Advertisement
Heart, Lung and Circulation
Review| Volume 32, ISSUE 1, P26-42, January 2023

Potential Biological Mediators of Myocardial and Vascular Complications of Air Pollution—A State-of-the-Art Review

  • Sina Fathieh
    Affiliations
    Kolling Institute of Medical Research, Sydney, NSW, Australia

    Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia

    Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
    Search for articles by this author
  • Stuart M. Grieve
    Affiliations
    Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia

    Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia

    Department of Radiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
    Search for articles by this author
  • Kazuaki Negishi
    Affiliations
    Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia

    Department of Cardiology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan

    Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia

    Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia
    Search for articles by this author
  • Gemma A. Figtree
    Correspondence
    Corresponding author at: Gemma Figtree, Interventional Cardiologist, Royal North Shore Hospital, Research Lead, Cardiovascular Discovery Group, Kolling Institute, University of Sydney, Level 12, Kolling Institute of Medical Research, 10 Westbourne Street, St Leonards, 2065, NSW, Australia.
    Affiliations
    Kolling Institute of Medical Research, Sydney, NSW, Australia

    Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia

    Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia

    Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
    Search for articles by this author
Published:December 28, 2022DOI:https://doi.org/10.1016/j.hlc.2022.11.014
      Ambient air pollution is recognised globally as a significant contributor to the burden of cardiovascular diseases. The evidence from both human and animal studies supporting the cardiovascular impact of exposure to air pollution has grown substantially, implicating numerous pathophysiological pathways and related signalling mediators. In this review, we summarise the list of activated mediators for each pathway that lead to myocardial and vascular injury in response to air pollutants. We performed a systematic search of multiple databases, including articles between 1990 and Jan 2022, summarising the evidence for activated pathways in response to each significant air pollutant. Particulate matter <2.5 μm (PM2.5) was the most studied pollutant, followed by particulate matter between 2.5 μm–10 μm (PM10), nitrogen dioxide (NO2) and ozone (O3). Key pathogenic pathways that emerged included activation of systemic and local inflammation, oxidative stress, endothelial dysfunction, and autonomic dysfunction. We looked at how potential mediators of each of these pathways were linked to both cardiovascular disease and air pollution and included the overlapping mediators. This review illustrates the complex relationship between air pollution and cardiovascular diseases, and discusses challenges in moving beyond associations, towards understanding causal contributions of specific pathways and markers that may inform us regarding an individual’s exposure, response, and likely risk.

