclerotic CVD (ASCVD) (HR 1.51 (95% CI, 1.08–2.11). Independent confirmation of these findings and analyses that follow the methods described above (see 'Evaluation of putative CVD risk factors' above) may help to support the use of BAC as a risk-enhancing factor for ASCVD. (See "Cardiovascular disease risk assessment for primary prevention: Risk calculators", section on 'ACC/AHA pooled cohort hard ASCVD risk calculator (2013), updated algorithm 2018'.)

Coronary artery calcification — Coronary artery calcification (CAC) detected by electron-beam CT or multidetector-row CT can be used to quantify the amount of calcium present in the coronary arteries of a given patient. The coronary calcium score correlates with the risk of cardiovascular events in both asymptomatic and symptomatic patients. Issues related to the possible utility of CAC scanning are discussed separately. (See "Coronary artery calcium scoring (CAC): Overview and clinical utilization".)

Electrocardiographic abnormalities

ECG changes — Asymptomatic persons with resting electrocardiogram (ECG) abnormalities such as ST depression, T-wave inversion, left ventricular hypertrophy (LVH) or strain, and premature ventricular contractions have a 2 to 10-fold increased risk of CHD compared with those with a normal ECG [23-26]. As examples:

Among a cohort of 6991 persons aged 65 years or older who participated in the Copenhagen Heart Study and had a baseline resting ECG, 30.6 percent had baseline ECG abnormalities [26]. Over a median follow-up of 12 years, during which 2236 participants developed a fatal CVD event, the risk of fatal CVD was significantly greater in persons with an abnormal baseline ECG (HR 1.33 compared with normal baseline ECG, 95% CI 1.29-1.36).

Among 2192 adults aged 70 to 79 without known CVD who were followed for eight years, persons with minor (defined as minor ST-T abnormalities) and major (defined as Q waves, LVH, bundle branch block, atrial fibrillation or flutter, Wolff-Parkinson-White (WPW) syndrome, or major ST-T changes) ECG abnormalities were more likely to develop CVD than those with normal baseline ECGs (HR 1.35, 95% CI 1.02-1.81 and HR 1.51, 95% CI 1.20-1.90, respectively) [25].

The usual transition zone, where the R and S wave are equal in amplitude in the precordial leads, usually occurs between V3 and V4. In a cohort of 9067 persons (44 percent male) followed for 24 years, clockwise rotation (also called "late transition") was associated with increased risk of CVD (HR 1.49, 95% CI 1.12-1.98), while counterclockwise rotation (also called "early transition") was inversely associated with CVD (HR 0.74, 95% CI 0.59-0.94) [27]. While these data are easily identified from the surface ECG, the exact mechanism by which cardiac rotation on ECG alters the risk of CVD remains undetermined. (See "ECG tutorial: Miscellaneous diagnoses", section on 'Early or late transition'.)

Left ventricular hypertrophy — LVH, which is associated with hypertension as well as with age and obesity, is a risk factor for CHD [28-31]. This has been identified in several large cohorts, as illustrated by the following examples:

Electrocardiographic and anatomic LVH, based upon the finding of an enlarged cardiac silhouette on a chest radiograph, each independently increased the risk of CVD in the Framingham study [29]. Manifestations of atherosclerosis occurred at two to three times the general population rate in persons with LVH.

Among more than 15,000 patients in the Atherosclerosis Risk in Communities Study who were followed for 15 years, both women and men with baseline ECG-identified LVH were significantly more likely to die from CVD than from non-CVD causes (HR in women 8.4, 95% CI 4.5-15.6, HR in men 4.9, 95% CI 3.0-7.8) [30].

Echocardiographic evidence of LVH (eccentric or concentric), which is more sensitive than the ECG, also is predictive of cardiovascular risk.

Endothelial dysfunction — Endothelial dysfunction induced by dyslipidemia and oxidative stress is an initial step in atherosclerosis. Several studies have found that coronary artery endothelial dysfunction and increased vascular oxidative stress predict long-term progression of atherosclerosis and an increased incidence of cardiovascular events [32-34]. (See "Coronary endothelial dysfunction: Clinical aspects".)

Endothelial progenitor cells — Endothelial progenitor cells (EPCs) are derived from the bone marrow and are thought to support the vascular endothelium and prevent the development of atherosclerosis. (See "Coronary endothelial dysfunction: Clinical aspects".)

Decreased numbers of circulation EPCs have been associated with endothelial dysfunction and a higher Framingham risk factor score in patients without established CVD and more frequent atherosclerotic events including death at 10 to 12-month follow-up of patients with established CVD [35-37].

The mechanisms by which EPCs might be reduced in patients with or at risk for CVD are not well understood. Two possible contributing factors are selective functional exhaustion of hematopoietic progenitor cells in bone marrow and peripheral blood in ischemic cardiomyopathy and an increased plasma concentration of asymmetrical dimethylarginine (ADMA) [38,39].

Asymmetrical dimethylarginine — ADMA is an endogenous nitric oxide synthase inhibitor that may be an independent risk factor for endothelial dysfunction and CVD [40]. Patients with and without established CHD and elevated levels of serum ADMA have an increased risk of acute coronary events compared with individuals with lower ADMA levels [41-43]. In a systematic review and meta-analysis which included 19,842 patients from 22 studies (mean follow-up 7.1 years), individuals in the top tertile of ADMA levels were at significantly greater risk of CVD (RR 1.42, 95% CI 1.29-1.56), CHD (RR 1.39, 95% CI 1.19-1.62), and stroke (RR 1.60, 95% CI 1.33-1.91) compared with patients with ADMA levels in the lowest tertile.

Proton pump inhibitors (PPIs), commonly used to reduce gastric acid secretion in a variety of gastrointestinal disorders, increase ADMA levels by inhibiting the enzymatic degradation of ADMA, a result which has been shown ex vivo in cultured human endothelial cells and saphenous vein segments, and in vivo in a mouse model [44]. This suggests a potential mechanism for the increased number of cardiac events following coronary artery intervention among patients taking both clopidogrel and a PPI. (See "Clopidogrel resistance and clopidogrel treatment failure".)

Because the elevation of ADMA in patients taking PPIs is not specific to those also taking clopidogrel, it stands to reason that an increased risk for cardiovascular events may also extend to the general population of PPI users. However, there are no published data to support or refute this.

Heart rate — Resting and peak exercise heart rate may be predictive of cardiovascular and CHD mortality [45-51]. This has been studied in several population-based cohorts, the largest of which involved 53,322 persons (24.5 percent women) who were followed for an average of 15 years (but some as long as 30 years) [49]. Compared with persons with a resting heart rate <60 beats per minute, those with a resting heart rate of 80 or more beats per minutes had significantly higher all-cause (HR 1.66, 95% CI 1.45-1.89) and cardiovascular (HR 1.87, 95% CI 1.52-2.30) mortality.

Changes in resting heart rate over time also correlate with an increased risk of CHD death. In a prospective cohort study of 29,325 Norwegians (46 percent men) without known CHD, resting heart rate was measured on two occasions approximately 10 years apart, after which the group was followed for an average for 12 years [52]. Persons with a resting heart rate <70 bpm at the initial but >85 bpm at second visit had a significantly high risk of CHD death (adjusted HR 1.9 compared with persons with resting heart rate <70 bpm at both visits, 95% CI 1.0-3.6). Similar results were noted for persons with a resting heart rate of 70 to 85 bpm at the initial but >85 bpm at second visit (adjusted HR 1.8, 95% CI 1.2-2.8).

A lower-than-expected peak heart rate during exercise, known as chronotropic incompetence, is also predictive of CHD and all-cause mortality [53]. (See "Prognostic features of stress testing in patients with known or suspected coronary disease", section on 'Heart rate response to exercise'.)

Reduced heart rate variability is also associated with a greater risk of CHD. (See "Evaluation of heart rate variability".)

Retinal vessel caliber — Retinal arteriolar narrowing or retinal venous dilation is associated with an increased risk for the development of CHD [54,55]. A meta-analysis of over 22,000 patients in six studies suggested that this relationship is present only in women [55].

OTHER SYSTEMIC CONDITIONS

Autoimmune disease — Autoimmune diseases, and inflammatory conditions in general, are associated with an increased risk of CVD, likely due to chronic and systemic inflammation disease processes attributed to proinflammatory cytokines and autoantibodies.

In a 2022 population-based matched cohort study of over 440,000 persons with a new diagnosis of an autoimmune disease who were free from CVD in the following 12 months, 15.3 percent of people with an autoimmune disease developed incident CVD during a median follow-up of 6.2 years compared with 11.0 percent of controls [56]. Among the autoimmune diseases, the risk of CVD was highest for systemic sclerosis (hazard ratio [HR] 3.59, 95% CI 2.81-4.59), Addison’s disease (HR 2.83 95% CI 1.96-4.09]), systemic lupus erythematosus (HR 2.82, 95% CI 2.38-3.33), and type 1 diabetes (HR 2.36, 95% CI 2.21-2.52). The risk was also higher among adults under the age of 45 years (HR 2.33, 95% CI 2.16-2.51) than in those 55 to 64 years (HR 1.76, 95% CI 1.67-1.85) or ≥75 years (HR 1.30, 95% CI 1.24-1.36). The risk of CVD increased with the number of autoimmune diseases present (one disease: HR 1.41, 95% CI 1.37-1.45; two diseases: 2.63, 95% CI 2.49-2.78; three or more diseases: 3.79, 95% CI 3.36-4.27).

A discussion of CVD in several autoimmune diseases is discussed elsewhere. (See "Coronary artery disease in rheumatoid arthritis: Pathogenesis, risk factors, clinical manifestations, and diagnostic implications" and "Coronary artery disease in systemic lupus erythematosus" and "Cardiac manifestations of systemic sclerosis (scleroderma)".)

Androgen deficiency — Epidemiologic studies have shown that reduced serum testosterone concentrations are associated with the subsequent development of metabolic syndrome, diabetes mellitus, and elevated C-reactive protein levels in addition to higher overall mortality [57]. Among a cohort of 930 British men with angiographically documented coronary heart disease (CHD) prospectively observed for a mean of seven years, those with low testosterone levels (bio-available testosterone <2.6 nmol/L, 21 percent of the group) had significantly greater mortality (21 versus 12 percent in those with normal testosterone levels) [58].

