Skip to main content
Erschienen in: Journal of Translational Medicine 1/2016

Open Access 01.12.2016 | Review

The Apoe−/− mouse model: a suitable model to study cardiovascular and respiratory diseases in the context of cigarette smoke exposure and harm reduction

verfasst von: Giuseppe Lo Sasso, Walter K. Schlage, Stéphanie Boué, Emilija Veljkovic, Manuel C. Peitsch, Julia Hoeng

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2016

Abstract

Atherosclerosis-prone apolipoprotein E-deficient (Apoe−/−) mice display poor lipoprotein clearance with subsequent accumulation of cholesterol ester-enriched particles in the blood, which promote the development of atherosclerotic plaques. Therefore, the Apoe−/− mouse model is well established for the study of human atherosclerosis. The systemic proinflammatory status of Apoe−/− mice also makes them good candidates for studying chronic obstructive pulmonary disease, characterized by pulmonary inflammation, airway obstruction, and emphysema, and which shares several risk factors with cardiovascular diseases, including smoking. Herein, we review the results from published studies using Apoe−/− mice, with a particular focus on work conducted in the context of cigarette smoke inhalation studies. The findings from these studies highlight the suitability of this animal model for researching the effects of cigarette smoking on atherosclerosis and emphysema.
Hinweise
Giuseppe Lo Sasso, Walter K. Schlage and Stéphanie Boué contributed equally to this manuscript

Overview

Atherosclerosis is a chronic disease in which systemic inflammation underlies the accumulation of plaques in the arterial intima. Plaques have a lipid-rich core and a thin fibrous cap that may rupture, causing thrombosis of the narrowed vessels [1]. If not properly controlled, atherosclerotic plaques can obstruct the arterial lumen, leading to cardiovascular diseases (CVDs), such as coronary heart disease (CHD), heart attack, stroke, or angina. More recently, an additional mechanism has been recognized, and it is responsible for fatal vascular occlusions predominantly in younger and female patients [2]. Indeed, thromboses can also arise from superficial erosion sites in vascular walls characterized by endothelial cell apoptosis on an underlying lipid-poor but proteoglycan- and glycosaminoglycan-rich plaque structure. It is important to highlight that statin treatment, lifestyle changes and other factors may favor the stabilization of the classical lipid-rich plaques [24]. Nevertheless, in 2011, CHD was responsible for around one in every seven deaths in the United States [5].
The development of CVD is associated with a number of risk factors, including smoking, physical inactivity, obesity, high blood pressure, diabetes, dyslipidemia, and genetic factors. Therapeutics targeted reducing CVD face a difficult pre-clinical hurdle because of a dearth of appropriate animal models that can capture the complexity of the human disease, which may take over 50 years to result in any clinically apparent event.
The development of animal models for CVD has increased our understanding of its pathophysiology, and allowed researchers to assess not only the etiologic effects of diet and environmental factors but also to evaluate the effectiveness of potential therapies [6]. The most widely used murine models for atherosclerosis are apolipoprotein E (Apoe) knockout (Apoe−/−) and low density lipoprotein (LDL) receptor deficient mice (Ldlr−/−), which both develop hypercholesterolemia [79]. The two models have both advantages and disadvantages depending on the goals of study. In fact, on a chow diet, Apoe−/− mice show higher plasma total cholesterol level compared with Ldlr−/−, and thus, develop severe atherosclerotic lesions as soon as a few weeks after birth [8, 10]. Moreover, the deficiency of the endogenous Apoe expression leads to an imbalance of cholesterol loading specifically in the macrophages. In turn, this stimulates cytokine and protease secretion, and triggers subsequent inflammation and extracellular matrix degradation [11]. These peculiar side effects related with the deficiency of Apoe but not Ldlr, confer to Apoe−/− mice greater adaptability for studying several other diseases associated with inflammation and extracellular matrix degradation [12], such as Alzheimer’s [13, 14], erectile dysfunction [15], diet-induced steatohepatitis [16, 17], and recently also chronic obstructive pulmonary disease [1821]. Other less frequently reported mouse models of atherosclerosis leverage alternative mechanisms of perturbing lipid metabolism. The hyperlipidemic APOE*3-Leiden mice express a human APOE isoform [22], and are described in greater detail below. In the scavenger receptor B1 (Scarb1) knockout mice, the atheroprotective HDL-C levels are elevated but—(apparently) paradoxically—atherosclerosis is increased [23, 24]. However, it has recently been shown that patients with a rare genetic inactivation of the SCARB1 also develop high levels of HDL-C associated with a higher atherosclerosis risk [24]. Double knockout mice for Scarb1 and Ldlr show more severe atherosclerosis and higher mortality than the single knockouts, particularly in combination with high fat diet [25]. For these mouse models of atherosclerosis, to the best of our knowledge, no smoke exposure studies have been reported in the literature.
The Apoe−/− model initiated a new era for CVD research. The Apoe−/− mouse was developed in 1992 by homologous recombination of embryonic stem cells, and it is currently the most widely used pre-clinical model of atherosclerosis [9, 26]. Apoe is a ligand for lipoprotein receptors involved in lipoprotein recognition and clearance. In particular, Apoe serves as a ligand that mediates the uptake of chylomicrons, very low-density lipoprotein (VLDL) and their remnants to hepatic receptors (LDL receptor and LDL receptor-related protein). Therefore, Apoe−/− mice show delayed lipoprotein clearance and consequently develop hyper- and dyslipoproteinemia [27], severe hypercholesterolemia, and atherosclerotic lesions even when on a normal diet. Arterial fat deposits are observed as early as 3 months after birth [28, 29]. By 8 months of age, the coronary arteries can be almost occluded when streptozotocin treatment is applied for additional acceleration [30, 31].
In the context of CVD development, the creation of genetically engineered mouse models became necessary also because of differences in the lipid homeostasis between human and murine organisms. For example, plasma cholesterol in wild-type mice on a regular chow diet is ~ 80 mg/dl, and it is primarily carried by high density lipoprotein (HDL) particles; moreover, these mice present small amounts of LDL and other atherogenic lipoproteins, such as VLDL remnants. This high HDL/LDL ratio is maintained even when mice are fed with high fat diet, indicating that wild-type mice have a high resistance to atherosclerosis development. In contrast, human lipid profiles show the majority of cholesterol in LDL particles (110 mg/dl) [32]. These characteristics make humans susceptible to HDL/LDL cholesterol ratio fluctuation, conferring a high risk of atherosclerosis and subsequent CVD. However, cholesterol transport and metabolism are sufficiently similar in the two species, suggesting that induced disturbances in plasma lipoprotein metabolism through gene manipulation would also lead to atherosclerosis in mice. Thus, the atherosclerotic lesions in Apoe−/− mice resemble human lesions in their sites of predilection and progression to the fibroproliferative stage [33, 34].
This review aims to highlight the usefulness of Apoe−/− mouse model in the context of inhalation toxicology studies addressing questions related to the role of cigarette smoke (CS), cessation, and candidate modified risk tobacco products (cMRTPs) [35] in the development of atherosclerosis and pulmonary diseases (Fig. 1).

Chronic obstructive pulmonary disease and its association with CVD

Chronic obstructive pulmonary disease (COPD) is typically considered to be a progressive lung disease, although it is increasingly thought to be a generalized disorder with a wide range of phenotypes [36]. COPD also shares some risk factors with CVD, including cigarette smoking.
Growing evidence suggests that, as in CVD, chronic systemic inflammation is an important factor in the development of COPD. Smoking causes, among other effects, a systemic inflammation, which may explain why smokers can develop both diseases, as well as other comorbidities such as cancer. Generally, COPD patients have an increased risk of developing CVD, even after correcting for risk factors such as smoking, and most COPD patients die from CVD or cancer rather than respiratory symptoms [37]. Despite their similarities, these disorders are often studied in isolation and the increased risk of CVD in COPD patients is often underestimated [36]. As interactions between the two diseases are not accounted for, the treatment of patients with both COPD and CVD is therefore often suboptimal [38].
Most animal models show resistance to the development of CS inhalation-related pathologies. This is why animal models with functional deficits are used to study the effects of CS [39]. The study of multiple disease endpoints in one mouse model would be very advantageous, as seen in a study of hyperlipidemic APOE*3-Leiden mice [22], which are prone to atherosclerosis [21]. These mice were intranasally administered low-dose lipopolysaccharide following the induction of emphysema by porcine pancreatic elastase. They exhibited pulmonary inflammation [increased interleukin (IL)-6 concentrations] and increased atherosclerotic lesion areas, but the presence of emphysema did not appear to accelerate atherosclerosis. This suggested that CVD/COPD therapy should aim to lower pulmonary and systemic inflammation, rather than simply treating emphysema.
Apoe−/− mice are a particularly useful strain to investigate comorbidities associated with cigarette smoking because, together with premature atherosclerosis [10], they show impaired alveologenesis [40] and develop emphysema [20, 41] (Fig. 2). In Apoe−/− mice fed a Western-type diet, severe systemic hypercholesterolemia accompanied by the abnormal cholesterol efflux induced pulmonary inflammation through a TLR4/inflammatory/MMP cascade, all of which ultimately result in emphysema development [20]. CS exposure of Apoe−/− mice clearly boosted these processes. In fact, CS increases oxidative stress, mitochondrial damage, and reduces glutathione levels, which in turn trigger arterial thrombosis and modulates the size and composition of neointimal lesions and thickening [4244].
In a recent study by Han and colleagues [21], the plasma cholesterol concentrations of Apoe−/− and Ldlr−/− mice responded differently to sidestream CS exposure, most probably because of the differences in lipoprotein metabolism between the two strains [45]. Those findings highlighted the valuable knowledge gained from studying two smoke-related pathologies in one animal model, but our recent literature searches on PubMed did not retrieve additional studies investigating CS exposure effects on Ldlr-/- mice.
The observed link between peripheral systemic changes in lipid metabolism and lung dysfunction seems to be a novel and clinically relevant factor in public health. As discussed above, it is already well established that patients with COPD have increased cardiovascular morbidity and mortality, but recent observations in both experimental models and humans have indicated that the reverse is also true, with the development of COPD in patients with atherosclerosis [46].

Human APOE polymorphism and CS: possible association with increased CVD risk

Diseases such as CVD and CHD are the result of physiological system homeostasis imbalance. Genetic (e.g., gene expression), environmental factors (e.g., smoking), or the interaction thereof may be the leading cause of these dangerous imbalances. Functional gene polymorphisms account for much of the different responses of human beings to environmental changes, increasing (or reducing) the risk of disease development.
In the context of CVD and APOE genotype, human populations show a very low frequency of APOE gene deficiencies. However, APOE is polymorphic in humans and plasma LDL cholesterol levels, hence atherosclerosis risks are strongly associated with the three common APOE isoforms, in the following order: APOE4, APOE3, and APOE2 [47]. Human APOE is a 299 amino acid protein whose isoforms are encoded by three APOE alleles (ε2, ε3, ε4) that give rise to six different genotypes (ε2/2, ε2/3, ε2/4 ε3/3, ε3/4 and ε4/4) [48]. The association between the three isoforms and the plasma cholesterol levels is attributed to the different affinity with the LDL receptor [49], secondary to the recognition and internalization of the APOE containing particles [5052]. Studies on the ε2 allele showed contrasting results. In fact, it was positively associated to hypertriglyceridemia [48], but also longevity [53] and negatively associated with myocardial infarction [54, 55]. APOE3 is the most common isoform, and it does not seem to be related to increased risk of CVD or other pathologies. On the contrary, the ε4 allele has been associated with CHD [47], Alzheimer’s disease [56], age-related cognitive decline [57], and other diseases [58]. Clinical studies conducted in the past 20 years showed not only the role of APOE polymorphisms on CVD occurrence, but also the interactions with environmental factors such as CS.
CS is associated with increased risk of CHD in men with all APOE genotypes, independently from the classical risk factors, including plasma lipid levels. The modification of the CHD risk associated with cigarette smoking by APOE polymorphism, was analyzed in the Northwick Park Heart Study II (NPHSII) [59], a prospective study of CHD in over 3000 UK men. In never-smokers, the CHD risk was similar in all APOE genotypes. The presence of the ε4 allele confined the CHD risk to current smokers alone and was independent of other classical CHD risk factors, including plasma lipid levels [60]. Interestingly in the ε4 ex-smokers, the risk decreased significantly, emphasizing the benefit of smoking cessation. The mechanism explaining this correlation could be detected in the higher levels of ROS in ε4 subjects [61]. Altogether, these studies highlighted the importance of the gene-environment interaction in the susceptibility to pathologies like CVD and CHD and its clear multifactorial nature.
The ability to study comorbidities in the Apoe−/− mouse model fulfils the “Reduction” (minimize the number of animals used per experiment or study) and “Refinement” (minimize the pain, suffering, distress or lasting harm that may be experienced by the animals) points of the 3R principle in animal research [62, 63]. Of note, the reduction and refinement together with the “Replacement” principle (aiming in developing methods which avoid or replace the use of animals in research), are fundamental towards achieving a good match between preclinical study outcomes and clinical trials in human beings.