      Keywords

      Introduction

      Public health efforts since the 1950s have resulted in an up to 80% reduction in the concentration of major air pollutants in megacities [
      • Fowler D.
      • Brimblecombe P.
      • Burrows J.
      • Heal M.R.
      • Grennfelt P.
      • Stevenson D.S.
      • et al.
      A chronology of global air quality.
      ]. However, outdoor ambient air pollution remains a leading environmental cause of cardiovascular morbidity and mortality, making it a significant public health consideration for policymakers [
      • Daiber A.
      • Lelieveld J.
      • Steven S.
      • Oelze M.
      • Kröller-Schön S.
      • Sørensen M.
      • et al.
      The “exposome” concept—how environmental risk factors influence cardiovascular health.
      ]. Particle matters with an aerodynamic diameter of less than 2.5 μm (PM2.5), and between 2.5 μm–10 μm (PM10), ozone (O3) and nitrogen dioxide (NO2) are amongst the major known hazardous air pollutants (HAP) that can impact cardiovascular health in the acute setting as well as chronic exposure.
      In 2015 alone, exposure to PM2.5 was found to be responsible for more than 4 million deaths, up from 3.5 million in 1990, putting it at the fifth highest ranking risk factor for mortality [
      • Cohen A.J.
      • Brauer M.
      • Burnett R.
      • Anderson H.R.
      • Frostad J.
      • Estep K.
      • et al.
      Estimates and 25-year trends of the global burden of disease attributable to ambient air pollution: an analysis of data from the Global Burden of Diseases Study 2015.
      ]. High-income countries comprised only 1% of deaths related to household air pollution but 12% of ambient air pollution-related mortality in that year. Other studies show direct causation on mortality and comorbidities, including a range of cardiovascular diseases [
      • Chan S.H.
      • Van Hee V.C.
      • Bergen S.
      • Szpiro A.A.
      • DeRoo L.A.
      • London S.J.
      • et al.
      Long-term air pollution exposure and blood pressure in the sister study.
      ,
      • Link M.S.
      • Dockery D.W.
      Air pollution and the triggering of cardiac arrhythmias.
      ,
      • Gold D.R.
      • Samet J.M.
      Air pollution, climate, and heart disease.
      ,
      • Newby D.E.
      • Mannucci P.M.
      • Tell G.S.
      • Baccarelli A.A.
      • Brook R.D.
      • Donaldson K.
      • et al.
      Expert position paper on air pollution and cardiovascular disease.
      ,
      • Beelen R.
      • Stafoggia M.
      • Raaschou-Nielsen O.
      • Andersen Z.J.
      • Xun W.W.
      • Katsouyanni K.
      • et al.
      Long-term exposure to air pollution and cardiovascular mortality: an analysis of 22 European cohorts.
      ], most notably coronary artery disease (CAD) [
      • Wang X.
      • Kindzierski W.
      • Kaul P.
      Air pollution and acute myocardial infarction hospital admission in Alberta, Canada: a three-step procedure case-crossover study.
      ,
      • Su C.
      • Breitner S.
      • Schneider A.
      • Liu L.
      • Franck U.
      • Peters A.
      • et al.
      Short-term effects of fine particulate air pollution on cardiovascular hospital emergency room visits: a time-series study in Beijing, China.
      ,
      • Weichenthal S.
      • Lavigne E.
      • Evans G.
      • Pollitt K.
      • Burnett R.T.
      Ambient PM2.5 and risk of emergency room visits for myocardial infarction: impact of regional PM2.5 oxidative potential: a case-crossover study.
      ], lung cancer [
      • Hamra G.B.
      • Guha N.
      • Cohen A.
      • Laden F.
      • Raaschou-Nielsen O.
      • Samet J.M.
      • et al.
      Outdoor particulate matter exposure and lung cancer: a systematic review and meta-analysis.
      ], diabetes [
      • Meo S.A.
      • Memon A.N.
      • Sheikh S.A.
      • Rouq F.A.
      • Usmani A.M.
      • Hassan A.
      • et al.
      Effect of environmental air pollution on type 2 diabetes mellitus.
      ] and stroke [
      • Stafoggia M.
      • Cesaroni G.
      • Peters A.
      • Andersen Z.J.
      • Badaloni C.
      • Beelen R.
      • et al.
      Long-term exposure to ambient air pollution and incidence of cerebrovascular events: results from 11 European cohorts within the ESCAPE project.
      ], to name a few. In addition to health impacts, air pollution causes significant direct and indirect economic burdens globally [
      • Landrigan P.J.
      • Fuller R.
      • Acosta N.J.R.
      • Adeyi O.
      • Arnold R.
      • Basu N.
      • et al.
      The Lancet Commission on pollution and health.
      ]. Furthermore, the latest advice from the World Health Organization (WHO) advises that there is no safe level for PM2.5 [
      • Daiber A.
      • Lelieveld J.
      • Steven S.
      • Oelze M.
      • Kröller-Schön S.
      • Sørensen M.
      • et al.
      The “exposome” concept—how environmental risk factors influence cardiovascular health.
      ,
      • Vineis P.
      What Is the exposome and how it can help research on air pollution.
      ].
      Studies focussing on the mechanism behind air pollution-related cardiovascular disease (CVD) since the 1980s have implicated the role of systemic and local inflammation, oxidative stress, vascular endothelial damage, plaque vulnerability, hypercoagulable state and autonomic dysfunction as some of the involved pathophysiological pathways. However, the exact mechanisms by which these pathways are activated remain a “hot” topic in environmental health studies. In this context, the role of cardio-respiratory interaction needs further research. The passage of smaller particulate matters from larger airways to alveoli and across the capillary bed to the systemic circulation is thought to cause local inflammation via local endothelial pathways and systemic inflammation by activation of airway receptors. However, little is known about the fate of micro-particulate matters in the bloodstream. Most of the biomarkers that are used for these studies are end-products of these processes, reflecting activity of these signalling pathways. Whilst they have been implicated in both cardiovascular disease processes and seen to be elevated in association with air pollution, the direct causal roles are less clear.
      Biological mediators have a new-found home in the systems biology model of diseases. The concepts of exposome (the measure of all the exposures of an individual in a lifetime, and how these exposures relate to health) and pollutome (the totality of all forms of pollution that have the potential to harm human health) aim to complement these models by addressing the interaction between the environment and an individual’s building blocks that results in diseases. It is therefore pertinent to have a summary of the available evidence for all the implicated biomarkers to pave the way for future research in this field.

      Aims

      For this review, we aimed to collect all the available data from human and animal studies for potential mediators involved in the pathogenesis of myocardial and vascular injuries and to categorise them based on their mechanism of action.