Additionally, several observational studies and retrospective analyses of randomized trials have shown an association between therapeutically reduced androgen levels (ie, as treatment for prostate cancer) and higher rates of CVD and mortality. However, a systematic review and meta-analysis of randomized trials of androgen deprivation therapy (ADT) showed that ADT was not associated with increased risk of cardiovascular death [59]. Detailed discussions of these data are presented elsewhere. (See "Approach to older males with low testosterone" and "Side effects of androgen deprivation therapy", section on 'Potential cardiovascular harm'.)

Testosterone therapy — Normalization of testosterone levels with testosterone replacement therapy (TRT) may affect the risk of CVD in men. Also, the effects of testosterone therapy on cardiovascular risk in post-menopausal women are not clear. These issues are discussed separately (See "Testosterone treatment of male hypogonadism", section on 'Cardiovascular risks' and "Overview of androgen deficiency and therapy in females", section on 'Risks and side effects of androgen therapy'.)

Collagen vascular disease — Patients with collagen vascular disease, especially those with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), have a significantly increased incidence of CVD. In the Nurses' Health Study, those with RA for more than 10 years, compared with those without RA, had an increased risk of CVD (relative risk [RR] 3.1, 95% CI 1.64-5.87) [60]. An even higher RR has been demonstrated for patients with SLE, especially among young women [61]. This is discussed in greater detail elsewhere (see "Coronary artery disease in rheumatoid arthritis: Pathogenesis, risk factors, clinical manifestations, and diagnostic implications" and "Coronary artery disease in systemic lupus erythematosus" and "Overview of established risk factors for cardiovascular disease").

Infection — Acute infectious illnesses may be associated with a transient increase in the risk of cardiovascular events [62-64]. In addition, it has been proposed that certain types of infections may play a role in the pathogenesis of atherosclerosis by establishing a low-grade persistent inflammatory process; acute or chronic inflammation may result in endothelial dysfunction and may be responsible for a cardiac event.

Specific infectious organisms – The major organisms that have been studied with respect to chronic inflammation are Chlamydia pneumoniae, cytomegalovirus, and Helicobacter pylori; however, enterovirus (coxsackie viral infection), hepatitis A virus, and herpes simplex virus type 1 and type 2 have also been implicated.

Large randomized trials do not support benefit from antibiotic therapy against C. pneumoniae to reduce coronary events. (See "Pathogenesis of atherosclerosis", section on 'Infection'.)

Influenza – Several studies have shown an association between influenza infection and increased risk of acute MI within seven days after respiratory infection. (See "Seasonal influenza in adults: Clinical manifestations and diagnosis", section on 'Cardiac complications'.)

SARS-CoV-2 – The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is responsible for the coronavirus disease 2019 (COVID-19) pandemic, has been associated with myocardial injury and potential increased risk for MI and other vascular disease events [65,66]. This is discussed in detail elsewhere. (See "COVID-19: Evaluation and management of cardiac disease in adults" and "COVID-19: Myocardial infarction and other coronary artery disease issues".)

Endotoxin – It has been proposed that exposure to sustained high levels of endotoxin constitutes a risk factor for atherosclerosis [67].

Periodontitis – The potential association between chronic periodontitis and CHD is discussed elsewhere. (See "Epidemiology, pathogenesis, and clinical manifestations of odontogenic infections", section on 'Association with cardiovascular risk'.)

Iron overload — Heart disease is a manifestation of iron overload in hereditary hemochromatosis (HH). Whether minimal degrees of iron overload in HH heterozygotes increases cardiovascular risk remains controversial. (See "Clinical manifestations and diagnosis of hereditary hemochromatosis", section on 'Cardiac iron overload'.)

Nonalcoholic fatty liver disease — Nonalcoholic fatty liver disease (also referred to as nonalcoholic steatohepatitis) is a clinico-histopathological entity with features that resemble alcohol-induced liver injury that occurs in patients with little or no history of alcohol consumption. Although its etiology is unknown, it is frequently associated with obesity, type 2 diabetes mellitus, and hyperlipidemia.

While patients with nonalcoholic fatty liver disease often meet the diagnostic criteria for the metabolic syndrome, there is evidence for an increased risk of incident CVD that is independent of the risk conferred by traditional risk factors and components of the metabolic syndrome [68,69]. (See "Overview of established risk factors for cardiovascular disease", section on 'Metabolic syndrome' and "Epidemiology, clinical features, and diagnosis of nonalcoholic fatty liver disease in adults", section on 'Associated disorders'.)

Abnormal sleep — Abnormal sleep duration, both too little and too much, has been associated with an increased risk of CVD [70-72]. Obstructive sleep apnea is associated with an increased risk for coronary artery disease, cardiac arrhythmias, systemic hypertension, and pulmonary hypertension. These relationships are discussed elsewhere. (See "Insufficient sleep: Definition, epidemiology, and adverse outcomes", section on 'Cardiovascular morbidity' and "Obstructive sleep apnea and cardiovascular disease in adults".)

Small for gestational age — Adults who were small-for-gestational-age infants are at increased risk for ischemic heart disease and related disorders including hypertension, stroke, diabetes, and hypercholesterolemia [73-76]. It is possible that the association between small-for-gestational-age and CVD can be explained by other maternal or socioeconomic factors [75]. (See "Possible role of low birth weight in the pathogenesis of primary (essential) hypertension".)

CIRCULATING SERUM OR PLASMA MARKERS

Aldosterone — Elevated plasma aldosterone levels are seen in numerous clinical conditions, including hypertension and heart failure, with evidence suggesting that increased aldosterone levels are associated with greater CVD mortality [77,78]. Results from one prospective cohort study of 3153 patients (average follow-up 7.7 years) in whom plasma aldosterone levels were measured at the time of coronary angiography suggested greater all-cause and cardiovascular mortality among patients in the top three quartiles of plasma aldosterone concentration (most of whom had an aldosterone concentration considered to be in the normal range for this assay) compared with those in the lowest quartile, even after adjusting for traditional coronary heart disease (CHD) risk factors [77]. Further studies are needed before recommending this test in the general population.

Antibodies against oxidized LDL — In healthy men aged 50 years, circulating immune complexes and autoantibodies against oxidized low-density lipoproteins (LDL) and cardiolipin predict the occurrence of a myocardial infarction (MI) within the subsequent 20 years; the predictive power was independent of that of other risk factors [79]. In patients with CHD an elevated titer of antibodies against oxidized LDL independently predicts the progression of atherosclerosis over six months [80].

BNP and N-terminal pro-BNP — Brain natriuretic peptide (BNP) is a natriuretic hormone released from myocardial cells in response to volume expansion and possibly increased wall stress. The N-terminal fragment, N-pro-BNP, is also released into the circulation. Serum BNP and N-pro-BNP are increased in patients with heart failure and are predictors of death and cardiovascular events in asymptomatic patients without heart failure [81]. (See "Natriuretic peptide measurement in heart failure".)

Coagulation factors — The following coagulation factors may be related to cardiovascular risk:

Plasma fibrinogen levels – Fibrinogen levels are independent risk factors for CVD [82,83]. In a 2005 meta-analysis of over 150,000 healthy, middle-aged adults in 31 prospective studies, there was an approximately log-linear relationship between plasma fibrinogen and any CHD, any stroke, and mortality due to other cardiovascular events [84]. However, in a 2013 meta-analysis of 28 genome-wide association studies involving over 90,000 persons, none of the genetic polymorphisms associated with variations in circulating fibrinogen levels was associated with any increased risk of coronary artery disease or stroke, which raises questions about any genetic component to circulating fibrinogen levels [85].

It is not known whether elevated plasma fibrinogen plays a causal role in CVD progression. Animal experiments have shown that an increase in plasma fibrinogen can accelerate thrombus formation and increase blood vessel intimal thickness [86,87]. However, elevated plasma fibrinogen could simply be a marker of chronic vascular inflammation that in itself does not contribute to CVD progression. A meta-analysis of genetic fibrin variants associated with hyperfibrinogenemia found no increased risk of CVD, suggesting that elevated fibrinogen alone does not cause atherosclerosis [88]. Additional studies are necessary to determine if true causal relationships exist between hyperfibrinogenemia and CVD in humans and to determine if therapeutic interventions that lower plasma fibrinogen (or improve fibrinolysis) could reduce CVD.

Fibrin turnover – Preliminary data suggest an association between CHD and elevated levels of fibrin D-dimer, which reflects fibrin turnover in the circulation [89,90].

Thrombomodulin – This is an integral membrane glycoprotein that has a major role in the regulation of intravascular coagulation. It has anticoagulant activity since, in concert with thrombin, it activates protein C (see "Overview of hemostasis"). Soluble thrombomodulin showed a strong, graded, inverse association with coronary artery disease in one study [91], and one polymorphism of thrombomodulin was associated with an increased risk of CHD in Black persons.

von Willebrand factor – There are conflicting data as to whether elevated plasma von Willebrand factor (vWF) concentrations predict an increased risk of CVD [83,90,92-94]. A report from the Framingham Offspring Study found the higher levels of vWF were associated with an increased risk of CVD only in patients with diabetes mellitus [95].

Hemophilia – Patients with hemophilia and carriers of hemophilia appear to have a reduced risk of CHD mortality [96,97]. The protective effect against CHD appears to be less in carriers of hemophilia [98].

Coagulation factors – The relationship between the activity of coagulation factors XI, XII, VIII and IX, and the risk of a first MI in men was increased with higher levels of factor XI activity and lower levels of factor XII activity, and the highest risk was found among men with both high levels of factor XI and low factor XII activities [99]. Higher levels of factor VIII and IX activity were also associated with an increased risk of MI in men.

Homoarginine — Homoarginine, an amino acid derived from lysine that serves as a precursor of the vasodilator nitric oxide (NO), may enhance endothelial function, thereby lowering cardiovascular risk. In a cohort of 3305 patients with angiographically proven CHD (78 percent) or at high risk for CHD who were prospectively followed for nearly eight years, those in the lowest quartile of homoarginine levels had a fourfold higher risk of death from CVD (adjusted hazard ratio [HR] 4.1 compared with highest quartile, 95% CI 3.0-5.7) [100]. Further studies are needed before recommending this test in the general population.