Lipoprotein homeostasis in humans and mice

Lipoproteins are a complex and heterogeneous population of soluble macromolecular aggregates responsible for the transport of water-insoluble lipid molecules (mainly triglycerides, cholesterol, fatty acids, and phospholipids) from their site of synthesis (e.g., liver) or absorption from food (e.g., gut), to sites of use (e.g., muscle and adipose tissue). There are five types of lipoprotein particles (from smallest to largest): HDL, LDL, IDL, VLDL (high, low, intermediate, and very low-density lipoproteins), and chylomicrons. The lipid cores of these lipoprotein particles are coated by phospholipids and specific apolipoproteins that play a crucial role in many fundamental processes, such as efflux and transport [64], enzyme activation [65], and receptor binding [50, 51, 66].
Lipoprotein metabolism pathways differ according to the source of contained lipids and show species specificity that is important for preclinical studies.
Exogenous lipoproteins contain dietary fat (triglycerides and cholesterol) that is assembled in chylomicron particles [67]. Chylomicrons are released into the lacteal vessels, and at the capillaries in peripheral tissue as free fatty acids, transforming themselves into APOE-coated chylomicron remnants (CM). CM particles penetrate the fenestrated endothelium in the liver and will subsequently directly interact with ApoE-receptors, like the LDL receptor or the LDL receptor related protein 1 (LRP1) [68, 69]. The involvement of the LDL receptor in CM remnant clearance has been investigated showing contradictory results. In fact, although it accounts for the bulk clearance of serum lipoproteins into the liver, including the apoE-containing CM remnants, LDLR deficiency in both patients and animal models does not result in a defective clearance of CM remnants, indicating the existence of alternative APOE-specific remnant receptors for their clearance [70]. The LDL receptor related proteins might significantly contribute to the CM clearance. LRP1 belongs to the LRP family that represents a group of structurally related transmembrane proteins involved in a diverse range of biological activities [71]. The discovery that LRP1 binds APOE led to the notion that it acts as a remnant receptor. Consistently, LRP1 deficiency or inhibition does result in a decreased clearance of CM remnants.It is noteworthy that the metabolism of exogenous lipoproteins is preserved across species [72].
Conversely, the endogenous lipoprotein metabolism is associated with different key players between humans and mice, thus contributing to the missed overlap between lipid profiles of the two species under normal conditions (Fig. 3). Of note, ApoB is the main apolipoprotein of LDL, IDL and VLDL, fundamental particles in the endogenous lipid transport. Two molecular species of ApoB exist: ApoB100 is the full-length molecule that is synthesized in the liver, where VLDL particles are assembled for release into the circulation [73]. ApoB48 is produced by RNA editing from the same gene (APOB), and corresponds to the N-terminal fragment of ApoB100, therefore lacking the LDLR binding region. In mice this latter isoform is expressed in both the liver and in intestine, while in humans, it is only expressed in the intestine [74].
At the peripheral tissue capillaries, VLDL particles are transformed into IDL [68] after releasing free fatty acids. Part of the remaining VLDL particles and/or partially catalyzed VLDL particles bind to VLDL receptors expressed in peripheral tissue [75]. In mice, ApoB48-containing VLDLs and IDLs are internalized by the LRP8 expressed in the liver. Since the fractional catabolic rate for ApoB48-containing lipoproteins is very high compared with that of ApoB100-containing lipoproteins [76], VLDL and IDL almost disappear from mouse circulation, with the slightest contribution to LDL formation. LDL cholesterol thus appears very low in wild-type mice compared with humans (Fig. 3).
The second important difference between human and mouse endogenous lipoprotein distribution pertains to the HDL cholesterol that, contrarily to LDL, is very high in mice and low in humans. This difference is mainly due to the absence of the cholesterol ester transfer protein (CETP) in mouse plasma compared with the human one [77]. Thus, during the reverse cholesterol transport from peripheral tissue to the liver, HDL particles receive free cholesterol from macrophages through scavenger receptor type B-I (SR-BI) and ATP-binding Cassette proteins (ABCs), such as ABCA1 [78]. In humans, after lecithin: cholesterol acyltransferase (LCAT)-mediated esterification, the esterified cholesterol in HDL particles is transferred to VLDL, IDL, and LDL thanks to the CETP [79]. The absence of this enzyme in mice prevents the cholesterol depletion of HDLs that therefore, represent the major lipid-containing particle in the mouse plasma.

Apoe−/− model, CS, and atherosclerosis: an overview

Apoe−/− mice have been used to study the effects of diverse risk factors on the development of atherosclerosis (Table 1). A long-term (12-month) study assessed the impact of mainstream smoke and a high-fat diet both separately and in combination [27]. It revealed that mainstream smoke significantly increased atherosclerotic plaque size in the brachiocephalic artery, while both risk factors combined increased the number of elastin-rich layers of the plaques, thus accelerating atherosclerosis.
Table 1
Functional endpoint categories related to atherogenesis in studies conducted in Apoe−/− mice
Exposure category
Exposure route
Ref
Dyslipidemia
Inflammation
Thrombosis
Oxidative stress
Plaque development
Vascular function (constriction)
Vascular function (relaxation)
Vascular remodeling
Motor vehicle emissions
Intraperitoneal
[128]
     
↓→
↓→
 
[129]
   
→↗
    
Nasal
[130]
→↘
  
→↘↗
   
[131]
    
   
Oropharyngeal aspiration
[132]
    
 
Whole body inhalation exposure
[133]
   
↑↗
 
↑→↗
 
↑→↘↗
[134]
 
↑→
 
↑→↗
↓→↘↗
↑→
 
↓↑→↘↗
[93]
    
↑→↗
 
[135]
  
   
[136]
   
↓↑
  
[137]
 
↑→
  
  
[138]
 
↑→↗
 
↑→
  
↓→
↑↗
Environmental air particles
Intraperitoneal
[139]
     
↓↘
 
Oropharyngeal aspiration
[140]
    
   
Whole body inhalation exposure
[141]
    
   
[142]
    
 
[143]
    
↑↗
   
[144]
 
  
↑↗
  
↑↗
[145]
 
 
↑↑→
  
[93]
    
   
[146]
    
↑↑
 
[147]
    
   
[148]
  
    
[149]
   
   
[150]
    
   
Motor vehicle emissions and environmental air particles
Whole body inhalation exposure
[151]
 
      
[146]
    
↑↗
 
Environmental tobacco smoke surrogate (sidestream smoke)
Whole body inhalation exposure
[80]
    
↑→↗
   
[81]
       
[85]
  
     
[21]
   
   
[91]
   
   
[152]
   
↑↗
   
[153]
    
   
[93]
    
↑→↗
   
Functional categories related to plaque development were created according to the specific endpoint investigated. This approach facilitated the analysis of dissimilar sets of data. Arrows indicate increase (upwards), decrease (downwards), no change (to the right), trend to increase (north east), and trend to decrease (south east) (Modified from [94])
Another study used an Apoe−/− mouse model rather than larger animal models to provide more defined genetic background, greater statistical power in association with larger sample numbers, and more accurate lesion measurements [80]. The results showed that exposure to sidestream CS increased the size of atherosclerotic lesions, particularly in the thoracic region, and thus, the severity of atherosclerosis. This model was later used to test the effectiveness of dietary coenzyme Q, a lipid antioxidant, for preventing sidestream CS-induced formation of atherosclerotic plaques. Although coenzyme Q reduced the growth of spontaneous lesions, it did not significantly reduce the growth of smoke-induced plaques [81].
Atherosclerosis in the abdominal aorta can lead to the development of abdominal aortic aneurysms, which are reported to be about nine times more common in smokers than in non-smokers [82]. This process was investigated in an Apoe−/− mouse model with angiotensin II-induced hypertension, which developed aneurysms in association with enhanced matrix metalloproteinase (MMP) gene expression [83]. Exposure to CS further upregulated MMP expression in the abdominal aorta, and exacerbated the development and severity of aneurysms.
To investigate the effects of CS exposure on plaque thrombogenicity, Apoe−/− mice fed a high cholesterol diet were exposed to CS for 8 weeks (5 days/week). Following atherosclerotic plaque disruption, the tissue factor (TF) formed a complex with factors VII and VIIa during thrombin formation. Mice exposed to CS showed significantly greater TF immunoreactive areas than those exposed to filtered air, consistent with the TF immunoreactivity seen in human carotid plaques, albeit at lower levels [84]. Treatment with aspirin, an inhibitor of lipopolysaccharide-induced TF expression, attenuated the observed changes in TF in Apoe−/− mice, which again paralleled the reduction in TF immunoreactive areas seen in smokers undergoing carotid endarterectomy who were treated with aspirin prior to surgery.
Thrombin formation activates the clotting cascade and platelet activity. Exposure to CS significantly increased platelet–fibrinogen binding in Apoe−/− mice, leading to significantly greater thrombotic occlusion in these mice than in Apoe−/− mice exposed to filtered air [85]. These changes were partially reversed by treatment with the Purinergic receptor P2Y, G-protein coupled, 12 (P2Y12) antagonist, cangrelor. Another P2Y12 antagonist, clopidogrel, also had positive clinical effects in human smokers [86], supporting the applicability of this mouse model to human studies.
The development of atherosclerosis involves a number of inflammatory and vascular remodeling events. The enzyme cyclooxygenase-2 (COX-2) is involved in prostaglandin biosynthesis and is overexpressed in atherosclerotic lesions. It can be activated by β-catenin, a component of cell–cell interactions. Apoe−/− mice exposed to CS have been found to increase serum concentrations of the inflammatory cytokine interleukin-1, and enhanced translocation of β-catenin to the nucleus, leading to upregulation of COX-2 and inflammatory genes [87]. These cellular changes were proposed to underlie the proatherogenic effects in vascular tissue, while CS also disrupted the VE-cadherin–β-catenin complexes, induced phosphorylation events and increased vascular permeability [88].
The effects of CS exposure on immune suppression and subsequent atherosclerosis development were also examined in Apoe−/− mice [89]. This study showed that intimal thickening induced by carotid arterial cuffing was greater in mice exposed to CS compared with mice exposed to filtered air. These effects of CS were associated with reduced anti-malondialdehyde oxidized LDL IgG titers. This immune modulation is thought to increase thickening of the vessel wall.
Some of the atherogenic effects of CS are mediated via oxidative stress. Systemic oxidative stress is increased in human smokers. This effect was modeled in a study using Apoe−/− mice that reported significantly increased serum oxidative stress markers, including thiobarbituric acid-reactive substances, and oxidatively modified low-density lipoproteins. The serum and aortic 3-nitrotyrosine concentrations were also increased by exposure to CS, and these changes were alleviated by treatment with the antioxidant vitamin E [90].
The activity of mitochondrial superoxide dismutase 2 (SOD2) is reduced in mice exposed to CS. Notably, Apoe−/−Sod2+/− mice displayed significant increases in atherosclerotic lesion formation compared with control Apoe−/− littermates [91]. Thus, increased mitochondrial oxidant production, which could be influenced by mitochondrial and nuclear genetic variation in humans, may directly enhance atherogenic susceptibility.
Apoe−/− mouse models are also appropriate to investigate the atherosclerotic effects of other pollutants such as combustion emissions and ambient air fine particulate matter [9294], which induce inflammatory responses and generate reactive oxygen species. Thus, as previously described [94], Apoe−/− mice have been used in a diverse range of inhalation toxicology studies. These studies have assessed exposure to motor vehicle emissions, environmental air particles, and mainstream and sidestream CS, with or without the effects of a high-fat diet. The delivery methods and exposure times varied between studies. The pathophysiological events, including inflammation, thrombosis, CVD, oxidative stress, and atherogenesis (Table 1), and endpoints such as plaque development, protein marker expression, and systems toxicology assessments, also varied among the studies.

Apoe−/− mouse model to investigate toxicological mechanisms of CS, smoking cessation, and harm reduction approaches

