Next Article in Journal
Evaluation of Medication-Related Osteonecrosis of the Jaw (MRONJ) in Terms of Staging and Treatment Strategies by Dental Students at Different Educational Levels
Next Article in Special Issue
Comparison of BODE and ADO Indices in Predicting COPD-Related Medical Costs
Previous Article in Journal
The Prone Lateral Approach for Lumbar Fusion—A Review of the Literature and Case Series
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Relationship of Cholesterol Responses to Mitochondrial Dysfunction and Lung Inflammation in Chronic Obstructive Pulmonary Disease

Department of Medicine, State University of New York Downstate Health Sciences University, New York, NY 11203, USA
*
Author to whom correspondence should be addressed.
Medicina 2023, 59(2), 253; https://doi.org/10.3390/medicina59020253
Submission received: 6 December 2022 / Revised: 25 January 2023 / Accepted: 26 January 2023 / Published: 28 January 2023
(This article belongs to the Special Issue Update on COPD and Cystic Fibrosis)

Abstract

:
Hyperlipidemia is frequently reported in chronic obstructive pulmonary disease (COPD) patients and is linked to the progression of the disease and its comorbidities. Hypercholesterolemia leads to cholesterol accumulation in many cell types, especially immune cells, and some recent studies suggest that cholesterol impacts lung epithelial cells’ inflammatory responses and mitochondrial responses. Several studies also indicate that targeting cholesterol responses with either statins or liver X receptor (LXR) agonists may be plausible means of improving pulmonary outcomes. Equally, cholesterol metabolism and signaling are linked to mitochondrial dysfunction and inflammation attributed to COPD progression. Here, we review the current literature focusing on the impact of cigarette smoke on cholesterol levels, cholesterol efflux, and the influence of cholesterol on immune and mitochondrial responses within the lungs.

1. Introduction to Hypercholesterolemia in COPD

Reverse lipid transport plays an important role in pulmonary cells, especially alveolar macrophages and type II pneumocytes. Hyperlipidemia, a condition with high levels of lipids in the blood, is common in chronic obstructive pulmonary disease (COPD) patients but its relationship to COPD and lung function is not well established [1]. COPD is the fourth leading cause of death in the US [2]. Cholesterol levels are an important prognostic marker of cardiovascular risk in patients with COPD [3]. Exposure to cigarette smoke is the primary environmental factor associated with the development of COPD. Alpha-1 antitrypsin deficiency is the most common genetic factor linked to COPD. Almost 20% of all coronary heart disease deaths are associated with smoking [2]. Several clinical studies suggest that high cholesterol levels may also significantly contribute to COPD progression or other comorbidities. A clinical randomized cross-sectional study enrolling 100 patients with severe and very severe COPD demonstrated that patients with very severe COPD had a positive correlation with low-density lipoprotein (LDL)-C (r = 0.895, p < 0.001) and had elevated levels of cholesterol (6.16 ± 1.5 vs. 5.61 ± 1.1, p = 0.039) [4]. Another recent multi-center COSYCONET (COPD and SYstemic consequences-COmorbidities NETwork) cohort included 1746 COPD (Gold class 1-4) patients who demonstrated that hyperlipidemia was associated with lower intrathoracic volume and higher forced expiratory volume in 1 s (FEV1) suggesting that COPD patients with hyperlipidemia showed less lung hyperinflation and airway obstruction [1]. Another cross-sectional observational study with 125 different classes of COPD severity patients found that hyperlipidemia was common in combination with COPD, but no statistical difference was observed for total cholesterol (TC), LDL, very low-density lipoprotein (VLDL), and triglycerides between mild, moderate, and severe COPD [5]. A cross-sectional study investigating 644 adolescents aged 15–16 years old showed that the percent predicted values of forced vital capacity (FVC) (p  =  0.036) and FEV1 (p  =  0.017) were also inversely associated with high-density lipoprotein (HDL) cholesterol [6]. HDL-cholesterol levels also correlate with muscle areas (pectoralis, rectus abdominis, and erector spinae), densities, and total energy expenditure in patients with or at risk for COPD [7]. Recently, several studies suggested a relationship between body mass index (BMI) and COPD. Some studies observed a negative impact of BMI on COPD [8,9], while others suggest a better prognosis with excess body weight [10]. This “obesity paradox” is a topic for discussion, and cholesterol could play a significant role, but additional research is needed on this lifestyle topic [11,12]. Patients with COPD are markedly inactive because of airflow limitation and shortness of breath [13]. Decreased active time correlates with cardiac event risk, mortality, and higher HDL cholesterol levels [14,15].
Smoking is believed to cause intracellular cholesterol accumulation that plays an important role in lung inflammation in COPD [16]. Cholesterol is also a vital component of surfactant, a complex mixture of lipids and proteins secreted by alveolar type II epithelial cells that helps reduce the work of breathing by lowering the surface tension during inspiration [17]. However, dysregulation of cholesterol homeostasis and overloading macrophages with cholesterol in COPD plays cytotoxic and inflammatory responses. Advanced COPD is associated with the generation of inducible bronchus-associated lymphoid tissue (iBALT). Oxysterols, the metabolites of cholesterol, are elevated in COPD patients and are involved in iBALT generation and immune pathogenesis [18]. Therefore, several studies have investigated the potential benefit of cholesterol-lowering medications in COPD patients. 3-hydroxy-3-methyl-3-glutaryl coenzyme A reductase inhibitors (statins), reduce the expression of chemokines, such as CCL2 and CXCL8, and matrix metalloprotease (MMP) 9, all of which are increased in COPD [19]. A recent meta-analysis that included eight studies and 1323 participants showed that statins reduced cholesterol and lung inflammation without any clear improvement in functional capacity or lung function [20]. Another network meta-analysis study suggested that statins reduced the risk of all-cause and cause-specific mortality, pulmonary hypertension, and the level of C-reactive protein (CRP), IL6, IL8, and TNF-α in COPD patients [21]. However, statins may not impact exacerbation rates in patients with COPD [22], which is an important factor in COPD progression. In animal models of COPD, inhibition of the oxysterol pathway with the CYP7B1 inhibitor clotrimazole resolved B cell-driven iBALT formation and attenuated cigarette smoke-induced emphysema [23]. Here, we will briefly discuss the role of smoke exposure on cholesterol levels and responses in COPD and review the literature on the subsequent impact of cholesterol on inflammation and mitochondrial function within the lungs.