Leptin — Leptin plays a role in fat metabolism and correlates with insulin resistance and other markers of the metabolic syndrome and was shown in the WOSCOPS trial of primary prevention to be predictive of CHD events after adjusting for body mass index (BMI) and other classic risk factors [101].

Serum phosphate — In patients with chronic kidney disease (CKD) or established CHD, a higher serum phosphate or calcium-phosphate product is associated with increased cardiovascular mortality. (See "Risk factors and epidemiology of coronary heart disease in end-stage kidney disease (dialysis)", section on 'Risk factors unique to chronic kidney disease' and "Vascular calcification in chronic kidney disease".)

There is also evidence that a higher serum phosphate level is associated with an increased risk of CVD in persons without known CHD or CKD [102,103].

Trimethylamine-N-oxide — Trimethylamine-N-oxide (TMAO), produced through intestinal microbial metabolism of dietary lecithin, appears to contribute to the development of atherosclerotic plaques through its interactions with macrophages and foam cells [104]. Plasma TMAO levels have been shown to decline following the suppression of intestinal microorganisms with oral broad-spectrum antibiotics and then return to near previous levels following the withdrawal of antibiotics [105].

TMAO levels were measured in a cohort of 4007 patients (mean age 63 years, 64 percent male) who underwent elective coronary angiography (74 percent of patients had one or more coronary artery with 50 percent or greater stenosis) and were followed for an average of three years [105]. After adjusting for standard cardiac risk factors, increased levels of plasma TMAO were associated with significantly greater risk of death, MI, or stroke (adjusted HR for highest versus lowest quartile 1.88, 95% CI 1.44-2.44).

In a systematic review and meta-analysis of 26,167 patients from 17 studies, with a mean follow-up of 4.3 years, high TMAO levels were associated with increased risk of all-cause death (HR 1.9, 95% CI 1.4-2.6), albeit with a high degree of heterogeneity between the studies (I2 = 94 percent) [106]. The risk of major adverse cardiac events was also significantly greater among patients with high TMAO levels (HR 1.7, 95% CI 1.3-2.1, I2 = 46 percent).

Increasing TMAO levels have also been correlated with increased atherosclerotic plaque burden [107].

Given the association between TMAO levels and atherosclerotic events as well as the potential for modification of TMAO levels based on intestinal microbiota, TMAO levels may be a future target for therapies aimed at lowering the risk of atherosclerotic events. Prior to this, however, these findings will need to be replicated in other populations.

TMAO levels have also been investigated as prognostic markers in patients with heart failure and possible acute coronary syndromes.

In a single-center cohort of 720 patients with heart failure who underwent coronary angiography and were followed for five years, median TMAO levels were higher in the heart failure cohort than in a healthy control cohort [108]. Within the heart failure cohort, 207 deaths occurred during the five-year follow-up; patients with TMAO levels in the highest quartile had significantly higher mortality (HR 3.4 compared with lowest quartile, 95% CI 2.2-5.2). As with TMAO levels and atherosclerotic events, TMAO levels in heart failure patients may be a future target for therapies aimed at lowering the risk of heart failure events, but additional data are needed that replicate these findings in other populations. (See "Epidemiology of heart failure".)

In a single-center cohort of 530 consecutive patients who presented with chest pain suspected to be cardiac in nature, elevated TMAO levels at presentation were associated with greater risk of major adverse cardiac events (MI, stroke, revascularization, and death) at 30 days (odds ratio [OR] 6.3, 95% CI 1.9-21.0) and six months (OR 5.7, 95% CI 1.9-16.7) as well as an increased risk of long-term mortality (OR 1.81 at 7 years, 95% CI 1.04-3.15) [109]. (See "Risk factors for adverse outcomes after non-ST elevation acute coronary syndromes", section on 'Biomarkers'.)

Urate — Large epidemiologic studies have shown that hyperuricemia is associated with an increased incidence of CHD and increased mortality in those with and without preexisting CHD [110-114]. Proposed mechanisms for such an increase in risk include the development of hypertension and oxidative stress [115,116]. It is unclear if hyperuricemia has a causal effect or, as has been more often suggested, is simply a marker for other risk factors such as hypertension, dyslipidemia, and diabetes [117-121].

Hyperuricemia has also been identified as a risk factor for mortality in patients with heart failure, where it may be a reflection of decreased tissue perfusion. (See "Predictors of survival in heart failure with reduced ejection fraction", section on 'Hyperuricemia'.)

Ceramides — Serum ceramides (the combination of sphingosine and a fatty acid) are being investigated as potential cardiovascular risk factors due to their role in atherosclerosis, diabetes, and inflammation [122]. Greater plasma ceramide levels are associated with an increased risk of cardiovascular death [123] and major adverse cardiac events [124] in patients with stable coronary artery disease, independent of traditional risk factors including lipid and C-reactive protein levels. Simvastatin has been reported to lower ceramide concentrations by approximately 25 percent [125]. However, measurement of serum ceramides is not widely available outside of research settings.

Angiotensin-converting enzyme 2 — In a global, community-based, case-cohort analysis of over 10,000 persons, analyzing data from the Prospective Urban Rural Epidemiology (PURE) study with a nine-year follow-up, higher plasma levels of angiotensin-converting enzyme (ACE) 2 were associated with greater risk of cardiovascular death (hazard ratio 1.40, 95% CI 1.27-1.54) as well as stroke and myocardial infarction, independent of age, sex, ancestry, systolic pressure, non-high-density lipoprotein (HDL) cholesterol, smoking, and diabetes [126].

MISCELLANEOUS FACTORS

Air pollution — In the early 21st century, air pollution emerged as a potentially modifiable risk factor for the development of CVD. Multiple observational studies have demonstrated an association between fine particulate air pollution (primarily from the use of fossil fuels in automobiles, power plants, and for heating purposes) and cardiovascular and cardiopulmonary outcomes, including mortality and acute coronary syndromes [102,127-136].

Specific components of air pollution that have been associated with CVD include fine particulate matter (eg, PM2.5), ozone, carbon monoxide, and sulfur oxides.

Both the American Heart Association (2020) and the European Society of Cardiology (2015) have issued official statements discussing the association between long-term exposure to air pollution and increased risk of developing CVD [137,138].

Short-term exposure to ozone and fine particulate matter have been associated with total mortality [139] and acute coronary ischemic events [132,140-142]. Conversely, regulation of pollution has been associated with short-term reductions in CVD events [143].

We summarize representative evidence on short-term air pollution exposure and risk of CVD:

Increased risk of myocardial infarction – In a systematic review and meta-analysis of data from 34 studies, exposure to carbon monoxide, nitrogen dioxide, sulfur dioxide, and small particulate matter (less than 10 microns and less than 2.5 microns) were all associated with an increased risk of myocardial infarction (MI), with the overall population attributable risk ranging from 1 to 5 percent [144].

Increased mortality at levels of air pollution that are lower than suggested air quality standards – In a case-crossover study of more than 22 million deaths between 2000 and 2012 from a United States Medicare database, short-term increases in two different types of air pollution were associated with an increased risk of all-cause mortality (fine particulate matter [RR increase 1.05 percent for each 10 mcg/m3 increase] and ozone [RR increase 0.51 percent for each 10 part-per-billion increase]) [139].

Exposure to air pollution immediately increases risk of acute coronary syndrome – A separate case-crossover study of 1.3 million people in China showed that an interquartile range increase in concentrations of fine particulate matter (PM2.5), nitric oxide, sulfur dioxide, and carbon monoxide in the 24 hours before onset of acute coronary syndrome were associated with 1.32, 3.89, 0.67, and 1.55 percent higher risks of acute coronary syndrome, respectively [145]. In this study, acute exposure to ozone was not associated with acute coronary syndrome. The association between PM2.5 and acute coronary syndrome was strongest in the concurrent hour of exposure and was then attenuated; the weakest effects were observed 15 to 29 hours after exposure.

Air pollution bans reduce CVD mortality – Further support for the significance of air pollution comes from a study of death rates in Dublin, Ireland before and after a ban on coal sales that led to a 70 percent reduction in black smoke concentrations [143]. Adjusted cardiovascular deaths fell by 10.3 percent in the six years after the ban.

In addition to short-term effects, there is evidence for an association between long-term air pollution exposure and CVD. We summarize representative evidence for long-term air pollution exposure and CVD:

Higher mortality from long-term exposure to air pollution – Fine particulate matter in the size 2.5 microns (known as PM2.5) is associated with cardiovascular mortality. Mortality data from nearly 450,000 patients in the American Cancer Society Cancer Prevention Study II database were correlated to geocoded air pollution data, including average concentrations of fine particulate matter (≤2.5 micrometers in diameter [PM2.5]) [133]. In multivariate analysis, PM2.5 (but not ozone) concentration was significantly associated with the risk of death from cardiovascular causes (RR 1.2, 95% CI 1.15-1.26).

Dose response relationship – Fine particulate matter (PM2.5) is associated with CVD and subclinical atherosclerosis in a dose response fashion. In the Women's Health Initiative Observational study of >65,000 postmenopausal women without prior CVD, the association between residential geocoded air pollution and a first cardiovascular event was evaluated [127]. Unlike prior studies, potential confounding factors such as age and other cardiovascular risk factors were taken into account. A participant’s exposure to a 10 mcg/m3 increase in pollution concentration was associated with an increased risk of any cardiovascular event (hazard ratio [HR] 1.24), death from CVD (HR 1.76), and cerebrovascular events (HR 1.35).

In the prospective MESA Air cohort study involving 6795 participants in six United States metropolitan areas, each increase in PM2.5 of 5 mcg/m3 was associated with 4.8 Agatston units per year progression of coronary calcification [146].