CS is a complex aerosol mixture, for which more than 8000 identified smoke constituents have been reported [95]. Although it is not clear which particular CS constituents plays which role in any specific disease pathways, there is a general consensus on the main classes of chemicals from the combustion of tobacco that are harmful and potentially harmful [35, 96].
In the recent years, there has been a growing interest in harm reduction approaches to address the health risks of smoking [97100]. Smoking cessation remains the most effective approach to minimizing the risk for smoking-related diseases [101103]. However, only a small percentage of individuals manage to achieve long-term abstinence without support [104]. Although nicotine replacement therapies (NRT) (e.g., patches, gums, and nasal sprays) are often used as smoking cessation aids, they do not address the sensory and behavioral aspects of the smoking ritual, thus limiting their efficacy [105]. Therefore, there is a space for tobacco harm reduction approaches, which includes the development of less harmful products as alternatives to smoking cigarettes. These products are designed to deliver nicotine, sensory and behavioral aspects that smokers find satisfying while significantly reducing or eliminating the harmful and potentially harmful chemicals in CS and may be referred to as modified risk tobacco products (MRTPs). MRTPs were defined by the US Family Smoking Prevention and Tobacco Control Act of 2009 as “any tobacco product that is sold or distributed for use to reduce harm or the risk of tobacco related disease associated with commercially marketed tobacco products” [35]. Importantly, according with the US Food and Drug Administration published draft guidance on “modified risk tobacco product applications” [35], applications must provide scientific evidence to demonstrate that the product will “significantly reduce harm and the risk of tobacco-related diseases to individual tobacco users, and benefit the health of the population as a whole, taking into account both users of tobacco products and persons who do not currently use tobacco products.”
Combustion of tobacco results in both pyrolysis and pyrosynthesis of many harmful or potentially harmful constituents (HPHCs) [106108]. As the health risk associated with CS is due to the HPHCs it contains, the main driver for risk reduction is the reduction in exposure to HPHCs. Therefore, preventing combustion can produce, as compared with CS, a simpler aerosol with a strongly reduced content of these toxicants [19, 109, 110]. This principle underlies products based on the “heat-not-burn” principle, in which tobacco is electrically heated in a controlled fashion to release nicotine and flavors.
In this context, non-clinical studies in Apoe−/− mice are an invaluable preclinical tool for the evaluation of candidate MRTPs (cMRTPs), as the effect of such products can be compared with well characterized CS effects in the Apoe−/− mouse model (see Table 2).
Using a systems toxicology approach [111, 112], we were able to show that CS activates several response pathways in both respiratory (lung and nasal) and non-respiratory (liver, heart, and aorta) tissues. These responses, including inflammation, cell proliferation, lipid accumulation, complement, and tissue remodeling, were deactivated or strongly attenuated in Apoe−/− mice upon smoking cessation, as well as upon the exposure to aerosol from a cMRTP [18, 19, 113115].
Among these studies, particular interest was brought to the investigation of lipidomic profiles in different tissues (plasma, aorta, liver, and lungs) of Apoe−/− mice exposed to CS. CS increased the levels of multiple lipids, including ceramides, cholesteryl esters, and phosphatidylcholine species, and increased the development of atherosclerotic plaques [27, 103]. One study conducted in 2012 confirmed these observations [18, 116, 117]. The study results showed the effects exerted by both CS (3-month exposure) and cessation (3-month CS + 3 months fresh air) on Apoe−/− mice plasma and vascular tissue lipidomic profile. The CS-dependent increase of plasma and aortic lipid levels was reversed following smoking cessation, with a consequent decrease of most lipid concentrations, including total cholesterol, VLDL, phosphatidylcholine, and sphingomyelin [116, 117]. More recently, by using a systems toxicology approach, exposure effects were investigated using the classical toxicological endpoints related to both atherosclerosis and respiratory diseases, such as physiology and histology, combined with in depth molecular characterization of the transcriptome, proteome and lipidome [19, 94].
This study, together with others developed as part of a more comprehensive systems toxicology assessment framework [112115], confirmed previous observations showing a CS-dependent increase of atherogenic lipid composition of plasma and vascular tissue. Phillips and colleagues used a different timing for the analysis, exposing mice up to 8 months [19]. The cessation group was defined as the group receiving 2 months of CS and 6 months of fresh air. The recovery in the cessation group in this particular condition was much more pronounced than in the previous study with a 3-month CS exposure plus 3-month cessation period [117]. This is consistent with the assumption that shorter exposure periods combined with longer post-exposure periods are more efficient at restoring lipids back to less atherogenic levels.
Table 2
Summary of studies using Apoe−/− mice conducted by PMI
Disease, mechanism
References
Biological matrix
Endpoint
Effect of CS exposure
Effect of cessation
CVD
[116]
Aorta
Lipidomics
↑↗
ND
CVD
[117]
Aorta
Lipidomics
↓→
CVD
[19]
Aorta
Lipidomics
↓→
CVD
[42]
Carotid artery
Thrombosis
ND
CVD
[42]
Carotid artery
Endothelial injury
ND
CVD
[116]
Liver
Lipidomics
↑→
ND
CVD
[118]
Liver
Lipidomics
↑↗
↓↑
CVD
[114]
Liver
Lipidomics
↑↗
↓↘
CVD
[118]
Liver
Transcriptomics
→↗
CVD
[114]
Liver
Transcriptomics
↑↗
↓↘
CVD
[114]
Liver
Proteomics
↑↗
↓↘
CVD
[116]
Plasma
Lipidomics
↑↗
ND
CVD
[117]
Plasma
Lipidomics
↑→
↓→
CVD
[19]
Plasma
Lipidomics
↑↗
↓↘
COPD
[113]
Lung
Lipidomics
↑↗
↓↘
COPD
[113]
Lung
Transcriptomics
↑↗
↓↘
COPD
[113]
Lung
Proteomics
↑↗
↓↘
Atherosclerosis
[116]
Aorta
Plaque size
ND
Atherosclerosis
[117]
Aorta
Plaque size
↑↘
Atherosclerosis
[19]
Aorta
Plaque size
Atherosclerosis
[118]
Aorta
Plaque size
ND
Atherosclerosis
[29]
Aorta
Plaque size
ND
Atherosclerosis
[27]
BA
Plaque size
↑→
ND
Atherosclerosis
[27]
Aortic arch
Plaque size
↑→
ND
Exposure markers
[18]
Blood
COHb
Exposure markers
[18]
Urine
Nicotine metabolites
Exposure marker
[117]
Blood
COHb
Exposure marker
[118]
Blood
COHb
Exposure marker
[117]
Urine
Nicotine metabolites
Exposure markers
[19]
Blood
COHb
Exposure markers
[19]
Urine
Nicotine metabolites
Inflammation
[18]
Lung
BALF
↓↘
Inflammation
[18]
NRE
Histopathology
Inflammation
[18]
Lung
Transcriptomics
↑↓
↓↑
Inflammation
[116]
Liver
Transcriptomics
↑↗
ND
Inflammation
[124]
Lung
BALF
↑→
Inflammation
[29]
Aorta
Transcriptomics
ND
Inflammation
[127]
Lung
Transcriptomics
↑↓
ND
Inflammation
[127]
Lung
BALF
ND
Inflammation
[127]
Respiratory
Histopathology
ND
Inflammation
[127]
Respiratory
Network model
ND
Inflammation
[113]
Lung
Lipidomics
Inflammation
[19]
Urine
Inflammatory biomarker
Inflammation
[19]
Lung
BALF
Inflammation
[19]
NRE
Histopathology
Inflammation
[19]
Lung
Histopathology
COPD—emphysema
[18]
Lung
Lung morphometry
COPD—emphysema
[19]
Lung
Lung function
COPD—emphysema
[19]
Lung
Lung morphometry
Inflammation/cell cycle/necroptosis
[19]
Lung
Network model
Inflammation/cell cycle/necroptosis
[19]
Lung
Transcriptomics
Inflammation/cell cycle/necroptosis
[19]
Lung
Proteomics
Inflammation/cell cycle/necroptosis
[124]
Lung
Network model
Inflammation/cell cycle/necroptosis
[124]
Lung
Transcriptomics
Oxidative stress
[18]
Lung
BALF
↓↘
Oxidative stress
[18]
NRE
Transcriptomics
↑↓
↓↑
Oxidative stress
[114]
Liver
Transcriptomics
Oxidative stress
[114]
Liver
Proteomics
Arrows indicate increase (upwards), decrease (downwards), no change (to the right), trend to increase (north east), and trend to decrease (south east). Arrows for the cigarette smoke (CS) effect indicate changes compared with unexposed mice, and arrows for the cessation effect indicate differences compared with continued CS exposure
COHb carboxyhemoglobin; BALF bronchoalveolar lavage fluid; CVD cardiovascular disease; NRE nasal respiratory epithelium; COPD chronic obstructive pulmonary disease; BA brachiocephalic artery; ND not done
The effects of CS exposure and smoking cessation were also determined on hepatic lipid, transcriptomic, and proteomic profiles in Apoe−/− mice [114, 116, 118]. De Leon and colleagues [118] showed that the concentrations of many lipids, including free cholesterol, ceramides, and sphingomyelin, decreased in the liver of Apoe−/− mice following smoking cessation, although the triacylglycerol concentration increased. Hepatic triacylglycerol accumulation is indicative of fatty liver disease, but the observed increase did not appear to reflect decreased hepatic lipolysis or decreased pancreatic lipase. Gene set enrichment analysis of the transcriptomes revealed that fewer genes were differentially regulated in the smoking cessation group than in the CS exposed group. Genes showing differential expression included those involved in the cell cycle, cholesterol biosynthesis, and platelet activation. A more recent study partially confirmed these results [114]. In fact, analysis of the liver proteome and transcriptome showed upregulation of several enzymes involved in fatty acid and cholesterol metabolism upon CS inhalation. It should also be noted that the Apoe deficiency per se already causes a skewed hepatic lipid metabolism and a higher baseline of hepatic triglyceride accumulation compared with wild-type mice [119, 120]. Therefore, while signs of overt hepatotoxicity were absent, livers of Apoe−/− mice exposed to CS did exhibit an exacerbated dysregulation of fundamental hepatic processes such as lipid, xenobiotic and iron homeostasis compared with the sham (fresh air exposed) group. These data suggested that smoking alone cannot be regarded as a causative agent for liver diseases but, rather, as an accelerator in the presence of synergistic factors (alcohol, chronic liver diseases) [121123]. Finally, these changes were significantly reduced or absent when mice were exposed continuously to the cMRTP aerosol, upon cessation regimen or switching to the cMRTP [114].
Thomson and colleagues [124] developed a means of assessing the biological impact of exposure to biological and chemical substances based on hypotheses (typically, the predicted upstream regulators of the measured differentially expressed genes), subnetworks, and networks. Transcriptomics data were evaluated by computational modeling that combined the data with a priori knowledge from a knowledgebase to determine the effects at the molecular and systems levels. This approach has been applied to a number of models, including two smoking cessation studies using Apoe−/− mice [18, 19]. Both these studies revealed that in respiratory tract tissues, genes involved in the cell cycle, pulmonary inflammation, senescence, and necroptosis were significantly perturbed by CS exposure. This was corroborated by a decrease in the perturbation of biological mechanisms related to cell proliferation, tissue repair/angiogenesis, cell stress, cell fate, and pulmonary inflammation in the smoking cessation group compared with the group under continuous CS exposure. Similarly, the predicted increase in the immune cell response subnetworks was supported by a significant increase in the immune cell count and their secreted inflammatory mediators in bronchoalveolar lavage fluid.
Because advanced atherosclerotic lesions are difficult to recreate experimentally, little is known about the mechanisms underlying plaque instability and rupture. Instead, this was modeled in a vascular inflammatory processes network [29], which, like the lung-specific models applied for respiratory tract evaluations, used reverse causal reasoning [125] to explain and integrate differential gene expression data from large datasets. The latter approach showed that distinct molecular pathways are involved in different stages of atherosclerosis in Apoe−/− mice exposed to CS. Thus, the Apoe−/− mouse model shares many causal mechanisms with those of advanced atherosclerotic lesions in human coronary arteries, including endothelial cell activation, endothelial/monocyte interaction, foam cell formation, and plaque destabilization.
Stegemann and colleagues analyzed the lipid composition of human atherosclerotic plaques from carotid endarterectomies and reported a list of plaque-enriched lipid species, also with a strong contribution from sterol lipids, sphingolipids, and glycerophospholipids [126]. In Fig. 4, we present a comparison of the Apoe−/− murine aortic arch lipid species (data from [19]) that have these human plaque-enriched lipids in common. With the exception of two phosphatidylcholines (PCs) and one phosphatidylethanolamine (PE), all these common lipids were also significantly higher in aortic arches from Apoe−/− mice exposed for 8 months to CS compared with tissue from the sham-exposed controls. Conversely, no significant changes were observed following exposure to aerosol from an electrically heated cMRTP, or in mice that were switched to the cMRTP or to fresh air (cessation group) after a 2-month CS exposure. This correlation between human and murine plaque-enriched lipids is suggestive of related alterations in lipid metabolism in this mouse model of atherosclerosis. Taken together, these findings suggest that similar mechanisms are common to both species at the early stages of smoking-related atherosclerosis.
Another study used a systems biology approach together with reverse causal reasoning to investigate the mechanisms associated with emphysema development in five different mouse models, including Apoe−/− mice [127]. Emphysema is a component of COPD, but it appears to have a strong underlying genetic element in its development because not all smokers develop this disease. Combining large-scale transcriptomics data sets from four different studies, 39 biological and chemical substances based on hypotheses were identified as potential mechanisms associated with emphysema induced by CS exposure. The transcription nuclear factor κB, Th1-associated cytokines (tumor necrosis factor-α, IL-2, and interferon-c), IL-17A and IL-17F were predicted to increase following CS exposure. These fundamental mechanisms are likely to be applicable to human COPD.

Conclusions

To understand disease mechanisms in human, it is sometimes necessary to use animal models that can be characterized in depth. Oftentimes, models are used to study one particular condition. It is possible, however, to bring the model closer to the human situation by studying multiple comorbidities at once, as it is discussed in this review for the Apoe−/− mouse. Although atherogenesis is accelerated in Apoe−/− mice compared with humans, this rodent model is useful for assessing the mechanisms underlying the progression of atherosclerosis that are also relevant to humans owing to the similarities in the underlying mechanisms. In this review article we discuss scientific publications that overall demonstrated that the Apoe−/− mouse model is a suitable and relevant tool for examining the effects of CS, smoking cessation, or any product aimed at reducing the risk associated with CS, not only on atherosclerosis but also on lung inflammation and emphysema. In this context, the versatility of the Apoe−/− mouse model is highlighted by the possibility not only to allow analysis in terms of genetic, molecular, and physiological mechanisms in cardiovascular systems, but also to apply a similar “systems” approach to other respiratory and non-respiratory tissues. Thus, the use of the Apoe−/− mouse model appears very appropriate in the framework of inhalation toxicology studies.

Authors’ contributions

GLS, WKS and SB contributed to retrieving the data relevant for this review. GLS, WKS and SB drafted and revised the manuscript. SB and GLS prepared the figures. EV, MCP and JH revised the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We thank Edanz Group Ltd. for their editorial assistance during the development of this manuscript.