2. Is There a Relationship between Smoking Status and Lung Cholesterol Levels?

Several studies investigated blood or bronchoalveolar lavage (BAL) fluid cholesterol levels and whether smoking or disease status are associated with these changes. The effects of smoking on serum lipids suggest that it induces an atherogenic lipid profile which in turn has ramifications for cardiovascular well-being as well as systemic inflammation. A one-year, prospective, double-blind, randomized, placebo-controlled clinical trial demonstrated that smoke cessation leads to increases in HDL cholesterol, total HDL, and large HDL compared to individuals that continue to smoke, especially in women [24]. There were no significant changes in LDL cholesterol with smoke cessation [24]. In current smokers, increased smoking inhalation is associated with increases in total cholesterol, LDL cholesterol, and triglycerides [25]. CO levels were also associated with lower HDL-cholesterol and total HDL particles [25]. Even with second-hand smoke, HDL cholesterol levels are lower in dyslipidemic children [26]. A recent publication demonstrated elevated plasma levels of cholesterol in COPD patients [27]. The exact mechanisms for how smoking affects the lipid profile are not understood but are postulated to be related to catecholamine release with subsequent changes in circulating free fatty acids and resultant increases in VLDL and LDL with a decrease in HDL [28].
Total lipid concentrations are decreased in the BAL fluids of COPD patients compared to healthy controls, and cholesterol is the second most abundant lipid in the airways [29]. In COPD patients, the amount of cholesterol in BAL fluid is reduced compared to healthy controls, which is also positively correlated [29]. This loss of BAL fluid cholesterol is also observed in smoke-exposed mice [29]. A recent preclinical study suggested that the foamy macrophage phenotype and inflammatory response (primarily IL-1α) following cigarette smoke exposure may be due to the direct damage made to pulmonary lipids [30]. The investigators also observed that intranasal delivery of human oxidized low-density lipoprotein led to a similar macrophage response to cigarette smoke exposure [30], which was attributed to altered surfactant responses.
Cigarette smoke can impair the surface tension lowering the ability of surfactants. The lipid constituents in surfactant aid in their ability to reduce surface tension, facilitate gas exchange, and help in surfactant production and immune modulation [31]. A systematic lipidomic analysis of BAL fluid in subjects with COPD that had achieved smoking cessation was compared with BAL from healthy subjects, and observed a decrease in alveolar lipids, which correlated with a reduction in lung function [29]. Smoking decreases BAL fluid lipids, especially phospholipids and cholesterol, followed by sphingolipids and glycerolipids [32]. Decreases in total BAL lipid, total PL, PC 30:0, PC 32:0, and total cholesterol, amongst other lipids, strongly correlated with decreased lung function, which was also observed in the second-hand smoke mice model [29]. Multiple mechanisms are postulated for the effect smoking has on pulmonary lipids, including the destruction of type II pneumocytes, decreased synthesis, and alterations in transport [33,34]. It is important to note that a small clinical trial showed improved lung function in patients with stable chronic bronchitis who were given supplemental surfactant therapy [35]. This further suggests that pulmonary lipids/surfactants may play a role in the pathophysiology of COPD.

3. Cholesterol Levels Can Influence Systemic and Local Lung Inflammation in COPD

Hypercholesterolemia leads to cholesterol accumulation in many cell types, especially immune cells, that contributes to elevated inflammatory responses, including inflammasome activation, Toll-like receptor (TLR) signaling, and the production of innate immune cells [36]. Elevated levels of LDL promote cholesterol accumulation and heightened inflammatory response. To opposes this, HDL promotes the cellular efflux of cholesterol and reduces inflammation. Since HDL can suppress inflammation, some investigators have suggested that it plays a protective effect on the smoke-exposed lung [37]. Decreased HDL could contribute to more inflammation within the lung. HDL stimulates the release of cholesterol from activated cholesterol-filled macrophages, which diminishes their inflammatory response in mice [38]. In acute septic patients and septic animal models, cholesterol efflux from macrophages to plasma or HDL enhances inflammation [39].
In macrophages, cigarette smoke impairs cholesterol efflux by reducing the expression of ATP binding cassette (ABC) A1 leading to enhanced TLR4/myeloid differentiation primary response 88 (MyD88) signaling, MMP9, MMP13, TNFα, IL1β, IL17, IFNγ expression, and suppression of IL10 that are also responsive to liver X receptor (LXR) agonists in mice [34]. The same study demonstrated that treatment with an LXR agonist, T0901317, prevented cigarette smoke-induced loss of lung function in AKR/J mice [34]. Cigarette smoke directly impacts cholesterol efflux in macrophages and reduces the frequency of foamy cell-like/lipid-laden macrophages in the lungs [34]. Another LXR agonist, GW3965, suppressed the production of lipopolysaccharide (LPS)-dependent CXC10 and CCL5 and elevated IL-10 production in alveolar macrophages [40]. A study conducted in Abca1 deficient mice demonstrated reduced airway eosinophils, T helper (Th) 2 cytokines, and Th2 cells, and increased airway neutrophils and IL-17 [41]. Therefore, ABCA1 plays a crucial role in cholesterol-mediated lung inflammation in cigarette smoke exposures. A recent transcriptome and lipidome study focusing on macrophages from COPD patients demonstrated that there are cholesterol metabolism and IFN-α and γ signatures within the macrophages that are GOLD grade-dependent [42]. In mice, cholesterol efflux pathways mediated by apolipoprotein (APO) E, ABCA1, and ABCG1 suppress the production of inflammatory cells in the bone marrow and the spleen [43]. This is also relevant to humans, as there is an inverse relationship between HDL cholesterol levels and blood monocyte numbers in children with familial hypercholesterolemia [44]. Therefore, elevated cholesterol influences immune cell proliferation in addition to inflammation responses. Promoting reverse lipid transport with recombinant ApoA-1 Milano/phospholipid complex (MDCO-216) prevented smoke-induced immune cell infiltration and partially protected lung function in mice [33].
Recently, Li et al. demonstrated that immune responses in airway epithelial cells are also influenced by cholesterol [27]. They observed that cigarette smoke extract (CSE)-induced expression of IL8 and IL6 in human bronchial epithelial (HBE) cells could be reversed by either a statin (atorvastatin) or LXR agonists (T0901317 and GW3965) [27]. Equally, they identified that the steroidogenic acute regulatory-related lipid transfer domain-3 (STARD3) was a critical player in cholesterol and cigarette smoke-induced IL6, IL8, and IL1β [27]. STARD3 is a sterol-binding protein that creates endoplasmic reticulum (ER)-endosome contact sites, and STARD3 can regulate cholesterol accumulation in endosomes rather than the plasma membrane [45]. Cholesterol regulation of STARD3 expression may be a mechanism for how smoke exposure modulates epithelial inflammation response sensitivity within the lung, and it also appears to influence cholesterol levels in the mitochondria. It may contribute to mitochondrial dysfunction [27]. STARD3 also significantly increases macrophage ABCA1 expression, enhances cholesterol efflux to APOA1, and reduces biosynthesis of cholesterol, cholesteryl ester, fatty acids, triacylglycerol, and phospholipids [46]. The overexpression of STARD3 (also known as MLN64) in osteoclasts enhances RANKL induction of inflammatory factors, including NFκB, TNFα, IL1β, IL6, and MMP1 [47]. Additionally, regulating plasma membrane cholesterol content may be necessary for regulating neutrophil-associated inflammation and adhesion, as maintaining cholesterol membrane content coincides with reduced inflammation in other lung diseases, such as cystic fibrosis [48].
The impact of elevated cholesterol on lung surfactants must also be considered as surfactant composition plays a major role in lung surface tension, but surfactant also has immune properties, with extracellular large surfactant aggregates observed to bind to non-typeable Haemophilus influenzae to inhibit adhesion of the bacterium to pneumocytes and prevent invasion [49]. Furthermore, in rodents, over 80% of the cholesterol in the rat lung comes from cholesterol uptake from circulation [50]. Therefore, elevated systemic cholesterol will influence cholesterol uptake levels in the lungs and could impact surfactant composition and function.
Finally, the impact of hypercholesterolemia on TLR signaling and 25-hydroxycholesterol (25HC), a metabolite of cholesterol that is produced and secreted by macrophages in response to TLR activation [51], is extensively studied in atherosclerosis. However, 25HC is also known to be elevated in COPD lungs, and its levels inversely correlate with important readouts of lung function, such as the percent of predicted FVC, FEV1, and diffusing capacity of carbon monoxide (DLCO) [52]. 25HC is encoded by the Ch25h gene, a known IFN-inducible gene. Many inflammation triggers, such as LPS, polyinosinic-polycytidylic acid (poly(I:C)), viruses, and interferons, can upregulate Ch25h expression and increase the production and release of 25HC in macrophages [53]. 25HC can also inhibit the growth of a broad range of viruses. Recently, 25HC inhibited SARS-CoV-2 and other coronaviruses by depleting membrane cholesterol [54]. Therefore, intra- and extra-cellular cholesterol, cholesterol efflux, and metabolites of cholesterol will affect immune responses in COPD and influence how the lung responds to inflammatory stimuli.