Possible mechanisms by which fine particulate air pollution may increase the risk of CVD include [137]:

An increase in mean resting arterial blood pressure through an increase in sympathetic tone and/or the modulation of basal systemic vascular tone [147]

An increase in the likelihood of intravascular thrombosis through transient increases in plasma viscosity and impaired endothelial dysfunction [148]

The initiation and promotion of atherosclerosis [146,149,150]

The impact of air pollution of the respiratory tract is discussed elsewhere. (See "Chronic obstructive pulmonary disease: Risk factors and risk reduction", section on 'Pollution, biomass, and occupational exposures'.)

Clonal hematopoiesis of indeterminate potential (CHIP) — The term "clonal hematopoiesis of indeterminate potential" (CHIP) is used to describe individuals who have a clonal mutation associated with hematologic neoplasia yet do not meet World Health Organization (WHO) criteria for diagnosis of a hematologic neoplasm. Patients identified with CHIP are thought to be at a higher risk of CVD. (See "Clonal hematopoiesis of indeterminate potential (CHIP) and related disorders of clonal hematopoiesis".)

Environmental noise — Observational studies have reported an association between chronic exposure to increased environmental noise levels (ie, road noise, noise from low-flying airplanes, etc) and an increased risk of developing CVD or death due to MI [151,152]. This effect is hypothesized to be due to stress-related dysregulation of the autonomic nervous system, leading to increases in hypertension and subsequent CVD.

Genetic markers — Several gene loci that affect the risk for the development of coronary heart disease (CHD) have been investigated in multiple studies, with a meta-analysis of 63,746 CHD cases and 130,681 controls reporting 46 single nucleotide polymorphisms (SNPs) across the genome which are significantly associated with an increased risk of CHD [153]. Among the various implicated SNPs, those appearing on locus 9p21 have shown the strongest association with CHD risk [154-157].

In a 2010 systematic review that evaluated 47 distinct data sets, including 35,872 cases and 95,837 controls, persons with two abnormal alleles at this locus were more likely to have CHD when compared with persons with one at-risk allele (odds ratio [OR] 1.25, 95% CI 1.21-1.29) [156].

The association with 9p21 SNPs and an increased risk of developing CHD has also been shown in older adult patients greater than 70 years of age without known prior CHD [158].

A 2014 systemic review and meta-analysis of 31 cohorts including 193,372 persons confirmed the association between 9p21 variants and the likelihood of a first CHD event (HR 1.19 per risk allele, 95% CI 1.17-1.22) [157]. However, 9p21 variants were not associated with an increased likelihood of subsequent CHD events among persons with known CHD (HR 1.01 per risk allele, 95% CI 0.97-1.06).

However, in spite of an apparently clear association between variants and incident CHD, locus 9p21 SNPs have not been definitively shown to significantly improve on the discrimination or classification of predicted CHD risk compared with the use of traditional risk factors [155,156,158].

A retrospective analysis evaluated the dose effect of abnormal 9p21 SNPs on the burden of CHD [159]. Among 950 non-diabetic patients with early-onset CHD (mean age 56 years) displaying at least one angiographic epicardial stenosis >50 percent seen by coronary angiography, the 9p21 genotype was associated with a risk of left main CHD (OR 2.38 per copy of risk allele, 95% CI 1.48-3.85), three-vessel CHD (OR 1.45 per copy of risk allele, 95% CI 1.18-1.79), and the need for bypass surgery (OR 1.37 per copy of risk allele, 95% CI 1.04-1.79). These data suggest more aggressive CHD occurring at a younger age in patients with abnormal 9p21 genotypes.

Different SNPs have been associated with different manifestations of CHD, with some increasing the risk of coronary atherosclerosis while others increase the risk of plaque rupture and acute MI, suggesting varying impacts of different mutations on the biology and pathophysiology of CHD [160,161]. Additionally, the pathophysiologic impact of genetic variants likely varies depending on other comorbid conditions. In a study which pooled CHD cases and controls from five large cohorts, the same SNP on chromosome 1q25 was associated with a significantly higher risk of CHD among patients with diabetes (OR 1.36 compared with diabetic patients without this SNP, 95% CI 1.22-1.51) but no change in risk of CHD among patients without diabetes (OR 0.99, 95% CI 0.87-1.13) [162]. This suggests the importance of other biologic factors in the development of CHD, and potentially different mechanisms in patients with and without diabetes.

Numerous other genetic abnormalities have been reported to be associated with the risk of CVD. As examples:

A specific polymorphism in the gene encoding 5-lipoxygenase activating protein (FLAP) has been associated with a twofold increase in the risk of MI and stroke [163]. A FLAP inhibitor reduced both serum C-reactive protein and other biomarkers of risk in patients who sustained an MI [164].

DAB2IP is a protein located on chromosome 9q33, which is thought to be involved in regulating cell proliferation apoptosis. In a case-control study of nearly 5000 people (1386 cases of CHD, 3532 controls), a polymorphism in this protein was associated with a great likelihood of CHD (OR 1.16, 95% CI 1.05-1.29) [165].

In a study of 180 genetic variants associated with height among 193,449 persons (including 65,066 cases with CHD and 128,383 controls without CHD), there was a significant increase in CHD risk with decreasing height (13.5 percent for each standard deviation [6 cm] decrease in height, 95% CI 5.4-22.1 percent), with a significant decrease in CHD risk with increasing numbers of height-raising genetic variants [166].

Peripheral blood cell gene expression has also been investigated as a means of estimating the risk of CVD, specifically obstructive CHD, and based on limited data the technique may be comparable to stress testing with myocardial perfusion imaging in terms of accuracy of diagnosing CHD [167-169]. However, the test cannot be used in patients with diabetes, which significantly limits its applicability. As of November 2014, peripheral blood cell gene expression is not widely available for clinical use.

Genetic studies of other cardiac risk factors (eg, cholesterol components) are discussed separately. (See "Inherited disorders of LDL-cholesterol metabolism other than familial hypercholesterolemia" and "HDL cholesterol: Clinical aspects of abnormal values", section on 'Low HDL cholesterol as a cause of ASCVD events'.)

Genetic risk scores — Although individual genetic markers are associated with CVD, their aggregate effect on risk beyond traditional factors has not been definitively established [170-174]. Several studies have evaluated the use of genetic risk scores as markers of cardiovascular risk:

A genetic risk score created from 101 SNPs associated with CVD did not improve discrimination or reclassification of risk after adjustment for traditional factors in a cohort of over 19,000 White women [171].

Using up to 50 SNPs to arrive at a polygenic risk score, the genetic risk for CHD was quantified in three prospective cohorts (ARIC, Women's Genome Health Study, and Malmo Diet and Cancer Study), along with assessment of healthy lifestyle factors (no current smoking, no obesity, regular physical activity, and healthy diet) [172]. Participants in the highest quintile of polygenic risk score had a 91 percent high risk of incident CHD events (HR 1.91, 95% CI 1.75-2.09). Participants with a favorable lifestyle (at least three of four healthy lifestyle factors) had a lower risk of CHD events at all levels of genetic risk, including a 46 percent reduction in risk among the highest quintile of polygenic risk score (HR 0.54, 95% CI 0.47-0.63), emphasizing the importance of lifestyle factors in CHD risk.

A coronary artery disease risk score that identified 163 coronary artery disease risk loci from genome-wide association study (GWAS) analyses has been published, and the authors state that approximately 30 to 40 percent of the heritability of coronary artery disease is accounted by the aggregate information in this analysis. Gene variants of special interest were associated with PCSK9, NPC1L1, LPA, LPL, APOC3, ANGPLT4, and ANGPTL3 [175].

In a study of over 55,000 participants, both genetic and lifestyle factors were associated coronary artery disease risk. Among participants at high genetic risk, a favorable lifestyle (defined as at least three of the four healthy lifestyle factors) was associated with a 46 percent lower relative risk of coronary events than an unfavorable lifestyle (defined as no or only one healthy lifestyle factor [172]. Other reports, based on more than 60,000 coronary artery disease cases and more than 120,000 controls, have shown that a coronary artery disease polygenic risk score is normally distributed and risk aggregates at right tail (high risk) of the distribution [176]. Investigators have begun to critically assess the relative merits of using polygenic risk scores to predict atherosclerotic CVD (ASCVD) events versus the traditional approach that uses information such as age, sex, smoking, diabetes mellitus status, lipid measures, blood pressure, and risk factors [177,178].

Socioeconomic and psychosocial factors

Socioeconomic factors – Social determinants of health, including education, occupation, place of residence, and income, or combinations of these, have been associated with an increased risk for CVD, especially CHD, and cardiovascular mortality [179-184].

Psychosocial factors – External situations that can potentially affect an individual's psychological health as well as the presence of mood disorders have both been linked with long term cardiovascular disease [185]. The precise role of psychological factors is difficult to delineate because of challenges in measuring, quantifying, and determining cause and effect. As an example, major depression can also be associated with established cardiovascular risk factors (such as smoking and low physical activity) [186,187].

Mood disorders – The topics most commonly considered have been depression and anxiety. In addition, psychological stress can affect risk factor control, compliance with healthy lifestyle patterns, and adherence to medications. (See "Psychosocial factors in coronary and cerebral vascular disease".)

Vitamins, antioxidants and homocysteine — Data are available assessing the effects of numerous vitamins, antioxidants, and homocysteine on CHD risk.

Oxidation of low-density lipoprotein (LDL) particles appears to be associated with an increased risk of atherosclerosis, suggesting that antioxidant therapy may reduce the incidence of CVD. Despite this association, randomized prospective studies of vitamin E, vitamin C, and beta-carotene have shown no clear evidence of benefit [188-190].

Both cross-sectional and prospective studies have linked elevated levels of homocysteine to increased risk for CHD. Higher serum homocysteine concentrations are frequently accompanied by reduced levels and intake of folate and vitamin B12. However, numerous prospective randomized trials of folate supplementation to lower serum homocysteine have demonstrated no reduction in major cardiovascular outcomes (eg, death, MI, stroke).

The addition of a serum homocysteine measurement to the Framingham risk score has been shown to improve risk prediction, with net reclassification of between 13 and 20 percent of patients from two cohorts [191]. The majority of those affected were reclassified to a higher risk level. The estimation of cardiovascular risk is discussed in greater detail separately. (See "Atherosclerotic cardiovascular disease risk assessment for primary prevention in adults: Our approach".)