Competing interests

Authors are employees of Philip Morris International. Philip Morris International is the sole source of funding and sponsor of this project. W. K. Schlage is contracted and paid by Philip Morris International. The candidate Modified Risk Tobacco Product (cMRTP) described in here is a product developed by Philip Morris International.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Jin ZX, Xiong Q, Jia F, Sun CL, Zhu HT, Ke FS. Investigation of RNA interference suppression of matrix metalloproteinase-9 in mouse model of atherosclerosis. Int J Clin Exp Med. 2015;8:5272–8.PubMedPubMedCentral Jin ZX, Xiong Q, Jia F, Sun CL, Zhu HT, Ke FS. Investigation of RNA interference suppression of matrix metalloproteinase-9 in mouse model of atherosclerosis. Int J Clin Exp Med. 2015;8:5272–8.PubMedPubMedCentral
2.
Zurück zum Zitat Yahagi K, Davis HR, Arbustini E, Virmani R. Sex differences in coronary artery disease: pathological observations. Atherosclerosis. 2015;239:260–7.PubMedCrossRef Yahagi K, Davis HR, Arbustini E, Virmani R. Sex differences in coronary artery disease: pathological observations. Atherosclerosis. 2015;239:260–7.PubMedCrossRef
3.
Zurück zum Zitat Libby P, Pasterkamp G. Requiem for the ‘vulnerable plaque’. Eur Heart J. 2015;36:2984–7.PubMed Libby P, Pasterkamp G. Requiem for the ‘vulnerable plaque’. Eur Heart J. 2015;36:2984–7.PubMed
4.
Zurück zum Zitat Bairey Merz CN, Mark S, Boyan BD, Jacobs AK, Shah PK, Shaw LJ, Taylor D, Marbán E. Proceedings from the scientific symposium: sex differences in cardiovascular disease and implications for therapies. J Womens Health (Larchmt). 2010;19:1059–72.CrossRef Bairey Merz CN, Mark S, Boyan BD, Jacobs AK, Shah PK, Shaw LJ, Taylor D, Marbán E. Proceedings from the scientific symposium: sex differences in cardiovascular disease and implications for therapies. J Womens Health (Larchmt). 2010;19:1059–72.CrossRef
5.
Zurück zum Zitat Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, et al. Heart disease and stroke statistics–2015 update: a report from the American Heart Association. Circulation. 2015;131:e29–322.PubMedCrossRef Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, et al. Heart disease and stroke statistics–2015 update: a report from the American Heart Association. Circulation. 2015;131:e29–322.PubMedCrossRef
6.
Zurück zum Zitat Zaragoza C, Gomez-Guerrero C, Martin-Ventura JL, Blanco-Colio L, Lavin B, Mallavia B, Tarin C, Mas S, Ortiz A, Egido J. Animal models of cardiovascular diseases. J Biomed Biotechnol. 2011;2011:497841.PubMedPubMedCentralCrossRef Zaragoza C, Gomez-Guerrero C, Martin-Ventura JL, Blanco-Colio L, Lavin B, Mallavia B, Tarin C, Mas S, Ortiz A, Egido J. Animal models of cardiovascular diseases. J Biomed Biotechnol. 2011;2011:497841.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Véniant MM, Withycombe S, Young SG. Lipoprotein size and atherosclerosis susceptibility in Apoe−/− and Ldlr−/− mice. Arterioscler Thromb Vasc Biol. 2001;21:1567–70.PubMedCrossRef Véniant MM, Withycombe S, Young SG. Lipoprotein size and atherosclerosis susceptibility in Apoe−/− and Ldlr−/− mice. Arterioscler Thromb Vasc Biol. 2001;21:1567–70.PubMedCrossRef
8.
Zurück zum Zitat Ishibashi S, Brown MS, Goldstein JL, Gerard RD, Hammer RE, Herz J. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J Clin Invest. 1993;92:883.PubMedPubMedCentralCrossRef Ishibashi S, Brown MS, Goldstein JL, Gerard RD, Hammer RE, Herz J. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J Clin Invest. 1993;92:883.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E. Science. 1992;258:468–71.PubMedCrossRef Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E. Science. 1992;258:468–71.PubMedCrossRef
10.
Zurück zum Zitat Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R. ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler Thromb Vasc Biol. 1994;14:133–40.CrossRef Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R. ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler Thromb Vasc Biol. 1994;14:133–40.CrossRef
11.
Zurück zum Zitat Shaw PX. Rethinking oxidized low-density lipoprotein, its role in atherogenesis and the immune responses associated with it. Arch Immunol Ther Exp (Warsz). 2004;52:225–39. Shaw PX. Rethinking oxidized low-density lipoprotein, its role in atherogenesis and the immune responses associated with it. Arch Immunol Ther Exp (Warsz). 2004;52:225–39.
12.
Zurück zum Zitat Bink DI, Ritz K, Aronica E, van der Weerd L, Daemen MJ. Mouse models to study the effect of cardiovascular risk factors on brain structure and cognition. J Cereb Blood Flow Metab. 2013;33:1666–84.PubMedPubMedCentralCrossRef Bink DI, Ritz K, Aronica E, van der Weerd L, Daemen MJ. Mouse models to study the effect of cardiovascular risk factors on brain structure and cognition. J Cereb Blood Flow Metab. 2013;33:1666–84.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Choi J, Forster MJ, McDonald SR, Weintraub ST, Carroll CA, Gracy RW. Proteomic identification of specific oxidized proteins in ApoE-knockout mice: relevance to Alzheimer’s disease. Free Radic Biol Med. 2004;36:1155–62.PubMedCrossRef Choi J, Forster MJ, McDonald SR, Weintraub ST, Carroll CA, Gracy RW. Proteomic identification of specific oxidized proteins in ApoE-knockout mice: relevance to Alzheimer’s disease. Free Radic Biol Med. 2004;36:1155–62.PubMedCrossRef
14.
Zurück zum Zitat Crisby M, Rahman S, Sylven C, Winblad B, Schultzberg M. Effects of high cholesterol diet on gliosis in apolipoprotein E knockout mice: implications for Alzheimer’s disease and stroke. Neurosci Lett. 2004;369:87–92.PubMedCrossRef Crisby M, Rahman S, Sylven C, Winblad B, Schultzberg M. Effects of high cholesterol diet on gliosis in apolipoprotein E knockout mice: implications for Alzheimer’s disease and stroke. Neurosci Lett. 2004;369:87–92.PubMedCrossRef
15.
Zurück zum Zitat Behr-Roussel D, Darblade B, Oudot A, Compagnie S, Bernabé J, Alexandre L, Giuliano F. Erectile dysfunction in hypercholesterolemic atherosclerotic apolipoprotein E knockout mice. J Sex Med. 2006;3:596–603.PubMedCrossRef Behr-Roussel D, Darblade B, Oudot A, Compagnie S, Bernabé J, Alexandre L, Giuliano F. Erectile dysfunction in hypercholesterolemic atherosclerotic apolipoprotein E knockout mice. J Sex Med. 2006;3:596–603.PubMedCrossRef
16.
Zurück zum Zitat Tous M, Ferré N, Camps J, Riu F, Joven J. Feeding apolipoprotein E-knockout mice with cholesterol and fat enriched diets may be a model of non-alcoholic steatohepatitis. Mol Cell Biochem. 2005;268:53–8.PubMedCrossRef Tous M, Ferré N, Camps J, Riu F, Joven J. Feeding apolipoprotein E-knockout mice with cholesterol and fat enriched diets may be a model of non-alcoholic steatohepatitis. Mol Cell Biochem. 2005;268:53–8.PubMedCrossRef
17.
Zurück zum Zitat Bonomini F, Rodella L, Moghadasian M, Lonati C, Rezzani R. Apolipoprotein E deficiency and a mouse model of accelerated liver aging. Biogerontology. 2013;14:209–20.PubMedCrossRef Bonomini F, Rodella L, Moghadasian M, Lonati C, Rezzani R. Apolipoprotein E deficiency and a mouse model of accelerated liver aging. Biogerontology. 2013;14:209–20.PubMedCrossRef
18.
Zurück zum Zitat Boué S, De León H, Schlage WK, Peck MJ, Weiler H, Berges A, Vuillaume G, Martin F, Friedrichs B, Lebrun S. Cigarette smoke induces molecular responses in respiratory tissues of ApoE−/− mice that are progressively deactivated upon cessation. Toxicology. 2013;314:112–24.PubMedCrossRef Boué S, De León H, Schlage WK, Peck MJ, Weiler H, Berges A, Vuillaume G, Martin F, Friedrichs B, Lebrun S. Cigarette smoke induces molecular responses in respiratory tissues of ApoE−/− mice that are progressively deactivated upon cessation. Toxicology. 2013;314:112–24.PubMedCrossRef
19.
Zurück zum Zitat Phillips B, Veljkovic E, Boue S, Schlage WK, Vuillaume G, Martin F, Titz B, Leroy P, Buettner A, Elamin A, et al. An 8-month systems toxicology inhalation/cessation study in Apoe−/− mice to investigate cardiovascular and respiratory exposure effects of a candidate modified risk tobacco product, THS 2.2, compared with conventional cigarettes. Toxicol Sci. 2016;149:411–32.PubMedPubMedCentralCrossRef Phillips B, Veljkovic E, Boue S, Schlage WK, Vuillaume G, Martin F, Titz B, Leroy P, Buettner A, Elamin A, et al. An 8-month systems toxicology inhalation/cessation study in Apoe−/− mice to investigate cardiovascular and respiratory exposure effects of a candidate modified risk tobacco product, THS 2.2, compared with conventional cigarettes. Toxicol Sci. 2016;149:411–32.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Goldklang M, Golovatch P, Zelonina T, Trischler J, Rabinowitz D, Lemaître V, D’Armiento J. Activation of the TLR4 signaling pathway and abnormal cholesterol efflux lead to emphysema in ApoE-deficient mice. Am J Physiol Lung Cell Mol Physiol. 2012;302:L1200–8.PubMedPubMedCentralCrossRef Goldklang M, Golovatch P, Zelonina T, Trischler J, Rabinowitz D, Lemaître V, D’Armiento J. Activation of the TLR4 signaling pathway and abnormal cholesterol efflux lead to emphysema in ApoE-deficient mice. Am J Physiol Lung Cell Mol Physiol. 2012;302:L1200–8.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Han SG, Howatt DA, Daugherty A, Gairola CG. Atherogenic and pulmonary responses of ApoE-and LDL receptor-deficient mice to sidestream cigarette smoke. Toxicology. 2012;299:133–8.PubMedCrossRef Han SG, Howatt DA, Daugherty A, Gairola CG. Atherogenic and pulmonary responses of ApoE-and LDL receptor-deficient mice to sidestream cigarette smoke. Toxicology. 2012;299:133–8.PubMedCrossRef
22.
Zurück zum Zitat Khedoe PP, Wong MC, Wagenaar GT, Plomp JJ, van Eck M, Havekes LM, Rensen PC, Hiemstra PS, Berbee JF. The effect of PPE-induced emphysema and chronic LPS-induced pulmonary inflammation on atherosclerosis development in APOE*3-LEIDEN mice. PLoS One. 2013;8:e80196.PubMedPubMedCentralCrossRef Khedoe PP, Wong MC, Wagenaar GT, Plomp JJ, van Eck M, Havekes LM, Rensen PC, Hiemstra PS, Berbee JF. The effect of PPE-induced emphysema and chronic LPS-induced pulmonary inflammation on atherosclerosis development in APOE*3-LEIDEN mice. PLoS One. 2013;8:e80196.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Hoekstra M, Van Eck M. Mouse models of disturbed HDL metabolism. Handb Exp Pharmacol. 2015;224:301–36.PubMedCrossRef Hoekstra M, Van Eck M. Mouse models of disturbed HDL metabolism. Handb Exp Pharmacol. 2015;224:301–36.PubMedCrossRef
24.
Zurück zum Zitat Zanoni P, Khetarpal SA, Larach DB, Hancock-Cerutti WF, Millar JS, Cuchel M, DerOhannessian S, Kontush A, Surendran P, Saleheen D, et al. Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease. Science. 2016;351:1166–71.PubMedCrossRef Zanoni P, Khetarpal SA, Larach DB, Hancock-Cerutti WF, Millar JS, Cuchel M, DerOhannessian S, Kontush A, Surendran P, Saleheen D, et al. Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease. Science. 2016;351:1166–71.PubMedCrossRef
25.
Zurück zum Zitat Fuller M, Dadoo O, Serkis V, Abutouk D, MacDonald M, Dhingani N, Macri J, Igdoura SA, Trigatti BL. The effects of diet on occlusive coronary artery atherosclerosis and myocardial infarction in scavenger receptor class B, type 1/low-density lipoprotein receptor double knockout mice. Arterioscler Thromb Vasc Biol. 2014;34:2394–403.PubMedCrossRef Fuller M, Dadoo O, Serkis V, Abutouk D, MacDonald M, Dhingani N, Macri J, Igdoura SA, Trigatti BL. The effects of diet on occlusive coronary artery atherosclerosis and myocardial infarction in scavenger receptor class B, type 1/low-density lipoprotein receptor double knockout mice. Arterioscler Thromb Vasc Biol. 2014;34:2394–403.PubMedCrossRef
26.
Zurück zum Zitat Jawien J. The role of an experimental model of atherosclerosis: apoE-knockout mice in developing new drugs against atherogenesis. Curr Pharm Biotechnol. 2012;13:2435–9.PubMedCrossRef Jawien J. The role of an experimental model of atherosclerosis: apoE-knockout mice in developing new drugs against atherogenesis. Curr Pharm Biotechnol. 2012;13:2435–9.PubMedCrossRef
27.
Zurück zum Zitat Von Holt K, Lebrun S, Stinn W, Conroy L, Wallerath T, Schleef R. Progression of atherosclerosis in the Apo E−/− model: 12-Month exposure to cigarette mainstream smoke combined with high-cholesterol/fat diet. Atherosclerosis. 2009;205:135–43.CrossRef Von Holt K, Lebrun S, Stinn W, Conroy L, Wallerath T, Schleef R. Progression of atherosclerosis in the Apo E−/− model: 12-Month exposure to cigarette mainstream smoke combined with high-cholesterol/fat diet. Atherosclerosis. 2009;205:135–43.CrossRef
28.
Zurück zum Zitat Tamminen M, Mottino G, Qiao J, Breslow J, Frank J. Ultrastructure of early lipid accumulation in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 1999;19:847–53.PubMedCrossRef Tamminen M, Mottino G, Qiao J, Breslow J, Frank J. Ultrastructure of early lipid accumulation in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 1999;19:847–53.PubMedCrossRef
29.
Zurück zum Zitat De Leon H, Boue S, Schlage WK, Boukharov N, Westra JW, Gebel S, VanHooser A, Talikka M, Fields RB, Veljkovic E, et al. A vascular biology network model focused on inflammatory processes to investigate atherogenesis and plaque instability. J Transl Med. 2014;12:185.PubMedPubMedCentralCrossRef De Leon H, Boue S, Schlage WK, Boukharov N, Westra JW, Gebel S, VanHooser A, Talikka M, Fields RB, Veljkovic E, et al. A vascular biology network model focused on inflammatory processes to investigate atherogenesis and plaque instability. J Transl Med. 2014;12:185.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Tse J, Martin-McNaulty B, Halks-Miller M, Kauser K, DelVecchio V, Vergona R, Sullivan ME, Rubanyi GM. Accelerated atherosclerosis and premature calcified cartilaginous metaplasia in the aorta of diabetic male Apo E knockout mice can be prevented by chronic treatment with 17β-estradiol. Atherosclerosis. 1999;144:303–13.PubMedCrossRef Tse J, Martin-McNaulty B, Halks-Miller M, Kauser K, DelVecchio V, Vergona R, Sullivan ME, Rubanyi GM. Accelerated atherosclerosis and premature calcified cartilaginous metaplasia in the aorta of diabetic male Apo E knockout mice can be prevented by chronic treatment with 17β-estradiol. Atherosclerosis. 1999;144:303–13.PubMedCrossRef