4. Hypercholesterolemia and Mitochondrial Dysfunction

The mitochondria play critical roles in energy production via oxidative phosphorylation, cell signaling, and maintaining tissue homeostasis. The mitochondria are regulated by a balance of mitochondrial membrane potential, mitochondrial calcium, mitochondrial DNA (mtDNA), and clearance of impaired mitochondria via mitophagy. However, many different insults may disrupt mitochondria regulation within the lung, including inflammation, infection, air pollution, cigarette smoke, and alteration of oxygen levels. These insults may cause mitochondria dysfunction via multiple different mechanisms, including mtDNA impairment and release, increased reactive oxygen species (ROS), reduced mitochondrial membrane potential, and dysregulation of calcium signaling and storage [55]. These alterations can ultimately induce metabolic reprogramming, inflammasome activation, apoptosis, senescence, oxidative stress, and inflammasome activation [56]. Recently, hypercholesteremia was demonstrated to induce mitochondria dysfunction in airway epithelial cells in COPD [27].
In preclinical models of the disease, hypercholesteremia is observed to induce mitochondrial dysfunction [57]. Feeding C57B1/6J mice a high-fat diet resulted in the downregulation of genes in mitochondrial oxidative phosphorylation [57]. This was further elucidated in humans where a high consumption of fat diet-induced dysregulation of target genes involved oxidative phosphorylation. Another potential mechanism identified is the accumulation of cholesterol within the mitochondria, causing a decrease in the mitochondrial glutathione stores, a key component in ATP production, by preventing its transport into the mitochondria [57]. Multiple studies have demonstrated that the accumulation of fatty acids within the inner mitochondrial membrane increases susceptibility to peroxidation leading to mitochondrial dysfunction [58]. The enzyme carnitine palmitoyltransferase (CPT) 1, which allows for the inward transportation of fatty acids, is linked to these fatty acids-associated mitochondrial dysfunction. Another method is through passive diffusion, described as the ‘flip-flop’ mechanism [59]. It explains how cholesterol bilayer motion influences cholesterol distribution within the cell membrane. Investigating the mitochondria genome may provide insights into understanding the physiological mechanisms by which lipid levels affect mitochondrial function and the possible influence of mtDNA on lipid changes [60]. One study identified 10 mitochondrial single nucleotide polymorphisms (mtSNPs) associated with HDL and cholesterol [60]. While there are many studies conducted to investigate the mechanisms by which hypercholesterolemia affects mitochondria function, further research is needed to identify these changes in the immunopathology of various diseases.
Mitochondrial dysfunction plays a critical role in driving the inflammatory process of lung inflammation in patients with COPD [61]. HBE cells (the cell line BEAS-2B) exposed to prolonged CSE or primary HBE cells isolated from COPD subjects have abnormal mitochondria, observed with increased mitochondrial mass, fragmentation, branching, and quality of cristae [62]. Reduced expression of the peroxisome proliferator-activated receptor-gamma coactivator (PGC) 1α, a key regulator of mitochondrial biogenesis regulated by sirtuin-1, is linked to the dysregulation of mitophagy, a cellular process to selectively remove aged and damaged mitochondria and cellular senescence in the lung [63]. Increased mitochondrial iron is also linked to mitochondrial dysfunction in COPD patients [64]. Mitochondrial dysfunction in macrophages from COPD patients is also linked to defective bacterial phagocytosis [65]. Extracellular mtDNA can activate the NLRP3 inflammasome [66] and the cytosolic DNA sensor cyclic guanosine monophosphate-adenosine monophosphate synthase [67]. Both of these responses would lead to the activation of innate immunity and the secretion of inflammation mediators. Increased mtDNA is observed in the urine of COPD patients and correlates with disease severity [68]. Mitochondrial dysfunction may play a key role in the pathogenesis and progression of COPD, and hypercholesterolemia may be a confounding factor for mitochondrial dysfunction in COPD.
There are large aggregates of mitochondria within muscle fibers of the diaphragm of a COPD patient [69] that may cause an increase in cytochrome oxidase activity and ROS production. The known mechanisms by which the mitochondria induce inflammation in COPD patients are a decrease in oxidative capacity, increased ROS production, and increased apoptosis and autophagy. The signaling pathways of iron-regulatory proteins, prohibitin complexes, PPAR-y coactivator, and peroxisome proliferator-activated receptors have also been demonstrated to contribute to lung inflammation in COPD patients. Interestingly, there is growing evidence that hypercholesteremia may significantly contribute to mitochondria dysfunction in COPD patients [27]. Preclinical studies suggest that there is an increase in cholesterol biosynthesis in lung epithelial and lipofibroblasts during aging, indicating a potential link between hypercholesteremia and HBE function in driving inflammation in disease [70]. A recent study by Li et al. in 2022 demonstrated that the external cholesterol level alterations influenced mitochondrial dynamics leading to a dysregulation of interleukin production via the down-regulation of the SREPB2/LDL-R pathway [27]. In addition, the STARD cholesterol transporters are key players in cholesterol distribution within and outside the cell and inflammation mediators, as mentioned in Section 3. The STARD family includes 15 different members. Of the 15 members, STARD1, STARD3, STARD4, and STARD5 regulate cholesterol homeostasis and assist in cholesterol transport. STARD3 can facilitate the transport of cholesterol intracellularly from endosomes to the mitochondria [45]. This may ultimately lead to cholesterol accumulation in the mitochondria resulting in increased oxidative stress and enhancement of steroidogenesis [45]. STARD3-mitochondrial fusion genes, including mitofusin (MFN) 2, may contribute to the induction of mitochondria dysfunction leading to the development of COPD [27].
Moreover, this study demonstrated that hypercholesteremia upregulates the expression of STARD3, which led to increased sensitivity of HBE cells to smoking and altered interleukin and cytokine production [27]. The authors suggested that there may be a dependency of STARD3 on airway epithelial sensitivity. When STARD3 is downregulated in preclinical models, it reversed the epithelial mitochondrial function and decreased sensitivity to various insults [27]. In addition, alterations in external cholesterol levels can directly influence mitochondria function [27]. It can downregulate inflammatory pathways, including the SREBP2/LDL-R pathway, and alters the transcriptomic expression of mitochondria-associated genes [27].
During cigarette smoke exposure, MFN2 expression levels were increased in human airway epithelia, while deletion of MFN2 altered the production of lipids [27]. MFN2 enhancement may be a compensatory response to cigarette smoke exposure. Chronic cigarette smoke exposure altered the STARD3 levels, which also altered the cholesterol levels within the mitochondria and externally [27]. This induces mitochondrial dysregulation leading to the accumulation of cholesterol, damaging its architecture and inducing activation of inflammatory signaling cascades. These findings demonstrated by Li provide new insights into how COPD patients with hypercholesteremia develop the disease, identify potential therapeutic targets, and possible indicators of disease severity. While significant progress has been made to identify the mechanisms by which hypercholesteremia induces mitochondria dysfunction leading to the development of disease, further research is needed to unravel new potential therapeutic targets and indicators of the disease process (See Figure 1 for possible mechanisms to cholesterol-induced inflammation and mitochondrial dysfunction in COPD).