Cysteine is structurally and metabolically related to homocysteine, and the plasma total cysteine concentration is strongly related to several risk factors for CVD [192]. Although both low and high plasma total cysteine concentrations have been associated with an increased risk of peripheral vascular disease, there is no significant relationship to coronary disease after adjustment for plasma homocysteine [193].

The effect of calcium supplementation on risk of CVD is controversial. While calcium supplementation may result in improvements in weight, blood pressure, and serum cholesterol concentrations (of approximately 5 percent) in patients with mild to moderate hypercholesterolemia, several meta-analyses have come to differing conclusions regarding CVD outcomes, with some suggesting an increased risk of CVD and others suggesting no increase in CVD risk. None of the trials (included in the meta-analyses), however, were designed to assess the effects of calcium or vitamin D on cardiovascular outcomes. As such, we do not restrict the use of calcium supplementation in patients with or at high risk for CVD and recommend optimal daily intake as discussed separately. (See "Calcium and vitamin D supplementation in osteoporosis", section on 'Side effects' and "Calcium and vitamin D supplementation in osteoporosis", section on 'Optimal intake'.)

MULTIVARIABLE RISK PROFILES — Multivariable risk models, such as the Framingham risk score and the SCORE project in Europe, have been developed in an attempt to estimate short-term (eg, 10-year) cardiovascular risk in patients without known CVD. These models and the clinical use of risk assessment are presented elsewhere. (See "Atherosclerotic cardiovascular disease risk assessment for primary prevention in adults: Our approach".)

Most risk-score algorithms assess risk over an intermediate follow-up interval of approximately 10 years and can help to guide interventions because categories of the risk factors are provided. Research from long-term cohort studies has also provided lifetime risk estimates.

Less detail on the individual risk factors is available with multivariable approaches to lifetime risk, but they can be used to help motivate patients to adhere to interventions. For example, the lifetime risk of CVD in a 50-year-old man with one major risk factor is approximately 10 percent by age 60 and rises to nearly 70 percent for men who live to age 90 years.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Assessment of cardiovascular risk".)

SUMMARY — An overview of the many possible emerging cardiovascular disease (CVD) risk factors that have a weak or proposed association with a greater risk of developing CVD is presented here. An overview of the established risk factors for CVD (eg, hyperlipidemia, hypertension, smoking) is discussed elsewhere. (See "Overview of established risk factors for cardiovascular disease".)

Cardiovascular factors – Cardiovascular factors associated with CVD include arterial intima-media thickness (IMT), arterial stiffness, arterial calcification, endothelial dysfunction, higher resting heart rates, and left ventricular hypertrophy (LVH). (See 'Cardiovascular factors' above.)

Systemic conditions – The systemic conditions associated with greater risk of CVD include collagen vascular disease, androgen deficiency, infection, iron overload, obstructive sleep apnea, and being born small for gestational age. (See 'Other systemic conditions' above.)

Circulating serum or plasma markers – These include aldosterone, antibodies against oxidized low-density lipoprotein (LDL), brain natriuretic peptide (BNP), coagulation factors, homoarginine, leptin, phosphate, trimethylamine-N-oxide (TMAO), and urate. (See 'Circulating serum or plasma markers' above.)

Other factors – Other factors such as genetic markers, air pollution, and socioeconomic factors have also been reported to be associated with greater risk of CVD. (See 'Miscellaneous factors' above.)