      Method

      Essential questions in this investigation before identifying potential biomarkers were:
      • 1.
        What are primary myocardial and vascular injuries attributed to acute or chronic air pollution exposure?
      • 2.
        Which air pollutants are implicated in the pathogenesis of those pathologies?What are the primary mechanisms by which these air pollutants mediate their injury?
      • 3.
        What are the mediators released after exposure to air pollutants that activate these pathological pathways that may act as markers of individual susceptibility and higher personal risk of cardiovascular complications?
      We examined all systematic reviews since 2015 to have the most recent summary of evidence for each sub-topic of interest and included relevant papers from 1990 on, since most technological advancements in detection and quantification of bio-mediators started in this period. Our choices of search engines were Google Scholar for its superior indexing and search algorithms, and PubMed for its greater number of indexed papers.
      Review papers addressing the first question were summarised first to allow a more targeted search for the next steps. Identified mechanisms of injury were atherosclerosis formation and progression leading to coronary artery disease (CAD) and acute coronary syndrome (ACS), congestive cardiac failure (CCF), cardiomyopathy, and pulmonary hypertension (type 1, 2 and 3). To identify relevant HAPs (essential question 2), we used the individual results from the previous step along with the individual pollutant’s name. A HAP was considered relevant if there was at least one review article or human study addressing that HAP—these included PM2.5, PM10, NO2 and O3. Finally, to generate the list of mediators that need to be included, we used the papers that were filtered in question 1 and question 2 and summarised the described pathways linking the identified HAPs to cardiovascular pathologies of interest. Our final list of pathophysiological pathways included systemic inflammation as the leading mechanism, along with local tissue inflammation, such as adipose tissue inflammation, that leads to the production of harmful adipokines, oxidative stress and mitochondrial dysfunction, endothelial dysfunction, impaired lipid metabolism, thrombogenesis, increased plaque vulnerability and autonomic dysfunction.
      We then identified individual mediators linked to cardiovascular injury for each pathway by searching for the pathway combined with Boolean keywords + or AND, along with terms cardiovascular disease, ischaemic heart disease, myocardial infarction, coronary artery disease, congestive cardiac failure, congestive heart failure, atherosclerosis, cardiomyopathy, and vascular disease. Using this method allowed us to have a more comprehensive list of mediators to rule out, one by one, by performing inquiries in our elected search engines using individual mediators, with the keywords Air Pollution, Particulate Matter 2.5, Particulate Matter 10, PM2.5, PM10, nitrogen dioxide, carbon dioxide and ozone. To better categorise the final set of mediators, we grouped them into inflammatory and non-inflammatory classes. We then grouped these papers based on individual bio-mediators or, in some instances, the covered pathways. Older papers that had been replaced by newer, more up-to-date evidence, and papers with similar findings were reviewed as a group, and papers that had the best evidence and were most up to date were kept.
      This method was chosen to have a maximal screening potential. Potential sources of assessment bias, however, include our reliance on review articles as the initial screening step, which seemed to primarily focus on the effects of PM2.5 and man-made sources of air pollution. Our complete search strategy is outlined in Supplementary material.

      Results

      Figure 1 outlines the potential biomarkers of myocardial and vascular complications of air pollution and their mechanism of injury.
      Figure thumbnail gr1
      Figure 1Potential biomarkers of myocardial and vascular complications of air pollution and their mechanisms of injury.

      Results, Part A: Inflammatory Mediators

      Systemic Inflammation

      The inflammatory response is triggered in response to tissue injuries as well as internal and external factors that get flagged as potentially harmful. These responses are classed as either acute or chronic, each with a specific set of mediators which result in cellular and vascular events. As shown in Table 1, air pollutants can result in the release of a range of mediators into the systemic circulation that can result in injury to the cardiovascular system.
      Table 1Potential air pollution induced systemic inflammatory mediators.
      MediatorDescriptionTriggersEffect on CVD
      Tumour Necrosis Factor-α (TNF-α)An important pro-inflammatory cytokine released by monocytes and macrophages.

      It has been implicated in a range of autoimmune conditions such as rheumatoid arthritis (RA).
      PM2.5 [
      • Pozzi R.
      • De Berardis B.
      • Paoletti L.
      • Guastadisegni C.
      Inflammatory mediators induced by coarse (PM2.5–10) and fine (PM2.5) urban air particles in RAW 264.7 cells.
      ]

      PM10 [
      • van Eeden S.F.
      • Tan W.C.
      • Suwa T.
      • Mukae H.
      • Terashima T.
      • Fujii T.
      • et al.
      Cytokines involved in the systemic inflammatory response induced by exposure to particulate matter air pollutants (PM10).
      ]

      O3 [
      • Fakhrzadeh L.
      • Laskin J.D.
      • Laskin D.L.
      Ozone-induced production of nitric oxide and TNF-α and tissue injury are dependent on NF-κB p50.
      ]

      NO2 [
      • Li H.
      • Han M.
      • Guo L.
      • Li G.
      • Sang N.
      Oxidative stress, endothelial dysfunction and inflammatory response in rat heart to NO2 inhalation exposure.
      ]
      CAD [
      • Tuomisto K.
      • Jousilahti P.
      • Sundvall J.
      • Pajunen P.
      • Salomaa V.
      C-reactive protein, interleukin-6 and tumor necrosis factor alpha as predictors of incident coronary and cardiovascular events and total mortality: a population-based, prospective study.
      ]