5. Conclusions

Smoke’s regulation of cholesterol transport, inflammation, and mitochondrial dysfunction appears to be mechanistically linked to each other and may play a significant role in the increased risk of COPD and targeting cholesterol responses may represent a novel therapeutic approach. Epidemiologic studies suggest a link between pulmonary lipids and changes in pulmonary function from cigarette smoke exposure. However, only a few studies have identified pathogenic links of cholesterol to COPD pathogenesis. The effect of cigarette smoke on the expression and activity of the major ABC transporters, STARD3, MFN2, inflammation sensitivity, and mitochondrial dysfunction within the lung suggests that the disruption of lipid homeostasis is an early event in the development of COPD after cigarette smoke exposure. Here, we primarily focused on cholesterol changes, but other lipids and mediators are linked to COPD progression and also need to be discussed. Effects of race, gender, and diet are also confounding factors.
Similarly, we did not discuss the effects of cigarette smoking on cholesterol metabolism, biosynthesis, maturation, remodeling of cholesterol, cholesterol subfractions, and cholesterol catabolism. It will also be important to investigate the effect of cigarette smoke-induced oxidative modifications on cholesterol, lipid peroxidation, and other induced dysfunctions. Genetic factors, such as alpha-1 antitrypsin deficiency, are also major factors in COPD development and progression. Alpha-1 antitrypsin augmentation therapy is reported to coincide with reduced cholesterol levels in an observational study [71], but further research is needed. Furthermore, new cholesterol-targeting therapies must be extensively investigated in COPD subjects, such as proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors and inclisiran, a small interfering RNA therapy. Recent studies suggest that metabolism does not progress as previously thought, with organ-specific metabolic activity remaining high throughout growth and development from infancy to adolescence. However, metabolism declines throughout adulthood and old age [72]. This may have many outcomes on our view of aging and COPD progression. Finally, there is evidence that vaping products, such as electronic cigarettes, can yield a similar lipid profile to conventional tobacco smoking [73], which could have many long-term complications. Therefore, significant work is required to elucidate the causality of altered lung cholesterol metabolism and signaling within the lungs, but it represents an important and exciting study area for pulmonary diseases such as COPD.