31.
Zurück zum Zitat Gräbner R, Lötzer K, Döpping S, Hildner M, Radke D, Beer M, Spanbroek R, Lippert B, Reardon CA, Getz GS. Lymphotoxin β receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged ApoE−/− mice. J Exp Med. 2009;206:233–48.PubMedPubMedCentralCrossRef Gräbner R, Lötzer K, Döpping S, Hildner M, Radke D, Beer M, Spanbroek R, Lippert B, Reardon CA, Getz GS. Lymphotoxin β receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged ApoE−/− mice. J Exp Med. 2009;206:233–48.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Havel RJ, Kane, JP. Introduction: structure and metabolism of plasma lipoprotein. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The metabolic basis of inherited disease. New York: McGraw-Hill; 1989. p. 1129–38. Havel RJ, Kane, JP. Introduction: structure and metabolism of plasma lipoprotein. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The metabolic basis of inherited disease. New York: McGraw-Hill; 1989. p. 1129–38.
33.
Zurück zum Zitat Smith J, Breslow J. The emergence of mouse models of atherosclerosis and their relevance to clinical research. J Intern Med. 1997;242:99–109.PubMedCrossRef Smith J, Breslow J. The emergence of mouse models of atherosclerosis and their relevance to clinical research. J Intern Med. 1997;242:99–109.PubMedCrossRef
34.
Zurück zum Zitat Daugherty A, Rateri DL. Development of experimental designs for atherosclerosis studies in mice. Methods. 2005;36:129–38.PubMedCrossRef Daugherty A, Rateri DL. Development of experimental designs for atherosclerosis studies in mice. Methods. 2005;36:129–38.PubMedCrossRef
35.
Zurück zum Zitat Food and drug administration F. Modified risk tobacco product applications: draft guidance for industry. 2012. Food and drug administration F. Modified risk tobacco product applications: draft guidance for industry. 2012.
36.
37.
Zurück zum Zitat Hansell AL, Walk JA, Soriano JB. What do chronic obstructive pulmonary disease patients die from? A multiple cause coding analysis. Eur Respir J. 2003;22:809–14.PubMedCrossRef Hansell AL, Walk JA, Soriano JB. What do chronic obstructive pulmonary disease patients die from? A multiple cause coding analysis. Eur Respir J. 2003;22:809–14.PubMedCrossRef
38.
Zurück zum Zitat Andrus MR, Loyed JV. Use of beta-adrenoceptor antagonists in older patients with chronic obstructive pulmonary disease and cardiovascular co-morbidity: safety issues. Drugs Aging. 2008;25:131–44.PubMedCrossRef Andrus MR, Loyed JV. Use of beta-adrenoceptor antagonists in older patients with chronic obstructive pulmonary disease and cardiovascular co-morbidity: safety issues. Drugs Aging. 2008;25:131–44.PubMedCrossRef
39.
Zurück zum Zitat Kodavanti UP, Costa DL. Rodent models of susceptibility: what is their place in inhalation toxicology? Respir Physiol. 2001;128:57–70.PubMedCrossRef Kodavanti UP, Costa DL. Rodent models of susceptibility: what is their place in inhalation toxicology? Respir Physiol. 2001;128:57–70.PubMedCrossRef
40.
Zurück zum Zitat Massaro D, Massaro GD. Apoetm1Unc mice have impaired alveologenesis, low lung function, and rapid loss of lung function. Am J Physiol Lung Cell Mol Physiol. 2008;294:L991–7.PubMedCrossRef Massaro D, Massaro GD. Apoetm1Unc mice have impaired alveologenesis, low lung function, and rapid loss of lung function. Am J Physiol Lung Cell Mol Physiol. 2008;294:L991–7.PubMedCrossRef
41.
Zurück zum Zitat Arunachalam G, Sundar IK, Hwang JW, Yao H, Rahman I. Emphysema is associated with increased inflammation in lungs of atherosclerosis-prone mice by cigarette smoke: implications in comorbidities of COPD. J Inflamm. 2010;7:34.CrossRef Arunachalam G, Sundar IK, Hwang JW, Yao H, Rahman I. Emphysema is associated with increased inflammation in lungs of atherosclerosis-prone mice by cigarette smoke: implications in comorbidities of COPD. J Inflamm. 2010;7:34.CrossRef
42.
Zurück zum Zitat Schroeter MR, Sawalich M, Humboldt T, Leifheit M, Meurrens K, Berges A, Xu H, Lebrun S, Wallerath T, Konstantinides S. Cigarette smoke exposure promotes arterial thrombosis and vessel remodeling after vascular injury in apolipoprotein E-deficient mice. J Vasc Res. 2008;45:480–92.PubMedCrossRef Schroeter MR, Sawalich M, Humboldt T, Leifheit M, Meurrens K, Berges A, Xu H, Lebrun S, Wallerath T, Konstantinides S. Cigarette smoke exposure promotes arterial thrombosis and vessel remodeling after vascular injury in apolipoprotein E-deficient mice. J Vasc Res. 2008;45:480–92.PubMedCrossRef
43.
Zurück zum Zitat Knight-Lozano CA, Young CG, Burow DL, Hu ZY, Uyeminami D, Pinkerton KE, Ischiropoulos H, Ballinger SW. Cigarette smoke exposure and hypercholesterolemia increase mitochondrial damage in cardiovascular tissues. Circulation. 2002;105:849–54.PubMedCrossRef Knight-Lozano CA, Young CG, Burow DL, Hu ZY, Uyeminami D, Pinkerton KE, Ischiropoulos H, Ballinger SW. Cigarette smoke exposure and hypercholesterolemia increase mitochondrial damage in cardiovascular tissues. Circulation. 2002;105:849–54.PubMedCrossRef
44.
Zurück zum Zitat Biswas SK, Newby DE, Rahman I, Megson IL. Depressed glutathione synthesis precedes oxidative stress and atherogenesis in Apo-E−/− mice. Biochem Biophys Res Commun. 2005;338:1368–73.PubMedCrossRef Biswas SK, Newby DE, Rahman I, Megson IL. Depressed glutathione synthesis precedes oxidative stress and atherogenesis in Apo-E−/− mice. Biochem Biophys Res Commun. 2005;338:1368–73.PubMedCrossRef
45.
Zurück zum Zitat Schick S, Glantz S. Philip Morris toxicological experiments with fresh sidestream smoke: more toxic than mainstream smoke. Tob Control. 2005;14:396–404.PubMedPubMedCentralCrossRef Schick S, Glantz S. Philip Morris toxicological experiments with fresh sidestream smoke: more toxic than mainstream smoke. Tob Control. 2005;14:396–404.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Leavitt BJ, Ross CS, Spence B, Surgenor SD, Olmstead EM, Clough RA, Charlesworth DC, Kramer RS, O’Connor GT, Group NNECDS. Long-term survival of patients with chronic obstructive pulmonary disease undergoing coronary artery bypass surgery. Circulation. 2006;114:I430–4.PubMedCrossRef Leavitt BJ, Ross CS, Spence B, Surgenor SD, Olmstead EM, Clough RA, Charlesworth DC, Kramer RS, O’Connor GT, Group NNECDS. Long-term survival of patients with chronic obstructive pulmonary disease undergoing coronary artery bypass surgery. Circulation. 2006;114:I430–4.PubMedCrossRef
47.
Zurück zum Zitat Wilson PW, Schaefer EJ, Larson MG, Ordovas JM. Apolipoprotein E alleles and risk of coronary disease A meta-analysis. Arterioscler Thromb Vasc Biol. 1996;16:1250–5.PubMedCrossRef Wilson PW, Schaefer EJ, Larson MG, Ordovas JM. Apolipoprotein E alleles and risk of coronary disease A meta-analysis. Arterioscler Thromb Vasc Biol. 1996;16:1250–5.PubMedCrossRef
48.
Zurück zum Zitat Davignon J, Gregg RE, Sing CF. Apolipoprotein E polymorphism and atherosclerosis. Arterioscler Thromb Vasc Biol. 1988;8:1–21.CrossRef Davignon J, Gregg RE, Sing CF. Apolipoprotein E polymorphism and atherosclerosis. Arterioscler Thromb Vasc Biol. 1988;8:1–21.CrossRef
49.
Zurück zum Zitat Knouff C, Hinsdale ME, Mezdour H, Altenburg MK, Watanabe M, Quarfordt SH, Sullivan PM, Maeda N. Apo E structure determines VLDL clearance and atherosclerosis risk in mice. J Clin Invest. 1999;103:1579.PubMedPubMedCentralCrossRef Knouff C, Hinsdale ME, Mezdour H, Altenburg MK, Watanabe M, Quarfordt SH, Sullivan PM, Maeda N. Apo E structure determines VLDL clearance and atherosclerosis risk in mice. J Clin Invest. 1999;103:1579.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Windler E, Kovanen PT, Chao Y-S, Brown M, Havel RJ, Goldstein J. The estradiol-stimulated lipoprotein receptor of rat liver. A binding site that membrane mediates the uptake of rat lipoproteins containing apoproteins B and E. J Biol Chem. 1980;255:10464–71.PubMed Windler E, Kovanen PT, Chao Y-S, Brown M, Havel RJ, Goldstein J. The estradiol-stimulated lipoprotein receptor of rat liver. A binding site that membrane mediates the uptake of rat lipoproteins containing apoproteins B and E. J Biol Chem. 1980;255:10464–71.PubMed
51.
Zurück zum Zitat Brown MS, Goldstein JL. A receptor-mediated pathway for cholesterol homeostasis. Science. 1986;232:34–47.PubMedCrossRef Brown MS, Goldstein JL. A receptor-mediated pathway for cholesterol homeostasis. Science. 1986;232:34–47.PubMedCrossRef
52.
Zurück zum Zitat Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. 1988;240:622–30.PubMedCrossRef Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. 1988;240:622–30.PubMedCrossRef
53.
Zurück zum Zitat Rea IM, Mc Dowell I, McMaster D, Smye M, Stout R, Evans A. Apolipoprotein E alleles in nonagenarian subjects in the belfast elderly longitudinal free-living ageing study (BELFAST). Mech Ageing Dev. 2001;122:1367–72.PubMedCrossRef Rea IM, Mc Dowell I, McMaster D, Smye M, Stout R, Evans A. Apolipoprotein E alleles in nonagenarian subjects in the belfast elderly longitudinal free-living ageing study (BELFAST). Mech Ageing Dev. 2001;122:1367–72.PubMedCrossRef
54.
Zurück zum Zitat Kolovou G, Yiannakouris N, Hatzivassiliou M, Malakos J, Daskalova D, Hatzigeorgiou G, Cariolou MA, Cokkinos DV. Association of apolipoprotein E polymorphism with myocardial infarction in Greek patients with coronary artery disease. Curr Med Res Opin. 2002;18:118–24.PubMedCrossRef Kolovou G, Yiannakouris N, Hatzivassiliou M, Malakos J, Daskalova D, Hatzigeorgiou G, Cariolou MA, Cokkinos DV. Association of apolipoprotein E polymorphism with myocardial infarction in Greek patients with coronary artery disease. Curr Med Res Opin. 2002;18:118–24.PubMedCrossRef
55.
Zurück zum Zitat Kolovou GD, Anagnostopoulou KK, Mikhailidis DP, Panagiotakos DB, Pilatis ND, Cariolou MA, Yiannakouris N, Degiannis D, Stavridis G, Cokkinos DV. Association of apolipoprotein E genotype with early onset of coronary heart disease in Greek men. Angiology. 2005;56:663–70.PubMedCrossRef Kolovou GD, Anagnostopoulou KK, Mikhailidis DP, Panagiotakos DB, Pilatis ND, Cariolou MA, Yiannakouris N, Degiannis D, Stavridis G, Cokkinos DV. Association of apolipoprotein E genotype with early onset of coronary heart disease in Greek men. Angiology. 2005;56:663–70.PubMedCrossRef
56.
Zurück zum Zitat Corder E, Saunders A, Strittmatter W, Schmechel D, Gaskell P, Small G, Roses A, Haines J, Pericak-Vance MA. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.PubMedCrossRef Corder E, Saunders A, Strittmatter W, Schmechel D, Gaskell P, Small G, Roses A, Haines J, Pericak-Vance MA. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.PubMedCrossRef
57.
Zurück zum Zitat Bretsky P, Guralnik J, Launer L, Albert M, Seeman TE. The role of APOE-ε4 in longitudinal cognitive decline MacArthur studies of successful aging. Neurology. 2003;60:1077–81.PubMedCrossRef Bretsky P, Guralnik J, Launer L, Albert M, Seeman TE. The role of APOE-ε4 in longitudinal cognitive decline MacArthur studies of successful aging. Neurology. 2003;60:1077–81.PubMedCrossRef
58.
Zurück zum Zitat Kolovou GD, Anagnostopoulou KK, Cokkinos DV. Apolipoprotein epsilon4 homozygosity and essential hypertension. South Med J. 1022;2004:97. Kolovou GD, Anagnostopoulou KK, Cokkinos DV. Apolipoprotein epsilon4 homozygosity and essential hypertension. South Med J. 1022;2004:97.
59.
Zurück zum Zitat Meade T, Brozovic M, Chakrabarti R, Haines A, Imeson J, Mellows S, Miller G, North W, Stirling Y, Thompson S. Haemostatic function and ischaemic heart disease: principal results of the Northwick Park heart study. Lancet. 1986;328:533–7.CrossRef Meade T, Brozovic M, Chakrabarti R, Haines A, Imeson J, Mellows S, Miller G, North W, Stirling Y, Thompson S. Haemostatic function and ischaemic heart disease: principal results of the Northwick Park heart study. Lancet. 1986;328:533–7.CrossRef
60.
Zurück zum Zitat Humphries SE, Talmud PJ, Hawe E, Bolla M, Day IN, Miller GJ. Apolipoprotein E4 and coronary heart disease in middle-aged men who smoke: a prospective study. Lancet. 2001;358:115–9.PubMedCrossRef Humphries SE, Talmud PJ, Hawe E, Bolla M, Day IN, Miller GJ. Apolipoprotein E4 and coronary heart disease in middle-aged men who smoke: a prospective study. Lancet. 2001;358:115–9.PubMedCrossRef
61.
Zurück zum Zitat Talmud P, Stephens J, Hawe E, Demissie S, Cupples L, Hurel S, Humphries S, Ordovas J. The significant increase in cardiovascular disease risk in APOEɛ4 carriers is evident only in men who smoke: potential relationship between reduced antioxidant status and APOE4. Ann Hum Genet. 2005;69:613–22.PubMedCrossRef Talmud P, Stephens J, Hawe E, Demissie S, Cupples L, Hurel S, Humphries S, Ordovas J. The significant increase in cardiovascular disease risk in APOEɛ4 carriers is evident only in men who smoke: potential relationship between reduced antioxidant status and APOE4. Ann Hum Genet. 2005;69:613–22.PubMedCrossRef
62.
Zurück zum Zitat Flecknell P. Replacement, reduction and refinement. ALTEX. 2002;19:73–8.PubMed Flecknell P. Replacement, reduction and refinement. ALTEX. 2002;19:73–8.PubMed
63.
Zurück zum Zitat Russell WMS, Burch RL, Hume CW. The principles of humane experimental technique. 1959. Russell WMS, Burch RL, Hume CW. The principles of humane experimental technique. 1959.
64.
Zurück zum Zitat Rothblat GH, Mahlberg FH, Johnson W, Phillips MC. Apolipoproteins, membrane cholesterol domains, and the regulation of cholesterol efflux. J Lipid Res. 1992;33:1091–7.PubMed Rothblat GH, Mahlberg FH, Johnson W, Phillips MC. Apolipoproteins, membrane cholesterol domains, and the regulation of cholesterol efflux. J Lipid Res. 1992;33:1091–7.PubMed
65.
Zurück zum Zitat Andrikoula M, McDowell I. The contribution of ApoB and ApoA1 measurements to cardiovascular risk assessment. Diabetes Obes Metab. 2008;10:271–8.PubMedCrossRef Andrikoula M, McDowell I. The contribution of ApoB and ApoA1 measurements to cardiovascular risk assessment. Diabetes Obes Metab. 2008;10:271–8.PubMedCrossRef