  1. Lloyd-Jones DM, Larson MG, Beiser A, Levy D. Lifetime risk of developing coronary heart disease. Lancet 1999; 353:89.
  2. Cooper R, Cutler J, Desvigne-Nickens P, et al. Trends and disparities in coronary heart disease, stroke, and other cardiovascular diseases in the United States: findings of the national conference on cardiovascular disease prevention. Circulation 2000; 102:3137.
  3. Benjamin EJ, Muntner P, Alonso A, et al. Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association. Circulation 2019; 139:e56.
  4. Yusuf S, Hawken S, Ounpuu S, et al. Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study. Lancet 2004; 364:937.
  5. Greenland P, Knoll MD, Stamler J, et al. Major risk factors as antecedents of fatal and nonfatal coronary heart disease events. JAMA 2003; 290:891.
  6. Khot UN, Khot MB, Bajzer CT, et al. Prevalence of conventional risk factors in patients with coronary heart disease. JAMA 2003; 290:898.
  7. Vasan RS, Sullivan LM, Wilson PW, et al. Relative importance of borderline and elevated levels of coronary heart disease risk factors. Ann Intern Med 2005; 142:393.
  8. Hackam DG, Anand SS. Emerging risk factors for atherosclerotic vascular disease: a critical review of the evidence. JAMA 2003; 290:932.
  9. Pencina MJ, D'Agostino RB Sr, D'Agostino RB Jr, Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med 2008; 27:157.
  10. de Groot E, Hovingh GK, Wiegman A, et al. Measurement of arterial wall thickness as a surrogate marker for atherosclerosis. Circulation 2004; 109:III33.
  11. Bots ML, Baldassarre D, Simon A, et al. Carotid intima-media thickness and coronary atherosclerosis: weak or strong relations? Eur Heart J 2007; 28:398.
  12. Davis PH, Dawson JD, Riley WA, Lauer RM. Carotid intimal-medial thickness is related to cardiovascular risk factors measured from childhood through middle age: The Muscatine Study. Circulation 2001; 104:2815.
  13. Järvisalo MJ, Jartti L, Näntö-Salonen K, et al. Increased aortic intima-media thickness: a marker of preclinical atherosclerosis in high-risk children. Circulation 2001; 104:2943.
  14. Vlachopoulos C, Aznaouridis K, Stefanadis C. Prediction of cardiovascular events and all-cause mortality with arterial stiffness: a systematic review and meta-analysis. J Am Coll Cardiol 2010; 55:1318.
  15. Cooper LL, Palmisano JN, Benjamin EJ, et al. Microvascular Function Contributes to the Relation Between Aortic Stiffness and Cardiovascular Events: The Framingham Heart Study. Circ Cardiovasc Imaging 2016; 9.
  16. Bos D, Leening MJ, Kavousi M, et al. Comparison of Atherosclerotic Calcification in Major Vessel Beds on the Risk of All-Cause and Cause-Specific Mortality: The Rotterdam Study. Circ Cardiovasc Imaging 2015; 8.
  17. Agmon Y, Khandheria BK, Meissner I, et al. Independent association of high blood pressure and aortic atherosclerosis: A population-based study. Circulation 2000; 102:2087.
  18. Iribarren C, Sidney S, Sternfeld B, Browner WS. Calcification of the aortic arch: risk factors and association with coronary heart disease, stroke, and peripheral vascular disease. JAMA 2000; 283:2810.
  19. Bastos Gonçalves F, Voûte MT, Hoeks SE, et al. Calcification of the abdominal aorta as an independent predictor of cardiovascular events: a meta-analysis. Heart 2012; 98:988.
  20. Hunziker PR, Imsand C, Keller D, et al. Bedside quantification of atherosclerosis severity for cardiovascular risk stratification: a prospective cohort study. J Am Coll Cardiol 2002; 39:702.
  21. Rifkin DE, Ix JH, Wassel CL, et al. Renal artery calcification and mortality among clinically asymptomatic adults. J Am Coll Cardiol 2012; 60:1079.
  22. Iribarren C, Chandra M, Lee C, et al. Breast Arterial Calcification: a Novel Cardiovascular Risk Enhancer Among Postmenopausal Women. Circ Cardiovasc Imaging 2022; 15:e013526.
  23. Daviglus ML, Liao Y, Greenland P, et al. Association of nonspecific minor ST-T abnormalities with cardiovascular mortality: the Chicago Western Electric Study. JAMA 1999; 281:530.
  24. Chou R, Arora B, Dana T, et al. Screening asymptomatic adults with resting or exercise electrocardiography: a review of the evidence for the U.S. Preventive Services Task Force. Ann Intern Med 2011; 155:375.
  25. Auer R, Bauer DC, Marques-Vidal P, et al. Association of major and minor ECG abnormalities with coronary heart disease events. JAMA 2012; 307:1497.
  26. Jørgensen PG, Jensen JS, Marott JL, et al. Electrocardiographic changes improve risk prediction in asymptomatic persons age 65 years or above without cardiovascular disease. J Am Coll Cardiol 2014; 64:898.
  27. Nakamura Y, Okamura T, Higashiyama A, et al. Prognostic values of clockwise and counterclockwise rotation for cardiovascular mortality in Japanese subjects: a 24-year follow-up of the National Integrated Project for Prospective Observation of Noncommunicable Disease and Its Trends in the Aged, 1980-2004 (NIPPON DATA80). Circulation 2012; 125:1226.
  28. Brown DW, Giles WH, Croft JB. Left ventricular hypertrophy as a predictor of coronary heart disease mortality and the effect of hypertension. Am Heart J 2000; 140:848.
  29. Kannel WB, Dannenberg AL, Levy D. Population implications of electrocardiographic left ventricular hypertrophy. Am J Cardiol 1987; 60:85I.
  30. Desai CS, Ning H, Lloyd-Jones DM. Competing cardiovascular outcomes associated with electrocardiographic left ventricular hypertrophy: the Atherosclerosis Risk in Communities Study. Heart 2012; 98:330.
  31. Estes EH, Zhang ZM, Li Y, et al. Individual components of the Romhilt-Estes left ventricular hypertrophy score differ in their prediction of cardiovascular events: The Atherosclerosis Risk in Communities (ARIC) study. Am Heart J 2015; 170:1220.
  32. Suwaidi JA, Hamasaki S, Higano ST, et al. Long-term follow-up of patients with mild coronary artery disease and endothelial dysfunction. Circulation 2000; 101:948.
  33. Schächinger V, Britten MB, Zeiher AM. Prognostic impact of coronary vasodilator dysfunction on adverse long-term outcome of coronary heart disease. Circulation 2000; 101:1899.
  34. Heitzer T, Schlinzig T, Krohn K, et al. Endothelial dysfunction, oxidative stress, and risk of cardiovascular events in patients with coronary artery disease. Circulation 2001; 104:2673.
  35. Hill JM, Zalos G, Halcox JP, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med 2003; 348:593.
  36. Schmidt-Lucke C, Rössig L, Fichtlscherer S, et al. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation 2005; 111:2981.
  37. Werner N, Kosiol S, Schiegl T, et al. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med 2005; 353:999.
  38. Kissel CK, Lehmann R, Assmus B, et al. Selective functional exhaustion of hematopoietic progenitor cells in the bone marrow of patients with postinfarction heart failure. J Am Coll Cardiol 2007; 49:2341.
  39. Thum T, Tsikas D, Stein S, et al. Suppression of endothelial progenitor cells in human coronary artery disease by the endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine. J Am Coll Cardiol 2005; 46:1693.
  40. Cooke JP. Asymmetrical dimethylarginine: the Uber marker? Circulation 2004; 109:1813.
  41. Valkonen VP, Päivä H, Salonen JT, et al. Risk of acute coronary events and serum concentration of asymmetrical dimethylarginine. Lancet 2001; 358:2127.
  42. Schnabel R, Blankenberg S, Lubos E, et al. Asymmetric dimethylarginine and the risk of cardiovascular events and death in patients with coronary artery disease: results from the AtheroGene Study. Circ Res 2005; 97:e53.
  43. Böger RH, Sullivan LM, Schwedhelm E, et al. Plasma asymmetric dimethylarginine and incidence of cardiovascular disease and death in the community. Circulation 2009; 119:1592.
  44. Ghebremariam YT, LePendu P, Lee JC, et al. Unexpected effect of proton pump inhibitors: elevation of the cardiovascular risk factor asymmetric dimethylarginine. Circulation 2013; 128:845.
  45. Kannel WB, Kannel C, Paffenbarger RS Jr, Cupples LA. Heart rate and cardiovascular mortality: the Framingham Study. Am Heart J 1987; 113:1489.
  46. Ekelund LG, Haskell WL, Johnson JL, et al. Physical fitness as a predictor of cardiovascular mortality in asymptomatic North American men. The Lipid Research Clinics Mortality Follow-up Study. N Engl J Med 1988; 319:1379.
  47. Greenland P, Daviglus ML, Dyer AR, et al. Resting heart rate is a risk factor for cardiovascular and noncardiovascular mortality: the Chicago Heart Association Detection Project in Industry. Am J Epidemiol 1999; 149:853.
  48. Kristal-Boneh E, Silber H, Harari G, Froom P. The association of resting heart rate with cardiovascular, cancer and all-cause mortality. Eight year follow-up of 3527 male Israeli employees (the CORDIS Study). Eur Heart J 2000; 21:116.
  49. Saxena A, Minton D, Lee DC, et al. Protective role of resting heart rate on all-cause and cardiovascular disease mortality. Mayo Clin Proc 2013; 88:1420.
  50. Ho JE, Larson MG, Ghorbani A, et al. Long-term cardiovascular risks associated with an elevated heart rate: the Framingham Heart Study. J Am Heart Assoc 2014; 3:e000668.
  51. Böhm M, Reil JC, Deedwania P, et al. Resting heart rate: risk indicator and emerging risk factor in cardiovascular disease. Am J Med 2015; 128:219.
  52. Nauman J, Janszky I, Vatten LJ, Wisløff U. Temporal changes in resting heart rate and deaths from ischemic heart disease. JAMA 2011; 306:2579.
  53. Lauer MS, Okin PM, Larson MG, et al. Impaired heart rate response to graded exercise. Prognostic implications of chronotropic incompetence in the Framingham Heart Study. Circulation 1996; 93:1520.
  54. Wong TY, Klein R, Sharrett AR, et al. Retinal arteriolar narrowing and risk of coronary heart disease in men and women. The Atherosclerosis Risk in Communities Study. JAMA 2002; 287:1153.
  55. McGeechan K, Liew G, Macaskill P, et al. Meta-analysis: retinal vessel caliber and risk for coronary heart disease. Ann Intern Med 2009; 151:404.
  56. Conrad N, Verbeke G, Molenberghs G, et al. Autoimmune diseases and cardiovascular risk: a population-based study on 19 autoimmune diseases and 12 cardiovascular diseases in 22 million individuals in the UK. Lancet 2022; 400:733.
  57. Ohlsson C, Barrett-Connor E, Bhasin S, et al. High serum testosterone is associated with reduced risk of cardiovascular events in elderly men. The MrOS (Osteoporotic Fractures in Men) study in Sweden. J Am Coll Cardiol 2011; 58:1674.
  58. Malkin CJ, Pugh PJ, Morris PD, et al. Low serum testosterone and increased mortality in men with coronary heart disease. Heart 2010; 96:1821.
  59. Nguyen PL, Je Y, Schutz FA, et al. Association of androgen deprivation therapy with cardiovascular death in patients with prostate cancer: a meta-analysis of randomized trials. JAMA 2011; 306:2359.
  60. Solomon DH, Karlson EW, Rimm EB, et al. Cardiovascular morbidity and mortality in women diagnosed with rheumatoid arthritis. Circulation 2003; 107:1303.
  61. Manzi S, Meilahn EN, Rairie JE, et al. Age-specific incidence rates of myocardial infarction and angina in women with systemic lupus erythematosus: comparison with the Framingham Study. Am J Epidemiol 1997; 145:408.
  62. Smeeth L, Thomas SL, Hall AJ, et al. Risk of myocardial infarction and stroke after acute infection or vaccination. N Engl J Med 2004; 351:2611.