Author Contributions

Conceptualization, B.J., H.A., J.R. and P.G.; writing—original draft preparation, B.J., H.A., J.R. and P.G.; writing—review and editing, B.J., H.A., J.R. and P.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants made available to P.G. (the Alpha-1 Foundation, 493373 and 614218).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kahnert, K.; Lucke, T.; Huber, R.M.; Behr, J.; Biertz, F.; Vogt, A.; Watz, H.; Alter, P.; Fahndrich, S.; Bals, R.; et al. Relationship of hyperlipidemia to comorbidities and lung function in COPD: Results of the COSYCONET cohort. PLoS ONE 2017, 12, e0177501. [Google Scholar] [CrossRef] [PubMed]
  2. U.S. Department of Health and Human Services. The Health Consequences of Smoking—50 Years of Progress: A Report of the Surgeon General; U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health: Atlanta, GA, USA, 2014.
  3. Shi, Y.; Zhang, J.; Huang, Y. Prediction of cardiovascular risk in patients with chronic obstructive pulmonary disease: A study of the National Health and Nutrition Examination Survey database. BMC Cardiovasc. Disord. 2021, 21, 417. [Google Scholar] [CrossRef] [PubMed]
  4. Zafirova-Ivanovska, B.; Stojkovikj, J.; Dokikj, D.; Anastasova, S.; Debresliovska, A.; Zejnel, S.; Stojkovikj, D. The Level of Cholesterol in COPD Patients with Severe and Very Severe Stage of the Disease. Open Access Maced J. Med. Sci. 2016, 4, 277–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Arora, S.; Jain, R.; Mohan, A.; Tiwari, P.; Guleria, R. Lipid Profile Status in Chronic Obstructive Pulmonary Disease and its Association with Disease Severity. Int. J. Respir. Pulm. Med. 2018, 5, 89. [Google Scholar] [CrossRef] [Green Version]
  6. Park, J.H.; Mun, S.; Choi, D.P.; Lee, J.Y.; Kim, H.C. Association between high-density lipoprotein cholesterol level and pulmonary function in healthy Korean adolescents: The JS high school study. BMC Pulm. Med. 2017, 17, 190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Shirahata, T.; Sato, H.; Yogi, S.; Inoue, K.; Niitsu, M.; Miyazawa, H.; Akagami, T.; Soma, M.; Mio, T.; Nagata, M.; et al. Possible association of high-density lipoprotein cholesterol levels with trunk muscle deficits and decrease in energy expenditure in patients with or at risk for COPD: A pilot study. Respir. Investig. 2022, 60, 720–724. [Google Scholar] [CrossRef] [PubMed]
  8. Kim, E.K.; Singh, D.; Park, J.H.; Park, Y.B.; Kim, S.I.; Park, B.; Park, J.; Kim, J.H.; Kim, M.A.; Lee, J.H.; et al. Impact of Body Mass Index Change on the Prognosis of Chronic Obstructive Pulmonary Disease. Respiration 2020, 99, 943–953. [Google Scholar] [CrossRef]
  9. Park, H.J.; Cho, J.H.; Kim, H.J.; Park, J.Y.; Lee, H.S.; Byun, M.K. The effect of low body mass index on the development of chronic obstructive pulmonary disease and mortality. J. Intern. Med. 2019, 286, 573–582. [Google Scholar] [CrossRef]
  10. Guo, Y.; Zhang, T.; Wang, Z.; Yu, F.; Xu, Q.; Guo, W.; Wu, C.; He, J. Body mass index and mortality in chronic obstructive pulmonary disease: A dose-response meta-analysis. Medicine 2016, 95, e4225. [Google Scholar] [CrossRef]
  11. Iyer, A.S.; Dransfield, M.T. The “Obesity Paradox” in Chronic Obstructive Pulmonary Disease: Can It Be Resolved? Ann. Am. Thorac. Soc. 2018, 15, 158–159. [Google Scholar] [CrossRef]
  12. Ma, Y.L.; Zhao, H.J.; Su, Y.H. Association between waist circumference change and incident chronic obstructive pulmonary disease among Chinese adults: A 10-year cohort study. Sci. Rep. 2022, 12, 18402. [Google Scholar] [CrossRef] [PubMed]
  13. Pitta, F.; Troosters, T.; Spruit, M.A.; Probst, V.S.; Decramer, M.; Gosselink, R. Characteristics of physical activities in daily life in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2005, 171, 972–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Bakrania, K.; Edwardson, C.L.; Bodicoat, D.H.; Esliger, D.W.; Gill, J.M.; Kazi, A.; Velayudhan, L.; Sinclair, A.J.; Sattar, N.; Biddle, S.J.; et al. Associations of mutually exclusive categories of physical activity and sedentary time with markers of cardiometabolic health in English adults: A cross-sectional analysis of the Health Survey for England. BMC Public Health 2016, 16, 25. [Google Scholar] [CrossRef] [Green Version]
  15. Young, D.R.; Hivert, M.F.; Alhassan, S.; Camhi, S.M.; Ferguson, J.F.; Katzmarzyk, P.T.; Lewis, C.E.; Owen, N.; Perry, C.K.; Siddique, J.; et al. Sedentary Behavior and Cardiovascular Morbidity and Mortality: A Science Advisory from the American Heart Association. Circulation 2016, 134, e262–e279. [Google Scholar] [CrossRef]
  16. Kotlyarov, S.; Kotlyarova, A. Molecular Mechanisms of Lipid Metabolism Disorders in Infectious Exacerbations of Chronic Obstructive Pulmonary Disease. Int. J. Mol. Sci. 2021, 22, 7634. [Google Scholar] [CrossRef] [PubMed]
  17. Keating, E.; Rahman, L.; Francis, J.; Petersen, A.; Possmayer, F.; Veldhuizen, R.; Petersen, N.O. Effect of cholesterol on the biophysical and physiological properties of a clinical pulmonary surfactant. Biophys. J. 2007, 93, 1391–1401. [Google Scholar] [CrossRef] [PubMed]
  18. Sheng, X.; Murphy, M.J.; MacDonald, T.M.; Schembri, S.; Simpson, W.; Winter, J.; Winter, J.H.; Wei, L. Effect of statins on total cholesterol concentrations, cardiovascular morbidity, and all-cause mortality in chronic obstructive pulmonary disease: A population-based cohort study. Clin. Ther. 2012, 34, 374–384. [Google Scholar] [CrossRef]