66.
Zurück zum Zitat Borén J, Lee I, Zhu W, Arnold K, Taylor S, Innerarity TL. Identification of the low density lipoprotein receptor-binding site in apolipoprotein B100 and the modulation of its binding activity by the carboxyl terminus in familial defective apo-B100. J Clin Invest. 1084;1998:101. Borén J, Lee I, Zhu W, Arnold K, Taylor S, Innerarity TL. Identification of the low density lipoprotein receptor-binding site in apolipoprotein B100 and the modulation of its binding activity by the carboxyl terminus in familial defective apo-B100. J Clin Invest. 1084;1998:101.
68.
Zurück zum Zitat Goldstein JL, Kita T, Brown MS. Defective lipoprotein receptors and atherosclerosis. Lessons from an animal counterpart of familial hypercholesterolemia. N Engl J Med. 1983;309:288.PubMedCrossRef Goldstein JL, Kita T, Brown MS. Defective lipoprotein receptors and atherosclerosis. Lessons from an animal counterpart of familial hypercholesterolemia. N Engl J Med. 1983;309:288.PubMedCrossRef
70.
Zurück zum Zitat Willnow TE. Mechanisms of hepatic chylomicron remnant clearance. Diabet Med. 1997;14(Suppl 3):S75–80.PubMedCrossRef Willnow TE. Mechanisms of hepatic chylomicron remnant clearance. Diabet Med. 1997;14(Suppl 3):S75–80.PubMedCrossRef
71.
Zurück zum Zitat Lillis AP, Van Duyn LB, Murphy-Ullrich JE, Strickland DK. LDL receptor-related protein 1: unique tissue-specific functions revealed by selective gene knockout studies. Physiol Rev. 2008;88:887–918.PubMedPubMedCentralCrossRef Lillis AP, Van Duyn LB, Murphy-Ullrich JE, Strickland DK. LDL receptor-related protein 1: unique tissue-specific functions revealed by selective gene knockout studies. Physiol Rev. 2008;88:887–918.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Shiomi M, Ishida T, Koike T. Genetically modified animal models for lipoprotein research. Rijeka: INTECH Open Access Publisher; 2012.CrossRef Shiomi M, Ishida T, Koike T. Genetically modified animal models for lipoprotein research. Rijeka: INTECH Open Access Publisher; 2012.CrossRef
73.
Zurück zum Zitat Olofsson SO, Borén J. Apolipoprotein B secretory regulation by degradation. Arterioscler Thromb Vasc Biol. 2012;32:1334–8.PubMedCrossRef Olofsson SO, Borén J. Apolipoprotein B secretory regulation by degradation. Arterioscler Thromb Vasc Biol. 2012;32:1334–8.PubMedCrossRef
74.
Zurück zum Zitat Greeve J, Altkemper I, Dieterich JH, Greten H, Windler E. Apolipoprotein B mRNA editing in 12 different mammalian species: hepatic expression is reflected in low concentrations of apoB-containing plasma lipoproteins. J Lipid Res. 1993;34:1367–83.PubMed Greeve J, Altkemper I, Dieterich JH, Greten H, Windler E. Apolipoprotein B mRNA editing in 12 different mammalian species: hepatic expression is reflected in low concentrations of apoB-containing plasma lipoproteins. J Lipid Res. 1993;34:1367–83.PubMed
75.
Zurück zum Zitat Takahashi S, Sakai J, Fujino T, Hattori H, Zenimaru Y, Suzuki J, Miyamori I, Yamamoto TT. The very low-density lipoprotein (VLDL) receptor: characterization and functions as a peripheral lipoprotein receptor. J Atheroscler Thromb. 2004;11:200–8.PubMedCrossRef Takahashi S, Sakai J, Fujino T, Hattori H, Zenimaru Y, Suzuki J, Miyamori I, Yamamoto TT. The very low-density lipoprotein (VLDL) receptor: characterization and functions as a peripheral lipoprotein receptor. J Atheroscler Thromb. 2004;11:200–8.PubMedCrossRef
76.
Zurück zum Zitat Li X, Catalina F, Grundy S, Patel S. Method to measure apolipoprotein B-48 and B-100 secretion rates in an individual mouse: evidence for a very rapid turnover of VLDL and preferential removal of B-48-relative to B-100-containing lipoproteins. J Lipid Res. 1996;37:210–20.PubMed Li X, Catalina F, Grundy S, Patel S. Method to measure apolipoprotein B-48 and B-100 secretion rates in an individual mouse: evidence for a very rapid turnover of VLDL and preferential removal of B-48-relative to B-100-containing lipoproteins. J Lipid Res. 1996;37:210–20.PubMed
77.
Zurück zum Zitat Agellon L, Walsh A, Hayek T, Moulin P, Jiang XC, Shelanski SA, Breslow J, Tall AR. Reduced high density lipoprotein cholesterol in human cholesteryl ester transfer protein transgenic mice. J Biol Chem. 1991;266:10796–801.PubMed Agellon L, Walsh A, Hayek T, Moulin P, Jiang XC, Shelanski SA, Breslow J, Tall AR. Reduced high density lipoprotein cholesterol in human cholesteryl ester transfer protein transgenic mice. J Biol Chem. 1991;266:10796–801.PubMed
78.
Zurück zum Zitat Rohrer L, Ohnsorg PM, Lehner M, Landolt F, Rinninger F, von Eckardstein A. High-density lipoprotein transport through aortic endothelial cells involves scavenger receptor BI and ATP-binding cassette transporter G1. Circ Res. 2009;104:1142–50.PubMedCrossRef Rohrer L, Ohnsorg PM, Lehner M, Landolt F, Rinninger F, von Eckardstein A. High-density lipoprotein transport through aortic endothelial cells involves scavenger receptor BI and ATP-binding cassette transporter G1. Circ Res. 2009;104:1142–50.PubMedCrossRef
79.
Zurück zum Zitat Son Y, Zilversmit D. Increased lipid transfer activities in hyperlipidemic rabbit plasma. Arterioscler Thromb Vasc Biol. 1986;6:345–51.CrossRef Son Y, Zilversmit D. Increased lipid transfer activities in hyperlipidemic rabbit plasma. Arterioscler Thromb Vasc Biol. 1986;6:345–51.CrossRef
80.
Zurück zum Zitat Gairola CG, Drawdy ML, Block AE, Daugherty A. Sidestream cigarette smoke accelerates atherogenesis in apolipoprotein E−/− mice. Atherosclerosis. 2001;156:49–55.PubMedCrossRef Gairola CG, Drawdy ML, Block AE, Daugherty A. Sidestream cigarette smoke accelerates atherogenesis in apolipoprotein E−/− mice. Atherosclerosis. 2001;156:49–55.PubMedCrossRef
81.
Zurück zum Zitat Gairola CG, Howatt DA, Daugherty A. Dietary coenzyme Q10 does not protect against cigarette smoke-augmented atherosclerosis in apoE-deficient mice. Free Radic Biol Med. 2010;48:1535–9.PubMedCrossRef Gairola CG, Howatt DA, Daugherty A. Dietary coenzyme Q10 does not protect against cigarette smoke-augmented atherosclerosis in apoE-deficient mice. Free Radic Biol Med. 2010;48:1535–9.PubMedCrossRef
82.
Zurück zum Zitat Takagi H, Umemoto T. Smoking promotes pathogenesis of aortic aneurysm through the 5-lipoxygenase pathway. Med Hypotheses. 2005;64:1117–9.PubMedCrossRef Takagi H, Umemoto T. Smoking promotes pathogenesis of aortic aneurysm through the 5-lipoxygenase pathway. Med Hypotheses. 2005;64:1117–9.PubMedCrossRef
83.
Zurück zum Zitat Stolle K, Berges A, Lietz M, Lebrun S, Wallerath T. Cigarette smoke enhances abdominal aortic aneurysm formation in angiotensin II-treated apolipoprotein E-deficient mice. Toxicol Lett. 2010;199:403–9.PubMedCrossRef Stolle K, Berges A, Lietz M, Lebrun S, Wallerath T. Cigarette smoke enhances abdominal aortic aneurysm formation in angiotensin II-treated apolipoprotein E-deficient mice. Toxicol Lett. 2010;199:403–9.PubMedCrossRef
84.
Zurück zum Zitat Matetzky S, Tani S, Kangavari S, Dimayuga P, Yano J, Xu H, Chyu KY, Fishbein MC, Shah PK, Cercek B. Smoking increases tissue factor expression in atherosclerotic plaques: implications for plaque thrombogenicity. Circulation. 2000;102:602–4.PubMedCrossRef Matetzky S, Tani S, Kangavari S, Dimayuga P, Yano J, Xu H, Chyu KY, Fishbein MC, Shah PK, Cercek B. Smoking increases tissue factor expression in atherosclerotic plaques: implications for plaque thrombogenicity. Circulation. 2000;102:602–4.PubMedCrossRef
85.
Zurück zum Zitat Dong A, Caicedo J, Han SG, Mueller P, Saha S, Smyth SS, Gairola CG. Enhanced platelet reactivity and thrombosis in Apoe−/− mice exposed to cigarette smoke is attenuated by P2Y12 antagonism. Thromb Res. 2010;126:e312–7.PubMedPubMedCentralCrossRef Dong A, Caicedo J, Han SG, Mueller P, Saha S, Smyth SS, Gairola CG. Enhanced platelet reactivity and thrombosis in Apoe−/− mice exposed to cigarette smoke is attenuated by P2Y12 antagonism. Thromb Res. 2010;126:e312–7.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Bliden KP, Dichiara J, Lawal L, Singla A, Antonino MJ, Baker BA, Bailey WL, Tantry US, Gurbel PA. The association of cigarette smoking with enhanced platelet inhibition by clopidogrel. J Am Coll Cardiol. 2008;52:531–3.PubMedCrossRef Bliden KP, Dichiara J, Lawal L, Singla A, Antonino MJ, Baker BA, Bailey WL, Tantry US, Gurbel PA. The association of cigarette smoking with enhanced platelet inhibition by clopidogrel. J Am Coll Cardiol. 2008;52:531–3.PubMedCrossRef
87.
Zurück zum Zitat Barbieri SS, Weksler BB. Tobacco smoke cooperates with interleukin-1beta to alter beta-catenin trafficking in vascular endothelium resulting in increased permeability and induction of cyclooxygenase-2 expression in vitro and in vivo. FASEB J. 2007;21:1831–43.PubMedCrossRef Barbieri SS, Weksler BB. Tobacco smoke cooperates with interleukin-1beta to alter beta-catenin trafficking in vascular endothelium resulting in increased permeability and induction of cyclooxygenase-2 expression in vitro and in vivo. FASEB J. 2007;21:1831–43.PubMedCrossRef
88.
Zurück zum Zitat Barbieri SS, Ruggiero L, Tremoli E, Weksler BB. Suppressing PTEN activity by tobacco smoke plus interleukin-1beta modulates dissociation of VE-cadherin/beta-catenin complexes in endothelium. Arterioscler Thromb Vasc Biol. 2008;28:732–8.PubMedCrossRef Barbieri SS, Ruggiero L, Tremoli E, Weksler BB. Suppressing PTEN activity by tobacco smoke plus interleukin-1beta modulates dissociation of VE-cadherin/beta-catenin complexes in endothelium. Arterioscler Thromb Vasc Biol. 2008;28:732–8.PubMedCrossRef
89.
Zurück zum Zitat Tani S, Dimayuga PC, Anazawa T, Chyu KY, Li H, Shah PK, Cercek B. Aberrant antibody responses to oxidized LDL and increased intimal thickening in apoE−/− mice exposed to cigarette smoke. Atherosclerosis. 2004;175:7–14.PubMedCrossRef Tani S, Dimayuga PC, Anazawa T, Chyu KY, Li H, Shah PK, Cercek B. Aberrant antibody responses to oxidized LDL and increased intimal thickening in apoE−/− mice exposed to cigarette smoke. Atherosclerosis. 2004;175:7–14.PubMedCrossRef
90.
Zurück zum Zitat Kunitomo M, Yamaguchi Y, Kagota S, Yoshikawa N, Nakamura K, Shinozuka K. Biochemical evidence of atherosclerosis progression mediated by increased oxidative stress in apolipoprotein E-deficient spontaneously hyperlipidemic mice exposed to chronic cigarette smoke. J Pharmacol Sci. 2009;110:354–61.PubMedCrossRef Kunitomo M, Yamaguchi Y, Kagota S, Yoshikawa N, Nakamura K, Shinozuka K. Biochemical evidence of atherosclerosis progression mediated by increased oxidative stress in apolipoprotein E-deficient spontaneously hyperlipidemic mice exposed to chronic cigarette smoke. J Pharmacol Sci. 2009;110:354–61.PubMedCrossRef
91.
Zurück zum Zitat Harrison CM, Pompilius M, Pinkerton KE, Ballinger SW. Mitochondrial oxidative stress significantly influences atherogenic risk and cytokine-induced oxidant production. Environ Health Perspect. 2011;119:676–81.PubMedPubMedCentralCrossRef Harrison CM, Pompilius M, Pinkerton KE, Ballinger SW. Mitochondrial oxidative stress significantly influences atherogenic risk and cytokine-induced oxidant production. Environ Health Perspect. 2011;119:676–81.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Seilkop SK, Campen MJ, Lund AK, McDonald JD, Mauderly JL. Identification of chemical components of combustion emissions that affect pro-atherosclerotic vascular responses in mice. Inhal Toxicol. 2012;24:270–87.PubMedPubMedCentralCrossRef Seilkop SK, Campen MJ, Lund AK, McDonald JD, Mauderly JL. Identification of chemical components of combustion emissions that affect pro-atherosclerotic vascular responses in mice. Inhal Toxicol. 2012;24:270–87.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Chen LC, Quan C, Hwang JS, Jin X, Li Q, Zhong M, Rajagopalan S, Sun Q. Atherosclerosis lesion progression during inhalation exposure to environmental tobacco smoke: a comparison to concentrated ambient air fine particles exposure. Inhal Toxicol. 2010;22:449–59.PubMedPubMedCentralCrossRef Chen LC, Quan C, Hwang JS, Jin X, Li Q, Zhong M, Rajagopalan S, Sun Q. Atherosclerosis lesion progression during inhalation exposure to environmental tobacco smoke: a comparison to concentrated ambient air fine particles exposure. Inhal Toxicol. 2010;22:449–59.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat De Leon H, Boue S, Szostak J, Peitsch MC, Hoeng J. Systems biology research into cardiovascular disease: contributions of lipidomics-based approaches to biomarker discovery. Curr Drug Discov Technol. 2015;12:129–54.PubMedCrossRef De Leon H, Boue S, Szostak J, Peitsch MC, Hoeng J. Systems biology research into cardiovascular disease: contributions of lipidomics-based approaches to biomarker discovery. Curr Drug Discov Technol. 2015;12:129–54.PubMedCrossRef
95.
Zurück zum Zitat Rodgman A, Perfetti TA. The chemical components of tobacco and tobacco smoke. Boca Raton: CRC Press; 2013.CrossRef Rodgman A, Perfetti TA. The chemical components of tobacco and tobacco smoke. Boca Raton: CRC Press; 2013.CrossRef
96.
Zurück zum Zitat Talhout R, Schulz T, Florek E, Van Benthem J, Wester P, Opperhuizen A. Hazardous compounds in tobacco smoke. Int J Environ Res Public Health. 2011;8:613–28.PubMedPubMedCentralCrossRef Talhout R, Schulz T, Florek E, Van Benthem J, Wester P, Opperhuizen A. Hazardous compounds in tobacco smoke. Int J Environ Res Public Health. 2011;8:613–28.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Sweanor D, Alcabes P, Drucker E. Tobacco harm reduction: how rational public policy could transform a pandemic. Int J Drug Policy. 2007;18:70–4.PubMedCrossRef Sweanor D, Alcabes P, Drucker E. Tobacco harm reduction: how rational public policy could transform a pandemic. Int J Drug Policy. 2007;18:70–4.PubMedCrossRef
98.
Zurück zum Zitat World Health Organization. The scientific basis of tobacco product regulation. Geneva: World Health Organization; 2007. World Health Organization. The scientific basis of tobacco product regulation. Geneva: World Health Organization; 2007.