  63. Madjid M, Miller CC, Zarubaev VV, et al. Influenza epidemics and acute respiratory disease activity are associated with a surge in autopsy-confirmed coronary heart disease death: results from 8 years of autopsies in 34,892 subjects. Eur Heart J 2007; 28:1205.
  64. Musher DM, Abers MS, Corrales-Medina VF. Acute Infection and Myocardial Infarction. N Engl J Med 2019; 380:171.
  65. Guan WJ, Ni ZY, Hu Y, et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 2020; 382:1708.
  66. Livingston E, Bucher K, Rekito A. Coronavirus Disease 2019 and Influenza 2019-2020. JAMA 2020; 323:1122.
  67. Wiedermann CJ, Kiechl S, Dunzendorfer S, et al. Association of endotoxemia with carotid atherosclerosis and cardiovascular disease: prospective results from the Bruneck Study. J Am Coll Cardiol 1999; 34:1975.
  68. Targher G, Day CP, Bonora E. Risk of cardiovascular disease in patients with nonalcoholic fatty liver disease. N Engl J Med 2010; 363:1341.
  69. Wong VW, Wong GL, Yip GW, et al. Coronary artery disease and cardiovascular outcomes in patients with non-alcoholic fatty liver disease. Gut 2011; 60:1721.
  70. Cappuccio FP, Cooper D, D'Elia L, et al. Sleep duration predicts cardiovascular outcomes: a systematic review and meta-analysis of prospective studies. Eur Heart J 2011; 32:1484.
  71. Lao XQ, Liu X, Deng HB, et al. Sleep Quality, Sleep Duration, and the Risk of Coronary Heart Disease: A Prospective Cohort Study With 60,586 Adults. J Clin Sleep Med 2018; 14:109.
  72. St-Onge MP, Grandner MA, Brown D, et al. Sleep Duration and Quality: Impact on Lifestyle Behaviors and Cardiometabolic Health: A Scientific Statement From the American Heart Association. Circulation 2016; 134:e367.
  73. Barker DJ. Fetal origins of coronary heart disease. BMJ 1995; 311:171.
  74. Barker DJ, Osmond C, Simmonds SJ, Wield GA. The relation of small head circumference and thinness at birth to death from cardiovascular disease in adult life. BMJ 1993; 306:422.
  75. Hübinette A, Cnattingius S, Ekbom A, et al. Birthweight, early environment, and genetics: a study of twins discordant for acute myocardial infarction. Lancet 2001; 357:1997.
  76. Lu D, Yu Y, Ludvigsson JF, et al. Birth Weight, Gestational Age, and Risk of Cardiovascular Disease in Early Adulthood: Influence of Familial Factors. Am J Epidemiol 2023; 192:866.
  77. Tomaschitz A, Pilz S, Ritz E, et al. Plasma aldosterone levels are associated with increased cardiovascular mortality: the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. Eur Heart J 2010; 31:1237.
  78. Ivanes F, Susen S, Mouquet F, et al. Aldosterone, mortality, and acute ischaemic events in coronary artery disease patients outside the setting of acute myocardial infarction or heart failure. Eur Heart J 2012; 33:191.
  79. Mustafa A, Nityanand S, Berglund L, et al. Circulating immune complexes in 50-year-old men as a strong and independent risk factor for myocardial infarction. Circulation 2000; 102:2576.
  80. Inoue T, Uchida T, Kamishirado H, et al. Antibody against oxidized low density lipoprotein may predict progression or regression of atherosclerotic coronary artery disease. J Am Coll Cardiol 2001; 37:1871.
  81. Wang TJ, Larson MG, Levy D, et al. Plasma natriuretic peptide levels and the risk of cardiovascular events and death. N Engl J Med 2004; 350:655.
  82. Stec JJ, Silbershatz H, Tofler GH, et al. Association of fibrinogen with cardiovascular risk factors and cardiovascular disease in the Framingham Offspring Population. Circulation 2000; 102:1634.
  83. Folsom AR, Wu KK, Rosamond WD, et al. Prospective study of hemostatic factors and incidence of coronary heart disease: the Atherosclerosis Risk in Communities (ARIC) Study. Circulation 1997; 96:1102.
  84. Fibrinogen Studies Collaboration, Danesh J, Lewington S, et al. Plasma fibrinogen level and the risk of major cardiovascular diseases and nonvascular mortality: an individual participant meta-analysis. JAMA 2005; 294:1799.
  85. Sabater-Lleal M, Huang J, Chasman D, et al. Multiethnic meta-analysis of genome-wide association studies in >100 000 subjects identifies 23 fibrinogen-associated Loci but no strong evidence of a causal association between circulating fibrinogen and cardiovascular disease. Circulation 2013; 128:1310.
  86. Kerlin B, Cooley BC, Isermann BH, et al. Cause-effect relation between hyperfibrinogenemia and vascular disease. Blood 2004; 103:1728.
  87. Machlus KR, Cardenas JC, Church FC, Wolberg AS. Causal relationship between hyperfibrinogenemia, thrombosis, and resistance to thrombolysis in mice. Blood 2011; 117:4953.
  88. Smith GD, Harbord R, Milton J, et al. Does elevated plasma fibrinogen increase the risk of coronary heart disease? Evidence from a meta-analysis of genetic association studies. Arterioscler Thromb Vasc Biol 2005; 25:2228.
  89. Danesh J, Whincup P, Walker M, et al. Fibrin D-dimer and coronary heart disease: prospective study and meta-analysis. Circulation 2001; 103:2323.
  90. Smith A, Patterson C, Yarnell J, et al. Which hemostatic markers add to the predictive value of conventional risk factors for coronary heart disease and ischemic stroke? The Caerphilly Study. Circulation 2005; 112:3080.
  91. Salomaa V, Matei C, Aleksic N, et al. Soluble thrombomodulin as a predictor of incident coronary heart disease and symptomless carotid artery atherosclerosis in the Atherosclerosis Risk in Communities (ARIC) Study: a case-cohort study. Lancet 1999; 353:1729.
  92. Wilson PW, Kauppila LI, O'Donnell CJ, et al. Abdominal aortic calcific deposits are an important predictor of vascular morbidity and mortality. Circulation 2001; 103:1529.
  93. Danesh J, Wheeler JG, Hirschfield GM, et al. C-reactive protein and other circulating markers of inflammation in the prediction of coronary heart disease. N Engl J Med 2004; 350:1387.
  94. Whincup PH, Danesh J, Walker M, et al. von Willebrand factor and coronary heart disease: prospective study and meta-analysis. Eur Heart J 2002; 23:1764.
  95. Frankel DS, Meigs JB, Massaro JM, et al. Von Willebrand factor, type 2 diabetes mellitus, and risk of cardiovascular disease: the framingham offspring study. Circulation 2008; 118:2533.
  96. Rosendaal FR, Varekamp I, Smit C, et al. Mortality and causes of death in Dutch haemophiliacs, 1973-86. Br J Haematol 1989; 71:71.
  97. Rosendaal FR, Briët E, Stibbe J, et al. Haemophilia protects against ischaemic heart disease: a study of risk factors. Br J Haematol 1990; 75:525.
  98. Srámek A, Kriek M, Rosendaal FR. Decreased mortality of ischaemic heart disease among carriers of haemophilia. Lancet 2003; 362:351.
  99. Doggen CJ, Rosendaal FR, Meijers JC. Levels of intrinsic coagulation factors and the risk of myocardial infarction among men: Opposite and synergistic effects of factors XI and XII. Blood 2006; 108:4045.
  100. März W, Meinitzer A, Drechsler C, et al. Homoarginine, cardiovascular risk, and mortality. Circulation 2010; 122:967.
  101. Wallace AM, McMahon AD, Packard CJ, et al. Plasma leptin and the risk of cardiovascular disease in the west of Scotland coronary prevention study (WOSCOPS). Circulation 2001; 104:3052.
  102. Dhingra R, Sullivan LM, Fox CS, et al. Relations of serum phosphorus and calcium levels to the incidence of cardiovascular disease in the community. Arch Intern Med 2007; 167:879.
  103. Foley RN, Collins AJ, Herzog CA, et al. Serum phosphorus levels associate with coronary atherosclerosis in young adults. J Am Soc Nephrol 2009; 20:397.
  104. Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011; 472:57.
  105. Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 2013; 368:1575.
  106. Schiattarella GG, Sannino A, Toscano E, et al. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J 2017; 38:2948.
  107. Senthong V, Li XS, Hudec T, et al. Plasma Trimethylamine N-Oxide, a Gut Microbe-Generated Phosphatidylcholine Metabolite, Is Associated With Atherosclerotic Burden. J Am Coll Cardiol 2016; 67:2620.
  108. Tang WH, Wang Z, Fan Y, et al. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol 2014; 64:1908.
  109. Li XS, Obeid S, Klingenberg R, et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur Heart J 2017; 38:814.
  110. Fang J, Alderman MH. Serum uric acid and cardiovascular mortality the NHANES I epidemiologic follow-up study, 1971-1992. National Health and Nutrition Examination Survey. JAMA 2000; 283:2404.
  111. Niskanen LK, Laaksonen DE, Nyyssönen K, et al. Uric acid level as a risk factor for cardiovascular and all-cause mortality in middle-aged men: a prospective cohort study. Arch Intern Med 2004; 164:1546.
  112. Choi HK, Curhan G. Independent impact of gout on mortality and risk for coronary heart disease. Circulation 2007; 116:894.
  113. Kuo CF, See LC, Yu KH, et al. Significance of serum uric acid levels on the risk of all-cause and cardiovascular mortality. Rheumatology (Oxford) 2013; 52:127.
  114. Kivity S, Kopel E, Maor E, et al. Association of serum uric acid and cardiovascular disease in healthy adults. Am J Cardiol 2013; 111:1146.
  115. Johnson RJ, Segal MS, Srinivas T, et al. Essential hypertension, progressive renal disease, and uric acid: a pathogenetic link? J Am Soc Nephrol 2005; 16:1909.
  116. Baker JF, Krishnan E, Chen L, Schumacher HR. Serum uric acid and cardiovascular disease: recent developments, and where do they leave us? Am J Med 2005; 118:816.
  117. Culleton BF, Larson MG, Kannel WB, Levy D. Serum uric acid and risk for cardiovascular disease and death: the Framingham Heart Study. Ann Intern Med 1999; 131:7.
  118. Wannamethee SG, Shaper AG, Whincup PH. Serum urate and the risk of major coronary heart disease events. Heart 1997; 78:147.
  119. Wheeler JG, Juzwishin KD, Eiriksdottir G, et al. Serum uric acid and coronary heart disease in 9,458 incident cases and 155,084 controls: prospective study and meta-analysis. PLoS Med 2005; 2:e76.
  120. Feig DI, Kang DH, Johnson RJ. Uric acid and cardiovascular risk. N Engl J Med 2008; 359:1811.
  121. Krishnan E, Sokolove J. Uric acid in heart disease: a new C-reactive protein? Curr Opin Rheumatol 2011; 23:174.
  122. Trayssac M, Hannun YA, Obeid LM. Role of sphingolipids in senescence: implication in aging and age-related diseases. J Clin Invest 2018; 128:2702.
  123. Laaksonen R, Ekroos K, Sysi-Aho M, et al. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur Heart J 2016; 37:1967.
  124. Meeusen JW, Donato LJ, Bryant SC, et al. Plasma Ceramides. Arterioscler Thromb Vasc Biol 2018; 38:1933.
  125. Tarasov K, Ekroos K, Suoniemi M, et al. Molecular lipids identify cardiovascular risk and are efficiently lowered by simvastatin and PCSK9 deficiency. J Clin Endocrinol Metab 2014; 99:E45.
  126. Narula S, Yusuf S, Chong M, et al. Plasma ACE2 and risk of death or cardiometabolic diseases: a case-cohort analysis. Lancet 2020; 396:968.
  127. Miller KA, Siscovick DS, Sheppard L, et al. Long-term exposure to air pollution and incidence of cardiovascular events in women. N Engl J Med 2007; 356:447.
  128. Brook RD, Franklin B, Cascio W, et al. Air pollution and cardiovascular disease: a statement for healthcare professionals from the Expert Panel on Population and Prevention Science of the American Heart Association. Circulation 2004; 109:2655.
  129. Pope CA 3rd, Burnett RT, Thun MJ, et al. Lung cancer, cardiopulmonary mortality, and long-term exposure to fine particulate air pollution. JAMA 2002; 287:1132.
  130. Samet JM, Dominici F, Curriero FC, et al. Fine particulate air pollution and mortality in 20 U.S. cities, 1987-1994. N Engl J Med 2000; 343:1742.