  19. Barnes, P.J.; Celli, B.R. Systemic manifestations and comorbidities of COPD. Eur. Respir. J. 2009, 33, 1165–1185. [Google Scholar] [CrossRef] [Green Version]
  20. Walsh, A.; Perrem, L.; Khashan, A.S.; Henry, M.T.; Ni Chroinin, M. Statins versus placebo for people with chronic obstructive pulmonary disease. Cochrane Database Syst. Rev. 2019, 7, CD011959. [Google Scholar] [CrossRef]
  21. Lu, Y.; Chang, R.; Yao, J.; Xu, X.; Teng, Y.; Cheng, N. Effectiveness of long-term using statins in COPD—A network meta-analysis. Respir. Res. 2019, 20, 17. [Google Scholar] [CrossRef]
  22. Criner, G.J.; Connett, J.E.; Aaron, S.D.; Albert, R.K.; Bailey, W.C.; Casaburi, R.; Cooper, J.A., Jr.; Curtis, J.L.; Dransfield, M.T.; Han, M.K.; et al. Simvastatin for the prevention of exacerbations in moderate-to-severe COPD. N. Engl. J. Med. 2014, 370, 2201–2210. [Google Scholar] [CrossRef] [Green Version]
  23. Jia, J.; Conlon, T.M.; Sarker, R.S.; Tasdemir, D.; Smirnova, N.F.; Srivastava, B.; Verleden, S.E.; Gunes, G.; Wu, X.; Prehn, C.; et al. Cholesterol metabolism promotes B-cell positioning during immune pathogenesis of chronic obstructive pulmonary disease. EMBO Mol. Med. 2018, 10, e8349. [Google Scholar] [CrossRef] [PubMed]
  24. Gepner, A.D.; Piper, M.E.; Johnson, H.M.; Fiore, M.C.; Baker, T.B.; Stein, J.H. Effects of smoking and smoking cessation on lipids and lipoproteins: Outcomes from a randomized clinical trial. Am. Heart J. 2011, 161, 145–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Gossett, L.K.; Johnson, H.M.; Piper, M.E.; Fiore, M.C.; Baker, T.B.; Stein, J.H. Smoking intensity and lipoprotein abnormalities in active smokers. J. Clin. Lipidol. 2009, 3, 372–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Neufeld, E.J.; Mietus-Snyder, M.; Beiser, A.S.; Baker, A.L.; Newburger, J.W. Passive cigarette smoking and reduced HDL cholesterol levels in children with high-risk lipid profiles. Circulation 1997, 96, 1403–1407. [Google Scholar] [CrossRef]
  27. Li, L.; Liu, Y.; Liu, X.; Zheng, N.; Gu, Y.; Song, Y.; Wang, X. Regulatory roles of external cholesterol in human airway epithelial mitochondrial function through STARD3 signalling. Clin. Transl. Med. 2022, 12, e902. [Google Scholar] [CrossRef] [PubMed]
  28. Chelland Campbell, S.; Moffatt, R.J.; Stamford, B.A. Smoking and smoking cessation—The relationship between cardiovascular disease and lipoprotein metabolism: A review. Atherosclerosis 2008, 201, 225–235. [Google Scholar] [CrossRef] [PubMed]
  29. Agudelo, C.W.; Kumley, B.K.; Area-Gomez, E.; Xu, Y.; Dabo, A.J.; Geraghty, P.; Campos, M.; Foronjy, R.; Garcia-Arcos, I. Decreased surfactant lipids correlate with lung function in chronic obstructive pulmonary disease (COPD). PLoS ONE 2020, 15, e0228279. [Google Scholar] [CrossRef]
  30. Morissette, M.C.; Shen, P.; Thayaparan, D.; Stampfli, M.R. Disruption of pulmonary lipid homeostasis drives cigarette smoke-induced lung inflammation in mice. Eur. Respir. J. 2015, 46, 1451–1460. [Google Scholar] [CrossRef] [Green Version]
  31. Lopez-Rodriguez, E.; Perez-Gil, J. Structure-function relationships in pulmonary surfactant membranes: From biophysics to therapy. Biochim. Biophys. Acta 2014, 1838, 1568–1585. [Google Scholar] [CrossRef]
  32. Hughes, D.A.; Haslam, P.L. Effect of smoking on the lipid composition of lung lining fluid and relationship between immunostimulatory lipids, inflammatory cells and foamy macrophages in extrinsic allergic alveolitis. Eur. Respir. J. 1990, 3, 1128–1139. [Google Scholar] [CrossRef] [PubMed]
  33. Jubinville, E.; Talbot, M.; Berube, J.C.; Hamel-Auger, M.; Maranda-Robitaille, M.; Beaulieu, M.J.; Aubin, S.; Pare, M.E.; Kallend, D.G.; Arsenault, B.; et al. Interplay between cigarette smoking and pulmonary reverse lipid transport. Eur. Respir. J. 2017, 50, 1700681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sonett, J.; Goldklang, M.; Sklepkiewicz, P.; Gerber, A.; Trischler, J.; Zelonina, T.; Westerterp, M.; Lemaitre, V.; Okada, Y.; D’Armiento, J. A critical role for ABC transporters in persistent lung inflammation in the development of emphysema after smoke exposure. FASEB J. 2018, 32, 6724. [Google Scholar] [CrossRef] [PubMed]
  35. Anzueto, A.; Jubran, A.; Ohar, J.A.; Piquette, C.A.; Rennard, S.I.; Colice, G.; Pattishall, E.N.; Barrett, J.; Engle, M.; Perret, K.A.; et al. Effects of aerosolized surfactant in patients with stable chronic bronchitis: A prospective randomized controlled trial. JAMA 1997, 278, 1426–1431. [Google Scholar] [CrossRef] [PubMed]
  36. Tall, A.R.; Yvan-Charvet, L. Cholesterol, inflammation and innate immunity. Nat. Rev. Immunol. 2015, 15, 104–116. [Google Scholar] [CrossRef] [Green Version]
  37. Cirillo, D.J.; Agrawal, Y.; Cassano, P.A. Lipids and pulmonary function in the Third National Health and Nutrition Examination Survey. Am. J. Epidemiol. 2002, 155, 842–848. [Google Scholar] [CrossRef] [Green Version]
  38. Yvan-Charvet, L.; Ranalletta, M.; Wang, N.; Han, S.; Terasaka, N.; Li, R.; Welch, C.; Tall, A.R. Combined deficiency of ABCA1 and ABCG1 promotes foam cell accumulation and accelerates atherosclerosis in mice. J. Clin. Investig. 2007, 117, 3900–3908. [Google Scholar] [CrossRef] [Green Version]