99.
Zurück zum Zitat Surgeons RCoPa. Harm reduction in nicotine addiction: helping people who can’t quit. London: Tobacco Advisory Group; 2007. Surgeons RCoPa. Harm reduction in nicotine addiction: helping people who can’t quit. London: Tobacco Advisory Group; 2007.
100.
Zurück zum Zitat Zeller M, Hatsukami D. The strategic dialogue on tobacco harm reduction: a vision and blueprint for action in the US. Tob Control. 2009;18:324–32.PubMedCrossRef Zeller M, Hatsukami D. The strategic dialogue on tobacco harm reduction: a vision and blueprint for action in the US. Tob Control. 2009;18:324–32.PubMedCrossRef
101.
Zurück zum Zitat Bakhru A, Erlinger TP. Smoking cessation and cardiovascular disease risk factors: results from the third national health and nutrition examination survey. PLoS Med. 2005;2:528.CrossRef Bakhru A, Erlinger TP. Smoking cessation and cardiovascular disease risk factors: results from the third national health and nutrition examination survey. PLoS Med. 2005;2:528.CrossRef
102.
Zurück zum Zitat Godtfredsen N, Lam T, Hansel T, Leon M, Gray N, Dresler C, Burns D, Prescott E, Vestbo J. COPD-related morbidity and mortality after smoking cessation: status of the evidence. Eur Respir J. 2008;32:844–53.PubMedCrossRef Godtfredsen N, Lam T, Hansel T, Leon M, Gray N, Dresler C, Burns D, Prescott E, Vestbo J. COPD-related morbidity and mortality after smoking cessation: status of the evidence. Eur Respir J. 2008;32:844–53.PubMedCrossRef
103.
Zurück zum Zitat Gepner AD, Piper ME, Johnson HM, Fiore MC, Baker TB, Stein JH. Effects of smoking and smoking cessation on lipids and lipoproteins: outcomes from a randomized clinical trial. Am Heart J. 2011;161:145–51.PubMedPubMedCentralCrossRef Gepner AD, Piper ME, Johnson HM, Fiore MC, Baker TB, Stein JH. Effects of smoking and smoking cessation on lipids and lipoproteins: outcomes from a randomized clinical trial. Am Heart J. 2011;161:145–51.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Hughes JR, Keely J, Naud S. Shape of the relapse curve and long-term abstinence among untreated smokers. Addiction. 2004;99:29–38.PubMedCrossRef Hughes JR, Keely J, Naud S. Shape of the relapse curve and long-term abstinence among untreated smokers. Addiction. 2004;99:29–38.PubMedCrossRef
105.
Zurück zum Zitat McRobbie H, Bullen C, Hartmann-Boyce J, Hajek P. Electronic cigarettes for smoking cessation and reduction. Cochrane Database Syst Rev. 2014;12:CD010216.PubMed McRobbie H, Bullen C, Hartmann-Boyce J, Hajek P. Electronic cigarettes for smoking cessation and reduction. Cochrane Database Syst Rev. 2014;12:CD010216.PubMed
106.
Zurück zum Zitat Hoffmann D, Rathkamp G. Quantitative determination of fluorenes in cigarette smoke and their formation by pyrosynthesis. Anal Chem. 1972;44:899–905.PubMedCrossRef Hoffmann D, Rathkamp G. Quantitative determination of fluorenes in cigarette smoke and their formation by pyrosynthesis. Anal Chem. 1972;44:899–905.PubMedCrossRef
107.
Zurück zum Zitat Brunnemann KD, Djordjevic MV, Feng R, Hoffmann D. Analysis and pyrolysis of some N-nitrosamino acids in tobacco and tobacco smoke. IARC Sci Publ. 1990:105;477–81. Brunnemann KD, Djordjevic MV, Feng R, Hoffmann D. Analysis and pyrolysis of some N-nitrosamino acids in tobacco and tobacco smoke. IARC Sci Publ. 1990:105;477–81.
108.
Zurück zum Zitat Hertz-Schunemann R, Ehlert S, Streibel T, Liu C, McAdam K, Baker RR, Zimmermann R. High-resolution time and spatial imaging of tobacco and its pyrolysis products during a cigarette puff by microprobe sampling photoionisation mass spectrometry. Anal Bioanal Chem. 2015;407:2293–9.PubMedCrossRef Hertz-Schunemann R, Ehlert S, Streibel T, Liu C, McAdam K, Baker RR, Zimmermann R. High-resolution time and spatial imaging of tobacco and its pyrolysis products during a cigarette puff by microprobe sampling photoionisation mass spectrometry. Anal Bioanal Chem. 2015;407:2293–9.PubMedCrossRef
109.
Zurück zum Zitat Baker RR. Smoke generation inside a burning cigarette: modifying combustion to develop cigarettes that may be less hazardous to health. Prog Energy Combust Sci. 2006;32:373–85.CrossRef Baker RR. Smoke generation inside a burning cigarette: modifying combustion to develop cigarettes that may be less hazardous to health. Prog Energy Combust Sci. 2006;32:373–85.CrossRef
110.
Zurück zum Zitat Borgerding MF, Bodnar JA, Chung HL, Mangan PP, Morrison CC, Risner CH, Rogers JC, Simmons DF, Uhrig MS, Wendelboe FN, et al. Chemical and biological studies of a new cigarette that primarily heats tobacco. Part 1. Chemical composition of mainstream smoke. Food Chem Toxicol. 1998;36:169–82.CrossRef Borgerding MF, Bodnar JA, Chung HL, Mangan PP, Morrison CC, Risner CH, Rogers JC, Simmons DF, Uhrig MS, Wendelboe FN, et al. Chemical and biological studies of a new cigarette that primarily heats tobacco. Part 1. Chemical composition of mainstream smoke. Food Chem Toxicol. 1998;36:169–82.CrossRef
111.
Zurück zum Zitat Hoeng J, Deehan R, Pratt D, Martin F, Sewer A, Thomson TM, Drubin DA, Waters CA, de Graaf D, Peitsch MC. A network-based approach to quantifying the impact of biologically active substances. Drug Discov Today. 2012;17:413–8.PubMedCrossRef Hoeng J, Deehan R, Pratt D, Martin F, Sewer A, Thomson TM, Drubin DA, Waters CA, de Graaf D, Peitsch MC. A network-based approach to quantifying the impact of biologically active substances. Drug Discov Today. 2012;17:413–8.PubMedCrossRef
112.
Zurück zum Zitat Sturla SJ, Boobis AR, FitzGerald RE, Hoeng J, Kavlock RJ, Schirmer K, Whelan M, Wilks MF, Peitsch MC. Systems toxicology: from basic research to risk assessment. Chem Res Toxicol. 2014;27:314–29.PubMedPubMedCentralCrossRef Sturla SJ, Boobis AR, FitzGerald RE, Hoeng J, Kavlock RJ, Schirmer K, Whelan M, Wilks MF, Peitsch MC. Systems toxicology: from basic research to risk assessment. Chem Res Toxicol. 2014;27:314–29.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Titz B, Boue S, Phillips B, Talikka M, Vihervaara T, Schneider T, Nury C, Elamin A, Guedj E, Peck MJ, et al. Effects of cigarette smoke, cessation, and switching to two heat-not-burn tobacco products on lung lipid metabolism in C57BL/6 and Apoe−/− mice-An integrative systems toxicology analysis. Toxicol Sci. 2016;149:441–57.PubMedPubMedCentralCrossRef Titz B, Boue S, Phillips B, Talikka M, Vihervaara T, Schneider T, Nury C, Elamin A, Guedj E, Peck MJ, et al. Effects of cigarette smoke, cessation, and switching to two heat-not-burn tobacco products on lung lipid metabolism in C57BL/6 and Apoe−/− mice-An integrative systems toxicology analysis. Toxicol Sci. 2016;149:441–57.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Lo Sasso G, Titz B, Nury C, Boue S, Phillips B, Belcastro V, Schneider T, Dijon S, Baumer K, Peric D, et al. Effects of cigarette smoke, cessation and switching to a candidate modified risk tobacco product on the liver in Apoe mice—a systems toxicology analysis. Inhal Toxicol. 2016;28:1–15.CrossRef Lo Sasso G, Titz B, Nury C, Boue S, Phillips B, Belcastro V, Schneider T, Dijon S, Baumer K, Peric D, et al. Effects of cigarette smoke, cessation and switching to a candidate modified risk tobacco product on the liver in Apoe mice—a systems toxicology analysis. Inhal Toxicol. 2016;28:1–15.CrossRef
115.
Zurück zum Zitat Szostak J, Boué S, Talikka M, Guedj E, Martin F, Phillips B, Ivanov N, Peitsch MC, Hoeng J. Long term exposure to cigarette smoke impacts the expression of genes involved in cardiac muscle structure and function in Apoe−/− mouse while exposure to the aerosol of a tobacco heating system does not. BMC Med Genomics. 2015. (personal communication) Szostak J, Boué S, Talikka M, Guedj E, Martin F, Phillips B, Ivanov N, Peitsch MC, Hoeng J. Long term exposure to cigarette smoke impacts the expression of genes involved in cardiac muscle structure and function in Apoe−/− mouse while exposure to the aerosol of a tobacco heating system does not. BMC Med Genomics. 2015. (personal communication)
116.
Zurück zum Zitat Boué S, Tarasov K, Jänis M, Lebrun S, Hurme R, Schlage W, Lietz M, Vuillaume G, Ekroos K, Steffen Y. Modulation of atherogenic lipidome by cigarette smoke in apolipoprotein E-deficient mice. Atherosclerosis. 2012;225:328–34.PubMedCrossRef Boué S, Tarasov K, Jänis M, Lebrun S, Hurme R, Schlage W, Lietz M, Vuillaume G, Ekroos K, Steffen Y. Modulation of atherogenic lipidome by cigarette smoke in apolipoprotein E-deficient mice. Atherosclerosis. 2012;225:328–34.PubMedCrossRef
117.
Zurück zum Zitat Lietz M, Berges A, Lebrun S, Meurrens K, Steffen Y, Stolle K, Schueller J, Boue S, Vuillaume G, Vanscheeuwijck P. Cigarette-smoke-induced atherogenic lipid profiles in plasma and vascular tissue of apolipoprotein E-deficient mice are attenuated by smoking cessation. Atherosclerosis. 2013;229:86–93.PubMedCrossRef Lietz M, Berges A, Lebrun S, Meurrens K, Steffen Y, Stolle K, Schueller J, Boue S, Vuillaume G, Vanscheeuwijck P. Cigarette-smoke-induced atherogenic lipid profiles in plasma and vascular tissue of apolipoprotein E-deficient mice are attenuated by smoking cessation. Atherosclerosis. 2013;229:86–93.PubMedCrossRef
118.
Zurück zum Zitat De León H, Boué S, Peitsch M, Hoeng J. Modulation of the hepatic lipidome and transcriptome of Apoe−/−mice in response to smoking cessation. J Liver. 2013;2(132):2167. De León H, Boué S, Peitsch M, Hoeng J. Modulation of the hepatic lipidome and transcriptome of Apoe−/−mice in response to smoking cessation. J Liver. 2013;2(132):2167.
119.
Zurück zum Zitat Wang L, Chen L, Tan Y, Wei J, Chang Y, Jin T, Zhu H. Betaine supplement alleviates hepatic triglyceride accumulation of apolipoprotein E deficient mice via reducing methylation of peroxisomal proliferator-activated receptor alpha promoter. Lipids Health Dis. 2013;12:34.PubMedPubMedCentralCrossRef Wang L, Chen L, Tan Y, Wei J, Chang Y, Jin T, Zhu H. Betaine supplement alleviates hepatic triglyceride accumulation of apolipoprotein E deficient mice via reducing methylation of peroxisomal proliferator-activated receptor alpha promoter. Lipids Health Dis. 2013;12:34.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Mensenkamp AR, van Luyn MJ, Havinga R, Teusink B, Waterman IJ, Mann CJ, Elzinga BM, Verkade HJ, Zammit VA, Havekes LM. The transport of triglycerides through the secretory pathway of hepatocytes is impaired in apolipoprotein E deficient mice. J Hepatol. 2004;40:599–606.PubMedCrossRef Mensenkamp AR, van Luyn MJ, Havinga R, Teusink B, Waterman IJ, Mann CJ, Elzinga BM, Verkade HJ, Zammit VA, Havekes LM. The transport of triglycerides through the secretory pathway of hepatocytes is impaired in apolipoprotein E deficient mice. J Hepatol. 2004;40:599–606.PubMedCrossRef
121.
Zurück zum Zitat Whitehead T, Robinson D, Allaway S. The effects of cigarette smoking and alcohol consumption on serum liver enzyme activities: a dose-related study in men. Ann Clin Biochem. 1996;33:530–5.PubMedCrossRef Whitehead T, Robinson D, Allaway S. The effects of cigarette smoking and alcohol consumption on serum liver enzyme activities: a dose-related study in men. Ann Clin Biochem. 1996;33:530–5.PubMedCrossRef
122.
Zurück zum Zitat Azzalini L, Ferrer E, Ramalho LN, Moreno M, Domínguez M, Colmenero J, Peinado VI, Barbera JA, Arroyo V, Gines P. Cigarette smoking exacerbates nonalcoholic fatty liver disease in obese rats. Hepatology. 2010;51:1567–76.PubMedCrossRef Azzalini L, Ferrer E, Ramalho LN, Moreno M, Domínguez M, Colmenero J, Peinado VI, Barbera JA, Arroyo V, Gines P. Cigarette smoking exacerbates nonalcoholic fatty liver disease in obese rats. Hepatology. 2010;51:1567–76.PubMedCrossRef
123.
Zurück zum Zitat Altamirano J, Bataller R. Cigarette smoking and chronic liver diseases. Gut. 2010;59:1159–62.PubMedCrossRef Altamirano J, Bataller R. Cigarette smoking and chronic liver diseases. Gut. 2010;59:1159–62.PubMedCrossRef
124.
Zurück zum Zitat Thomson TM, Sewer A, Martin F, Belcastro V, Frushour BP, Gebel S, Park J, Schlage WK, Talikka M, Vasilyev DM, et al. Quantitative assessment of biological impact using transcriptomic data and mechanistic network models. Toxicol Appl Pharmacol. 2013;272:863–78.PubMedCrossRef Thomson TM, Sewer A, Martin F, Belcastro V, Frushour BP, Gebel S, Park J, Schlage WK, Talikka M, Vasilyev DM, et al. Quantitative assessment of biological impact using transcriptomic data and mechanistic network models. Toxicol Appl Pharmacol. 2013;272:863–78.PubMedCrossRef
125.
Zurück zum Zitat Catlett NL, Bargnesi AJ, Ungerer S, Seagaran T, Ladd W, Elliston KO, Pratt D. Reverse causal reasoning: applying qualitative causal knowledge to the interpretation of high-throughput data. BMC Bioinformatics. 2013;14:340.PubMedPubMedCentralCrossRef Catlett NL, Bargnesi AJ, Ungerer S, Seagaran T, Ladd W, Elliston KO, Pratt D. Reverse causal reasoning: applying qualitative causal knowledge to the interpretation of high-throughput data. BMC Bioinformatics. 2013;14:340.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Stegemann C, Drozdov I, Shalhoub J, Humphries J, Ladroue C, Didangelos A, Baumert M, Allen M, Davies AH, Monaco C, et al. Comparative lipidomics profiling of human atherosclerotic plaques. Circ Cardiovasc Genet. 2011;4:232–42.PubMedCrossRef Stegemann C, Drozdov I, Shalhoub J, Humphries J, Ladroue C, Didangelos A, Baumert M, Allen M, Davies AH, Monaco C, et al. Comparative lipidomics profiling of human atherosclerotic plaques. Circ Cardiovasc Genet. 2011;4:232–42.PubMedCrossRef
127.