  131. Forastiere F, Stafoggia M, Picciotto S, et al. A case-crossover analysis of out-of-hospital coronary deaths and air pollution in Rome, Italy. Am J Respir Crit Care Med 2005; 172:1549.
  132. Pope CA 3rd, Muhlestein JB, May HT, et al. Ischemic heart disease events triggered by short-term exposure to fine particulate air pollution. Circulation 2006; 114:2443.
  133. Jerrett M, Burnett RT, Pope CA 3rd, et al. Long-term ozone exposure and mortality. N Engl J Med 2009; 360:1085.
  134. Kälsch H, Hennig F, Moebus S, et al. Are air pollution and traffic noise independently associated with atherosclerosis: the Heinz Nixdorf Recall Study. Eur Heart J 2014; 35:853.
  135. Xie W, Li G, Zhao D, et al. Relationship between fine particulate air pollution and ischaemic heart disease morbidity and mortality. Heart 2015; 101:257.
  136. Alexeeff SE, Deosaransingh K, Van Den Eeden S, et al. Association of Long-term Exposure to Particulate Air Pollution With Cardiovascular Events in California. JAMA Netw Open 2023; 6:e230561.
  137. Newby DE, Mannucci PM, Tell GS, et al. Expert position paper on air pollution and cardiovascular disease. Eur Heart J 2015; 36:83.
  138. Kaufman JD, Elkind MSV, Bhatnagar A, et al. Guidance to Reduce the Cardiovascular Burden of Ambient Air Pollutants: A Policy Statement From the American Heart Association. Circulation 2020; 142:e432.
  139. Di Q, Dai L, Wang Y, et al. Association of Short-term Exposure to Air Pollution With Mortality in Older Adults. JAMA 2017; 318:2446.
  140. Ruidavets JB, Cournot M, Cassadou S, et al. Ozone air pollution is associated with acute myocardial infarction. Circulation 2005; 111:563.
  141. Tonne C, Wilkinson P. Long-term exposure to air pollution is associated with survival following acute coronary syndrome. Eur Heart J 2013; 34:1306.
  142. Sinharay R, Gong J, Barratt B, et al. Respiratory and cardiovascular responses to walking down a traffic-polluted road compared with walking in a traffic-free area in participants aged 60 years and older with chronic lung or heart disease and age-matched healthy controls: a randomised, crossover study. Lancet 2018; 391:339.
  143. Clancy L, Goodman P, Sinclair H, Dockery DW. Effect of air-pollution control on death rates in Dublin, Ireland: an intervention study. Lancet 2002; 360:1210.
  144. Mustafic H, Jabre P, Caussin C, et al. Main air pollutants and myocardial infarction: a systematic review and meta-analysis. JAMA 2012; 307:713.
  145. Chen R, Jiang Y, Hu J, et al. Hourly Air Pollutants and Acute Coronary Syndrome Onset in 1.29 Million Patients. Circulation 2022; 145:1749.
  146. Kaufman JD, Adar SD, Barr RG, et al. Association between air pollution and coronary artery calcification within six metropolitan areas in the USA (the Multi-Ethnic Study of Atherosclerosis and Air Pollution): a longitudinal cohort study. Lancet 2016; 388:696.
  147. Brook RD, Brook JR, Urch B, et al. Inhalation of fine particulate air pollution and ozone causes acute arterial vasoconstriction in healthy adults. Circulation 2002; 105:1534.
  148. Pekkanen J, Peters A, Hoek G, et al. Particulate air pollution and risk of ST-segment depression during repeated submaximal exercise tests among subjects with coronary heart disease: the Exposure and Risk Assessment for Fine and Ultrafine Particles in Ambient Air (ULTRA) study. Circulation 2002; 106:933.
  149. Sun Q, Hong X, Wold LE. Cardiovascular effects of ambient particulate air pollution exposure. Circulation 2010; 121:2755.
  150. Bauer M, Moebus S, Möhlenkamp S, et al. Urban particulate matter air pollution is associated with subclinical atherosclerosis: results from the HNR (Heinz Nixdorf Recall) study. J Am Coll Cardiol 2010; 56:1803.
  151. Münzel T, Gori T, Babisch W, Basner M. Cardiovascular effects of environmental noise exposure. Eur Heart J 2014; 35:829.
  152. Héritier H, Vienneau D, Foraster M, et al. A systematic analysis of mutual effects of transportation noise and air pollution exposure on myocardial infarction mortality: a nationwide cohort study in Switzerland. Eur Heart J 2019; 40:598.
  153. CARDIoGRAMplusC4D Consortium, Deloukas P, Kanoni S, et al. Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet 2013; 45:25.
  154. Samani NJ, Erdmann J, Hall AS, et al. Genomewide association analysis of coronary artery disease. N Engl J Med 2007; 357:443.
  155. Paynter NP, Chasman DI, Buring JE, et al. Cardiovascular disease risk prediction with and without knowledge of genetic variation at chromosome 9p21.3. Ann Intern Med 2009; 150:65.
  156. Palomaki GE, Melillo S, Bradley LA. Association between 9p21 genomic markers and heart disease: a meta-analysis. JAMA 2010; 303:648.
  157. Patel RS, Asselbergs FW, Quyyumi AA, et al. Genetic variants at chromosome 9p21 and risk of first versus subsequent coronary heart disease events: a systematic review and meta-analysis. J Am Coll Cardiol 2014; 63:2234.
  158. Dutta A, Henley W, Lang IA, et al. The coronary artery disease-associated 9p21 variant and later life 20-year survival to cohort extinction. Circ Cardiovasc Genet 2011; 4:542.
  159. Dandona S, Stewart AF, Chen L, et al. Gene dosage of the common variant 9p21 predicts severity of coronary artery disease. J Am Coll Cardiol 2010; 56:479.
  160. Fan M, Dandona S, McPherson R, et al. Two chromosome 9p21 haplotype blocks distinguish between coronary artery disease and myocardial infarction risk. Circ Cardiovasc Genet 2013; 6:372.
  161. Hernesniemi JA, Lyytikäinen LP, Oksala N, et al. Predicting sudden cardiac death using common genetic risk variants for coronary artery disease. Eur Heart J 2015; 36:1669.
  162. Qi L, Qi Q, Prudente S, et al. Association between a genetic variant related to glutamic acid metabolism and coronary heart disease in individuals with type 2 diabetes. JAMA 2013; 310:821.
  163. Helgadottir A, Manolescu A, Thorleifsson G, et al. The gene encoding 5-lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nat Genet 2004; 36:233.
  164. Hakonarson H, Thorvaldsson S, Helgadottir A, et al. Effects of a 5-lipoxygenase-activating protein inhibitor on biomarkers associated with risk of myocardial infarction: a randomized trial. JAMA 2005; 293:2245.
  165. Harrison SC, Cooper JA, Li K, et al. Association of a sequence variant in DAB2IP with coronary heart disease. Eur Heart J 2012; 33:881.
  166. Nelson CP, Hamby SE, Saleheen D, et al. Genetically determined height and coronary artery disease. N Engl J Med 2015; 372:1608.
  167. Elashoff MR, Wingrove JA, Beineke P, et al. Development of a blood-based gene expression algorithm for assessment of obstructive coronary artery disease in non-diabetic patients. BMC Med Genomics 2011; 4:26.
  168. Rosenberg S, Elashoff MR, Beineke P, et al. Multicenter validation of the diagnostic accuracy of a blood-based gene expression test for assessing obstructive coronary artery disease in nondiabetic patients. Ann Intern Med 2010; 153:425.
  169. Thomas GS, Voros S, McPherson JA, et al. A blood-based gene expression test for obstructive coronary artery disease tested in symptomatic nondiabetic patients referred for myocardial perfusion imaging the COMPASS study. Circ Cardiovasc Genet 2013; 6:154.
  170. Khan SS, Post WS, Guo X, et al. Coronary Artery Calcium Score and Polygenic Risk Score for the Prediction of Coronary Heart Disease Events. JAMA 2023; 329:1768.
  171. Paynter NP, Chasman DI, Paré G, et al. Association between a literature-based genetic risk score and cardiovascular events in women. JAMA 2010; 303:631.
  172. Khera AV, Emdin CA, Drake I, et al. Genetic Risk, Adherence to a Healthy Lifestyle, and Coronary Disease. N Engl J Med 2016; 375:2349.
  173. Inouye M, Abraham G, Nelson CP, et al. Genomic Risk Prediction of Coronary Artery Disease in 480,000 Adults: Implications for Primary Prevention. J Am Coll Cardiol 2018; 72:1883.
  174. Gibson G. On the utilization of polygenic risk scores for therapeutic targeting. PLoS Genet 2019; 15:e1008060.
  175. Erdmann J, Kessler T, Munoz Venegas L, Schunkert H. A decade of genome-wide association studies for coronary artery disease: the challenges ahead. Cardiovasc Res 2018; 114:1241.
  176. Khera AV, Chaffin M, Aragam KG, et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat Genet 2018; 50:1219.
  177. Mosley JD, Gupta DK, Tan J, et al. Predictive Accuracy of a Polygenic Risk Score Compared With a Clinical Risk Score for Incident Coronary Heart Disease. JAMA 2020; 323:627.
  178. Elliott J, Bodinier B, Bond TA, et al. Predictive Accuracy of a Polygenic Risk Score-Enhanced Prediction Model vs a Clinical Risk Score for Coronary Artery Disease. JAMA 2020; 323:636.
  179. Kaplan GA, Keil JE. Socioeconomic factors and cardiovascular disease: a review of the literature. Circulation 1993; 88:1973.
  180. Diez Roux AV, Merkin SS, Arnett D, et al. Neighborhood of residence and incidence of coronary heart disease. N Engl J Med 2001; 345:99.
  181. Boehm JK, Peterson C, Kivimaki M, Kubzansky LD. Heart health when life is satisfying: evidence from the Whitehall II cohort study. Eur Heart J 2011; 32:2672.
  182. Kivimäki M, Nyberg ST, Batty GD, et al. Job strain as a risk factor for coronary heart disease: a collaborative meta-analysis of individual participant data. Lancet 2012; 380:1491.
  183. Virtanen M, Heikkilä K, Jokela M, et al. Long working hours and coronary heart disease: a systematic review and meta-analysis. Am J Epidemiol 2012; 176:586.
  184. Virtanen M, Nyberg ST, Batty GD, et al. Perceived job insecurity as a risk factor for incident coronary heart disease: systematic review and meta-analysis. BMJ 2013; 347:f4746.
  185. Wang YX, Sun Y, Missmer SA, et al. Association of early life physical and sexual abuse with premature mortality among female nurses: prospective cohort study. BMJ 2023; 381:e073613.
  186. Fluharty M, Taylor AE, Grabski M, Munafò MR. The Association of Cigarette Smoking With Depression and Anxiety: A Systematic Review. Nicotine Tob Res 2017; 19:3.
  187. Win S, Parakh K, Eze-Nliam CM, et al. Depressive symptoms, physical inactivity and risk of cardiovascular mortality in older adults: the Cardiovascular Health Study. Heart 2011; 97:500.
  188. Vivekananthan DP, Penn MS, Sapp SK, et al. Use of antioxidant vitamins for the prevention of cardiovascular disease: meta-analysis of randomised trials. Lancet 2003; 361:2017.
  189. Lonn E, Bosch J, Yusuf S, et al. Effects of long-term vitamin E supplementation on cardiovascular events and cancer: a randomized controlled trial. JAMA 2005; 293:1338.
  190. Cook NR, Albert CM, Gaziano JM, et al. A randomized factorial trial of vitamins C and E and beta carotene in the secondary prevention of cardiovascular events in women: results from the Women's Antioxidant Cardiovascular Study. Arch Intern Med 2007; 167:1610.
  191. Veeranna V, Zalawadiya SK, Niraj A, et al. Homocysteine and reclassification of cardiovascular disease risk. J Am Coll Cardiol 2011; 58:1025.
  192. El-Khairy L, Ueland PM, Nygård O, et al. Lifestyle and cardiovascular disease risk factors as determinants of total cysteine in plasma: the Hordaland Homocysteine Study. Am J Clin Nutr 1999; 70:1016.
  193. El-Khairy L, Ueland PM, Refsum H, et al. Plasma total cysteine as a risk factor for vascular disease: The European Concerted Action Project. Circulation 2001; 103:2544.
Topic 16307 Version 68.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