  39. Annema, W.; Nijstad, N.; Tolle, M.; de Boer, J.F.; Buijs, R.V.; Heeringa, P.; van der Giet, M.; Tietge, U.J. Myeloperoxidase and serum amyloid A contribute to impaired in vivo reverse cholesterol transport during the acute phase response but not group IIA secretory phospholipase A(2). J. Lipid Res. 2010, 51, 743–754. [Google Scholar] [CrossRef] [Green Version]
  40. Higham, A.; Lea, S.; Plumb, J.; Maschera, B.; Simpson, K.; Ray, D.; Singh, D. The role of the liver X receptor in chronic obstructive pulmonary disease. Respir. Res. 2013, 14, 106. [Google Scholar] [CrossRef] [Green Version]
  41. Gowdy, K.M.; Fessler, M.B. Emerging roles for cholesterol and lipoproteins in lung disease. Pulm. Pharmacol. Ther. 2013, 26, 430–437. [Google Scholar] [CrossRef]
  42. Fujii, W.; Kapellos, T.S.; Bassler, K.; Handler, K.; Holsten, L.; Knoll, R.; Warnat-Herresthal, S.; Oestreich, M.; Hinkley, E.R.; Hasenauer, J.; et al. Alveolar macrophage transcriptomic profiling in COPD shows major lipid metabolism changes. ERJ Open Res. 2021, 7, 00915-2020. [Google Scholar] [CrossRef] [PubMed]
  43. Murphy, A.J.; Akhtari, M.; Tolani, S.; Pagler, T.; Bijl, N.; Kuo, C.L.; Wang, M.; Sanson, M.; Abramowicz, S.; Welch, C.; et al. ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J. Clin. Investig. 2011, 121, 4138–4149. [Google Scholar] [CrossRef] [Green Version]
  44. Tolani, S.; Pagler, T.A.; Murphy, A.J.; Bochem, A.E.; Abramowicz, S.; Welch, C.; Nagareddy, P.R.; Holleran, S.; Hovingh, G.K.; Kuivenhoven, J.A.; et al. Hypercholesterolemia and reduced HDL-C promote hematopoietic stem cell proliferation and monocytosis: Studies in mice and FH children. Atherosclerosis 2013, 229, 79–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Wilhelm, L.P.; Wendling, C.; Vedie, B.; Kobayashi, T.; Chenard, M.P.; Tomasetto, C.; Drin, G.; Alpy, F. STARD3 mediates endoplasmic reticulum-to-endosome cholesterol transport at membrane contact sites. EMBO J. 2017, 36, 1412–1433. [Google Scholar] [CrossRef] [PubMed]
  46. Borthwick, F.; Allen, A.M.; Taylor, J.M.; Graham, A. Overexpression of STARD3 in human monocyte/macrophages induces an anti-atherogenic lipid phenotype. Clin. Sci. 2010, 119, 265–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Lei, C.; Xueming, H.; Ruihang, D. MLN64 deletion suppresses RANKL-induced osteoclastic differentiation and attenuates diabetic osteoporosis in streptozotocin (STZ)-induced mice. Biochem. Biophys. Res. Commun. 2018, 505, 1228–1235. [Google Scholar] [CrossRef]
  48. White, M.M.; Geraghty, P.; Hayes, E.; Cox, S.; Leitch, W.; Alfawaz, B.; Lavelle, G.M.; McElvaney, O.J.; Flannery, R.; Keenan, J.; et al. Neutrophil Membrane Cholesterol Content is a Key Factor in Cystic Fibrosis Lung Disease. EBioMedicine 2017, 23, 173–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Garcia-Fojeda, B.; Gonzalez-Carnicero, Z.; de Lorenzo, A.; Minutti, C.M.; de Tapia, L.; Euba, B.; Iglesias-Ceacero, A.; Castillo-Lluva, S.; Garmendia, J.; Casals, C. Lung Surfactant Lipids Provide Immune Protection Against Haemophilus influenzae Respiratory Infection. Front. Immunol. 2019, 10, 458. [Google Scholar] [CrossRef] [Green Version]
  50. Turley, S.D.; Andersen, J.M.; Dietschy, J.M. Rates of sterol synthesis and uptake in the major organs of the rat in vivo. J. Lipid Res. 1981, 22, 551–569. [Google Scholar] [CrossRef]
  51. McDonald, J.G.; Russell, D.W. Editorial: 25-Hydroxycholesterol: A new life in immunology. J. Leukoc. Biol. 2010, 88, 1071–1072. [Google Scholar] [CrossRef]
  52. Sugiura, H.; Koarai, A.; Ichikawa, T.; Minakata, Y.; Matsunaga, K.; Hirano, T.; Akamatsu, K.; Yanagisawa, S.; Furusawa, M.; Uno, Y.; et al. Increased 25-hydroxycholesterol concentrations in the lungs of patients with chronic obstructive pulmonary disease. Respirology 2012, 17, 533–540. [Google Scholar] [CrossRef] [PubMed]
  53. Gold, E.S.; Diercks, A.H.; Podolsky, I.; Podyminogin, R.L.; Askovich, P.S.; Treuting, P.M.; Aderem, A. 25-Hydroxycholesterol acts as an amplifier of inflammatory signaling. Proc. Natl. Acad. Sci. USA 2014, 111, 10666–10671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Wang, S.; Li, W.; Hui, H.; Tiwari, S.K.; Zhang, Q.; Croker, B.A.; Rawlings, S.; Smith, D.; Carlin, A.F.; Rana, T.M. Cholesterol 25-Hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterol. EMBO J. 2020, 39, e106057. [Google Scholar] [CrossRef] [PubMed]
  55. Clayton, S.A.; MacDonald, L.; Kurowska-Stolarska, M.; Clark, A.R. Mitochondria as Key Players in the Pathogenesis and Treatment of Rheumatoid Arthritis. Front. Immunol. 2021, 12, 673916. [Google Scholar] [CrossRef]
  56. Meyers, A.K.; Zhu, X. The NLRP3 Inflammasome: Metabolic Regulation and Contribution to Inflammaging. Cells 2020, 9, 1808. [Google Scholar] [CrossRef]
  57. Torres, S.; Solsona-Vilarrasa, E.; Nunez, S.; Matias, N.; Insausti-Urkia, N.; Castro, F.; Casasempere, M.; Fabrias, G.; Casas, J.; Enrich, C.; et al. Acid ceramidase improves mitochondrial function and oxidative stress in Niemann-Pick type C disease by repressing STARD1 expression and mitochondrial cholesterol accumulation. Redox Biol. 2021, 45, 102052. [Google Scholar] [CrossRef]
  58. Paradies, G.; Paradies, V.; Ruggiero, F.M.; Petrosillo, G. Oxidative stress, cardiolipin and mitochondrial dysfunction in nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 14205–14218. [Google Scholar] [CrossRef]
  59. Gu, R.X.; Baoukina, S.; Tieleman, D.P. Cholesterol Flip-Flop in Heterogeneous Membranes. J. Chem. Theory Comput. 2019, 15, 2064–2070. [Google Scholar] [CrossRef] [Green Version]