Zurück zum Zitat Cabanski M, Fields B, Boue S, Boukharov N, DeLeon H, Dror N, Geertz M, Guedj E, Iskandar A, Kogel U, et al. Transcriptional profiling and targeted proteomics reveals common molecular changes associated with cigarette smoke-induced lung emphysema development in five susceptible mouse strains. Inflamm Res. 2015;64:471–86.PubMedPubMedCentralCrossRef Cabanski M, Fields B, Boue S, Boukharov N, DeLeon H, Dror N, Geertz M, Guedj E, Iskandar A, Kogel U, et al. Transcriptional profiling and targeted proteomics reveals common molecular changes associated with cigarette smoke-induced lung emphysema development in five susceptible mouse strains. Inflamm Res. 2015;64:471–86.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Hansen CS, Sheykhzade M, Moller P, Folkmann JK, Amtorp O, Jonassen T, Loft S. Diesel exhaust particles induce endothelial dysfunction in apoE−/− mice. Toxicol Appl Pharmacol. 2007;219:24–32.PubMedCrossRef Hansen CS, Sheykhzade M, Moller P, Folkmann JK, Amtorp O, Jonassen T, Loft S. Diesel exhaust particles induce endothelial dysfunction in apoE−/− mice. Toxicol Appl Pharmacol. 2007;219:24–32.PubMedCrossRef
129.
Zurück zum Zitat Folkmann JK, Risom L, Hansen CS, Loft S, Moller P. Oxidatively damaged DNA and inflammation in the liver of dyslipidemic ApoE−/− mice exposed to diesel exhaust particles. Toxicology. 2007;237:134–44.PubMedCrossRef Folkmann JK, Risom L, Hansen CS, Loft S, Moller P. Oxidatively damaged DNA and inflammation in the liver of dyslipidemic ApoE−/− mice exposed to diesel exhaust particles. Toxicology. 2007;237:134–44.PubMedCrossRef
130.
Zurück zum Zitat Cassee FR, Campbell A, Boere AJ, McLean SG, Duffin R, Krystek P, Gosens I, Miller MR. The biological effects of subacute inhalation of diesel exhaust following addition of cerium oxide nanoparticles in atherosclerosis-prone mice. Environ Res. 2012;115:1–10.PubMedPubMedCentralCrossRef Cassee FR, Campbell A, Boere AJ, McLean SG, Duffin R, Krystek P, Gosens I, Miller MR. The biological effects of subacute inhalation of diesel exhaust following addition of cerium oxide nanoparticles in atherosclerosis-prone mice. Environ Res. 2012;115:1–10.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Poss J, Lorenz D, Werner C, Pavlikova V, Gensch C, Speer T, Alessandrini F, Berezowski V, Kuntz M, Mempel M, et al. Diesel exhaust particles impair endothelial progenitor cells, compromise endothelial integrity, reduce neoangiogenesis, and increase atherogenesis in mice. Cardiovasc Toxicol. 2013;13:290–300.PubMedCrossRef Poss J, Lorenz D, Werner C, Pavlikova V, Gensch C, Speer T, Alessandrini F, Berezowski V, Kuntz M, Mempel M, et al. Diesel exhaust particles impair endothelial progenitor cells, compromise endothelial integrity, reduce neoangiogenesis, and increase atherogenesis in mice. Cardiovasc Toxicol. 2013;13:290–300.PubMedCrossRef
132.
Zurück zum Zitat Miller MR, McLean SG, Duffin R, Lawal AO, Araujo JA, Shaw CA, Mills NL, Donaldson K, Newby DE, Hadoke PW. Diesel exhaust particulate increases the size and complexity of lesions in atherosclerotic mice. Part Fibre Toxicol. 2013;10:61.PubMedPubMedCentralCrossRef Miller MR, McLean SG, Duffin R, Lawal AO, Araujo JA, Shaw CA, Mills NL, Donaldson K, Newby DE, Hadoke PW. Diesel exhaust particulate increases the size and complexity of lesions in atherosclerotic mice. Part Fibre Toxicol. 2013;10:61.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Lund AK, Knuckles TL, Obot Akata C, Shohet R, McDonald JD, Gigliotti A, Seagrave JC, Campen MJ. Gasoline exhaust emissions induce vascular remodeling pathways involved in atherosclerosis. Toxicol Sci. 2007;95:485–94.PubMedCrossRef Lund AK, Knuckles TL, Obot Akata C, Shohet R, McDonald JD, Gigliotti A, Seagrave JC, Campen MJ. Gasoline exhaust emissions induce vascular remodeling pathways involved in atherosclerosis. Toxicol Sci. 2007;95:485–94.PubMedCrossRef
134.
Zurück zum Zitat Campen MJ, Lund AK, Knuckles TL, Conklin DJ, Bishop B, Young D, Seilkop S, Seagrave J, Reed MD, McDonald JD. Inhaled diesel emissions alter atherosclerotic plaque composition in ApoE(−/−) mice. Toxicol Appl Pharmacol. 2010;242:310–7.PubMedPubMedCentralCrossRef Campen MJ, Lund AK, Knuckles TL, Conklin DJ, Bishop B, Young D, Seilkop S, Seagrave J, Reed MD, McDonald JD. Inhaled diesel emissions alter atherosclerotic plaque composition in ApoE(−/−) mice. Toxicol Appl Pharmacol. 2010;242:310–7.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Bai N, Kido T, Suzuki H, Yang G, Kavanagh TJ, Kaufman JD, Rosenfeld ME, van Breemen C, Eeden SF. Changes in atherosclerotic plaques induced by inhalation of diesel exhaust. Atherosclerosis. 2011;216:299–306.PubMedPubMedCentralCrossRef Bai N, Kido T, Suzuki H, Yang G, Kavanagh TJ, Kaufman JD, Rosenfeld ME, van Breemen C, Eeden SF. Changes in atherosclerotic plaques induced by inhalation of diesel exhaust. Atherosclerosis. 2011;216:299–306.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Bai N, Kido T, Kavanagh TJ, Kaufman JD, Rosenfeld ME, van Breemen C, van Eeden SF. Exposure to diesel exhaust up-regulates iNOS expression in ApoE knockout mice. Toxicol Appl Pharmacol. 2011;255:184–92.PubMedPubMedCentralCrossRef Bai N, Kido T, Kavanagh TJ, Kaufman JD, Rosenfeld ME, van Breemen C, van Eeden SF. Exposure to diesel exhaust up-regulates iNOS expression in ApoE knockout mice. Toxicol Appl Pharmacol. 2011;255:184–92.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Bai N, Tranfield EM, Kavanagh TJ, Kaufman JD, Rosenfeld ME, van Eeden SF. Exposure to diesel exhaust upregulates COX-2 expression in ApoE knockout mice. Inhal Toxicol. 2012;24:518–27.PubMedPubMedCentralCrossRef Bai N, Tranfield EM, Kavanagh TJ, Kaufman JD, Rosenfeld ME, van Eeden SF. Exposure to diesel exhaust upregulates COX-2 expression in ApoE knockout mice. Inhal Toxicol. 2012;24:518–27.PubMedPubMedCentralCrossRef
138.
Zurück zum Zitat Campen M, Robertson S, Lund A, Lucero J, McDonald J. Engine exhaust particulate and gas phase contributions to vascular toxicity. Inhal Toxicol. 2014;26:353–60.PubMedPubMedCentralCrossRef Campen M, Robertson S, Lund A, Lucero J, McDonald J. Engine exhaust particulate and gas phase contributions to vascular toxicity. Inhal Toxicol. 2014;26:353–60.PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Vesterdal LK, Folkmann JK, Jacobsen NR, Sheykhzade M, Wallin H, Loft S, Moller P. Modest vasomotor dysfunction induced by low doses of C60 fullerenes in apolipoprotein E knockout mice with different degree of atherosclerosis. Part Fibre Toxicol. 2009;6:5.PubMedPubMedCentralCrossRef Vesterdal LK, Folkmann JK, Jacobsen NR, Sheykhzade M, Wallin H, Loft S, Moller P. Modest vasomotor dysfunction induced by low doses of C60 fullerenes in apolipoprotein E knockout mice with different degree of atherosclerosis. Part Fibre Toxicol. 2009;6:5.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Ross AF, Green WN, Hartman DS, Claudio T. Efficiency of acetylcholine receptor subunit assembly and its regulation by cAMP. J Cell Biol. 1991;113:623–36.PubMedCrossRef Ross AF, Green WN, Hartman DS, Claudio T. Efficiency of acetylcholine receptor subunit assembly and its regulation by cAMP. J Cell Biol. 1991;113:623–36.PubMedCrossRef
141.
Zurück zum Zitat Chen LC, Nadziejko C. Effects of subchronic exposures to concentrated ambient particles (CAPs) in mice. V. CAPs exacerbate aortic plaque development in hyperlipidemic mice. Inhal Toxicol. 2005;17:217–24.PubMedCrossRef Chen LC, Nadziejko C. Effects of subchronic exposures to concentrated ambient particles (CAPs) in mice. V. CAPs exacerbate aortic plaque development in hyperlipidemic mice. Inhal Toxicol. 2005;17:217–24.PubMedCrossRef
142.
Zurück zum Zitat Sun Q, Wang A, Jin X, Natanzon A, Duquaine D, Brook RD, Aguinaldo JG, Fayad ZA, Fuster V, Lippmann M, et al. Long-term air pollution exposure and acceleration of atherosclerosis and vascular inflammation in an animal model. JAMA. 2005;294:3003–10.PubMedCrossRef Sun Q, Wang A, Jin X, Natanzon A, Duquaine D, Brook RD, Aguinaldo JG, Fayad ZA, Fuster V, Lippmann M, et al. Long-term air pollution exposure and acceleration of atherosclerosis and vascular inflammation in an animal model. JAMA. 2005;294:3003–10.PubMedCrossRef
143.
Zurück zum Zitat Araujo JA, Barajas B, Kleinman M, Wang X, Bennett BJ, Gong KW, Navab M, Harkema J, Sioutas C, Lusis AJ, Nel AE. Ambient particulate pollutants in the ultrafine range promote early atherosclerosis and systemic oxidative stress. Circ Res. 2008;102:589–96.PubMedPubMedCentralCrossRef Araujo JA, Barajas B, Kleinman M, Wang X, Bennett BJ, Gong KW, Navab M, Harkema J, Sioutas C, Lusis AJ, Nel AE. Ambient particulate pollutants in the ultrafine range promote early atherosclerosis and systemic oxidative stress. Circ Res. 2008;102:589–96.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Sun Q, Yue P, Kirk RI, Wang A, Moatti D, Jin X, Lu B, Schecter AD, Lippmann M, Gordon T, et al. Ambient air particulate matter exposure and tissue factor expression in atherosclerosis. Inhal Toxicol. 2008;20:127–37.PubMedCrossRef Sun Q, Yue P, Kirk RI, Wang A, Moatti D, Jin X, Lu B, Schecter AD, Lippmann M, Gordon T, et al. Ambient air particulate matter exposure and tissue factor expression in atherosclerosis. Inhal Toxicol. 2008;20:127–37.PubMedCrossRef
145.
Zurück zum Zitat Ying Z, Kampfrath T, Thurston G, Farrar B, Lippmann M, Wang A, Sun Q, Chen LC, Rajagopalan S. Ambient particulates alter vascular function through induction of reactive oxygen and nitrogen species. Toxicol Sci. 2009;111:80–8.PubMedPubMedCentralCrossRef Ying Z, Kampfrath T, Thurston G, Farrar B, Lippmann M, Wang A, Sun Q, Chen LC, Rajagopalan S. Ambient particulates alter vascular function through induction of reactive oxygen and nitrogen species. Toxicol Sci. 2009;111:80–8.PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Quan C, Sun Q, Lippmann M, Chen LC. Comparative effects of inhaled diesel exhaust and ambient fine particles on inflammation, atherosclerosis, and vascular dysfunction. Inhal Toxicol. 2010;22:738–53.PubMedPubMedCentralCrossRef Quan C, Sun Q, Lippmann M, Chen LC. Comparative effects of inhaled diesel exhaust and ambient fine particles on inflammation, atherosclerosis, and vascular dysfunction. Inhal Toxicol. 2010;22:738–53.PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Chen T, Jia G, Wei Y, Li J. Beijing ambient particle exposure accelerates atherosclerosis in ApoE knockout mice. Toxicol Lett. 2013;223:146–53.PubMedCrossRef Chen T, Jia G, Wei Y, Li J. Beijing ambient particle exposure accelerates atherosclerosis in ApoE knockout mice. Toxicol Lett. 2013;223:146–53.PubMedCrossRef
148.
Zurück zum Zitat Rao X, Zhong J, Maiseyeu A, Gopalakrishnan B, Villamena FA, Chen LC, Harkema JR, Sun Q, Rajagopalan S. CD36-dependent 7-ketocholesterol accumulation in macrophages mediates progression of atherosclerosis in response to chronic air pollution exposure. Circ Res. 2014;115:770–80.PubMedPubMedCentralCrossRef Rao X, Zhong J, Maiseyeu A, Gopalakrishnan B, Villamena FA, Chen LC, Harkema JR, Sun Q, Rajagopalan S. CD36-dependent 7-ketocholesterol accumulation in macrophages mediates progression of atherosclerosis in response to chronic air pollution exposure. Circ Res. 2014;115:770–80.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Keebaugh AJ, Sioutas C, Pakbin P, Schauer JJ, Mendez LB, Kleinman MT. Is atherosclerotic disease associated with organic components of ambient fine particles? Sci Total Environ. 2015;533:69–75.PubMedCrossRef Keebaugh AJ, Sioutas C, Pakbin P, Schauer JJ, Mendez LB, Kleinman MT. Is atherosclerotic disease associated with organic components of ambient fine particles? Sci Total Environ. 2015;533:69–75.PubMedCrossRef
150.
Zurück zum Zitat Wan Q, Cui X, Shao J, Zhou F, Jia Y, Sun X, Zhao X, Chen Y, Diao J, Zhang L. Beijing ambient particle exposure accelerates atherosclerosis in ApoE knockout mice by upregulating visfatin expression. Cell Stress Chaperones. 2014;19:715–24.PubMedPubMedCentralCrossRef Wan Q, Cui X, Shao J, Zhou F, Jia Y, Sun X, Zhao X, Chen Y, Diao J, Zhang L. Beijing ambient particle exposure accelerates atherosclerosis in ApoE knockout mice by upregulating visfatin expression. Cell Stress Chaperones. 2014;19:715–24.PubMedPubMedCentralCrossRef
151.
Zurück zum Zitat Mauderly J, Kracko D, Brower J, Doyle-Eisele M, McDonald J, Lund A, Seilkop S. The National Environmental Respiratory Center (NERC) experiment in multi-pollutant air quality health research: IV. Vascular effects of repeated inhalation exposure to a mixture of five inorganic gases. Inhal Toxicol. 2014;26:691–6.PubMedCrossRef Mauderly J, Kracko D, Brower J, Doyle-Eisele M, McDonald J, Lund A, Seilkop S. The National Environmental Respiratory Center (NERC) experiment in multi-pollutant air quality health research: IV. Vascular effects of repeated inhalation exposure to a mixture of five inorganic gases. Inhal Toxicol. 2014;26:691–6.PubMedCrossRef
152.
Zurück zum Zitat Yang Z, Knight CA, Mamerow MM, Vickers K, Penn A, Postlethwait EM, Ballinger SW. Prenatal environmental tobacco smoke exposure promotes adult atherogenesis and mitochondrial damage in apolipoprotein E−/− mice fed a chow diet. Circulation. 2004;110:3715–20.PubMedCrossRef Yang Z, Knight CA, Mamerow MM, Vickers K, Penn A, Postlethwait EM, Ballinger SW. Prenatal environmental tobacco smoke exposure promotes adult atherogenesis and mitochondrial damage in apolipoprotein E−/− mice fed a chow diet. Circulation. 2004;110:3715–20.PubMedCrossRef
153.
Zurück zum Zitat Cakir Y, Yang Z, Knight CA, Pompilius M, Westbrook D, Bailey SM, Pinkerton KE, Ballinger SW. Effect of alcohol and tobacco smoke on mtDNA damage and atherogenesis. Free Radic Biol Med. 2007;43:1279–88.PubMedCrossRef Cakir Y, Yang Z, Knight CA, Pompilius M, Westbrook D, Bailey SM, Pinkerton KE, Ballinger SW. Effect of alcohol and tobacco smoke on mtDNA damage and atherogenesis. Free Radic Biol Med. 2007;43:1279–88.PubMedCrossRef
Metadaten
Titel
The Apoe−/− mouse model: a suitable model to study cardiovascular and respiratory diseases in the context of cigarette smoke exposure and harm reduction
verfasst von
Giuseppe Lo Sasso
Walter K. Schlage
Stéphanie Boué
Emilija Veljkovic
Manuel C. Peitsch
Julia Hoeng
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2016
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-016-0901-1

Weitere Artikel der Ausgabe 1/2016

Journal of Translational Medicine 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.