  60. Flaquer, A.; Rospleszcz, S.; Reischl, E.; Zeilinger, S.; Prokisch, H.; Meitinger, T.; Meisinger, C.; Peters, A.; Waldenberger, M.; Grallert, H.; et al. Mitochondrial GWA Analysis of Lipid Profile Identifies Genetic Variants to Be Associated with HDL Cholesterol and Triglyceride Levels. PLoS ONE 2015, 10, e0126294. [Google Scholar] [CrossRef]
  61. Kang, M.J.; Shadel, G.S. A Mitochondrial Perspective of Chronic Obstructive Pulmonary Disease Pathogenesis. Tuberc. Respir. Dis. 2016, 79, 207–213. [Google Scholar] [CrossRef]
  62. Hoffmann, R.F.; Zarrintan, S.; Brandenburg, S.M.; Kol, A.; de Bruin, H.G.; Jafari, S.; Dijk, F.; Kalicharan, D.; Kelders, M.; Gosker, H.R.; et al. Prolonged cigarette smoke exposure alters mitochondrial structure and function in airway epithelial cells. Respir. Res. 2013, 14, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Mizumura, K.; Cloonan, S.M.; Nakahira, K.; Bhashyam, A.R.; Cervo, M.; Kitada, T.; Glass, K.; Owen, C.A.; Mahmood, A.; Washko, G.R.; et al. Mitophagy-dependent necroptosis contributes to the pathogenesis of COPD. J. Clin. Investig. 2014, 124, 3987–4003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cloonan, S.M.; Glass, K.; Laucho-Contreras, M.E.; Bhashyam, A.R.; Cervo, M.; Pabon, M.A.; Konrad, C.; Polverino, F.; Siempos, I.I.; Perez, E.; et al. Mitochondrial iron chelation ameliorates cigarette smoke-induced bronchitis and emphysema in mice. Nat. Med. 2016, 22, 163–174. [Google Scholar] [CrossRef] [PubMed]
  65. Belchamber, K.B.R.; Singh, R.; Batista, C.M.; Whyte, M.K.; Dockrell, D.H.; Kilty, I.; Robinson, M.J.; Wedzicha, J.A.; Barnes, P.J.; Donnelly, L.E.; et al. Defective bacterial phagocytosis is associated with dysfunctional mitochondria in COPD macrophages. Eur. Respir. J. 2019, 54, 1802244. [Google Scholar] [CrossRef] [PubMed]
  66. Wu, G.; Zhu, Q.; Zeng, J.; Gu, X.; Miao, Y.; Xu, W.; Lv, T.; Song, Y. Extracellular mitochondrial DNA promote NLRP3 inflammasome activation and induce acute lung injury through TLR9 and NF-kappaB. J. Thorac. Dis. 2019, 11, 4816–4828. [Google Scholar] [CrossRef] [PubMed]
  67. West, A.P.; Khoury-Hanold, W.; Staron, M.; Tal, M.C.; Pineda, C.M.; Lang, S.M.; Bestwick, M.; Duguay, B.A.; Raimundo, N.; MacDuff, D.A.; et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 2015, 520, 553–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Zhang, W.Z.; Rice, M.C.; Hoffman, K.L.; Oromendia, C.; Barjaktarevic, I.Z.; Wells, J.M.; Hastie, A.T.; Labaki, W.W.; Cooper, C.B.; Comellas, A.P.; et al. Association of urine mitochondrial DNA with clinical measures of COPD in the SPIROMICS cohort. JCI Insight 2020, 5, e133984. [Google Scholar] [CrossRef] [Green Version]
  69. Lloreta, J.; Orozco, M.; Gea, J.; Corominas, J.M.; Serrano, S. Selective diaphragmatic mitochondrial abnormalities in a patient with marked air flow obstruction. Ultrastruct. Pathol. 1996, 20, 67–71. [Google Scholar] [CrossRef]
  70. Angelidis, I.; Simon, L.M.; Fernandez, I.E.; Strunz, M.; Mayr, C.H.; Greiffo, F.R.; Tsitsiridis, G.; Ansari, M.; Graf, E.; Strom, T.M.; et al. An atlas of the aging lung mapped by single cell transcriptomics and deep tissue proteomics. Nat. Commun. 2019, 10, 963. [Google Scholar] [CrossRef] [Green Version]
  71. Fahndrich, S.; Biertz, F.; Karch, A.; Kleibrink, B.; Koch, A.; Teschler, H.; Welte, T.; Kauczor, H.U.; Janciauskiene, S.; Jorres, R.A.; et al. Cardiovascular risk in patients with alpha-1-antitrypsin deficiency. Respir. Res. 2017, 18, 171. [Google Scholar] [CrossRef]
  72. Pontzer, H.; Yamada, Y.; Sagayama, H.; Ainslie, P.N.; Andersen, L.F.; Anderson, L.J.; Arab, L.; Baddou, I.; Bedu-Addo, K.; Blaak, E.E.; et al. Daily energy expenditure through the human life course. Science 2021, 373, 808–812. [Google Scholar] [CrossRef] [PubMed]
  73. Majid, S.; Keith, R.J.; Fetterman, J.L.; Weisbrod, R.M.; Nystoriak, J.; Wilson, T.; Stokes, A.C.; Blaha, M.J.; Srivastava, S.; Robertson, R.M.; et al. Lipid profiles in users of combustible and electronic cigarettes. Vasc. Med. 2021, 26, 483–488. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Possible cholesterol signaling that contributes to mitochondrial dysfunction, inflammation, and loss of lung function in COPD. Created with BioRender.com.
Figure 1. Possible cholesterol signaling that contributes to mitochondrial dysfunction, inflammation, and loss of lung function in COPD. Created with BioRender.com.
Medicina 59 00253 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jundi, B.; Ahmed, H.; Reece, J.; Geraghty, P. The Relationship of Cholesterol Responses to Mitochondrial Dysfunction and Lung Inflammation in Chronic Obstructive Pulmonary Disease. Medicina 2023, 59, 253. https://doi.org/10.3390/medicina59020253

AMA Style

Jundi B, Ahmed H, Reece J, Geraghty P. The Relationship of Cholesterol Responses to Mitochondrial Dysfunction and Lung Inflammation in Chronic Obstructive Pulmonary Disease. Medicina. 2023; 59(2):253. https://doi.org/10.3390/medicina59020253

Chicago/Turabian Style

Jundi, Bakr, Huma Ahmed, Joshua Reece, and Patrick Geraghty. 2023. "The Relationship of Cholesterol Responses to Mitochondrial Dysfunction and Lung Inflammation in Chronic Obstructive Pulmonary Disease" Medicina 59, no. 2: 253. https://doi.org/10.3390/medicina59020253

Article Metrics

Back to TopTop