      CCF [
      • Vasan R.S.
      • Sullivan L.M.
      • Roubenoff R.
      • Dinarello C.A.
      • Harris T.
      • Benjamin E.J.
      • et al.
      Inflammatory markers and risk of heart failure in elderly subjects without prior myocardial infarction: the Framingham Heart Study.
      ]

      Lipid metabolism, insulin resistance, endothelial dysfunction, ROS formation and hypercoagulable state [
      • Yuan S.
      • Carter P.
      • Bruzelius M.
      • Vithayathil M.
      • Kar S.
      • Mason A.M.
      • et al.
      Effects of tumour necrosis factor on cardiovascular disease and cancer: a two-sample Mendelian randomization study.
      ]
      Interleukin 1 beta (IL-1 β)Part of the IL-1 family that is released systemically from monocytes, smooth muscle cells and endothelial cells in response to microbial products and endogenous triggers that act on IL-1 receptors.PM2.5 [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]

      PM10 [
      • van Eeden S.F.
      • Tan W.C.
      • Suwa T.
      • Mukae H.
      • Terashima T.
      • Fujii T.
      • et al.
      Cytokines involved in the systemic inflammatory response induced by exposure to particulate matter air pollutants (PM10).
      ]

      NO2 [
      • Li H.
      • Han M.
      • Guo L.
      • Li G.
      • Sang N.
      Oxidative stress, endothelial dysfunction and inflammatory response in rat heart to NO2 inhalation exposure.
      ]

      O3 [
      • Barker J.S.
      • Wu Z.
      • Hunter D.D.
      • Dey R.D.
      Ozone exposure initiates a sequential signaling cascade in airways involving interleukin-1beta release, nerve growth factor secretion, and substance P upregulation.
      ]
      Atherogenesis by activating inflammasome-IL-1β pathway [
      • Qamar A.
      • Rader D.J.
      Effect of interleukin 1β inhibition in cardiovascular disease.
      ]
      Interleukin-2 (IL-2)Responsible for eliminating diseased cells by contributing to the inflammatory cascade via activating CD4 and CD8 T-cells. Also has a significant role in the resolution of inflammation.Chronic HAP [
      • Tomei G.
      • Ciarrocca M.
      • Bernardini A.
      • Capozzella A.
      • Fortunato B.R.
      • Pimpinella B.
      • et al.
      Plasma IL-2, NK, IFN-γ, and C3 in male workers exposed to traffic pollutants.
      ]

      NOx [
      • Mostafavi N.
      • Vlaanderen J.
      • Chadeau-Hyam M.
      • Beelen R.
      • Modig L.
      • Palli D.
      • et al.
      Inflammatory markers in relation to long-term air pollution.
      ] ↓
      CAD [
      • Ding R.
      • Gao W.
      • Ostrodci D.H.
      • He Z.
      • Song Y.
      • Ma L.
      • et al.
      Effect of interleukin-2 level and genetic variants on coronary artery disease.
      ]

      Stronger association with stable angina than ACS [
      • Simon A.D.
      • Yazdani S.
      • Wang W.
      • Schwartz A.
      • Rabbani L.E.
      Elevated plasma levels of interleukin-2 and soluble il-2 receptor in ischemic heart disease.
      ]
      Interleukin-6 (IL-6)A pivotal multifunctional cytokine that triggers the release of many other downstream inflammatory cytokines and is the main stimulus for C-reactive protein (CRP) synthesis.PM2.5 [
      • Pozzi R.
      • De Berardis B.
      • Paoletti L.
      • Guastadisegni C.
      Inflammatory mediators induced by coarse (PM2.5–10) and fine (PM2.5) urban air particles in RAW 264.7 cells.
      ] PM10 [
      • van Eeden S.F.
      • Tan W.C.
      • Suwa T.
      • Mukae H.
      • Terashima T.
      • Fujii T.
      • et al.
      Cytokines involved in the systemic inflammatory response induced by exposure to particulate matter air pollutants (PM10).
      ] O3 [
      • Mirowsky J.E.
      • Dailey L.A.
      • Devlin R.B.
      Differential expression of pro-inflammatory and oxidative stress mediators induced by nitrogen dioxide and ozone in primary human bronchial epithelial cells.
      ] SO2 [
      • Panasevich S.
      • Leander K.
      • Rosenlund M.
      • Ljungman P.
      • Bellander T.
      • de Faire U.
      • et al.
      Associations of long- and short-term air pollution exposure with markers of inflammation and coagulation in a population sample.
      ] NO2 [
      • Perret J.L.
      • Bowatte G.
      • Lodge C.J.
      • Knibbs L.D.
      • Gurrin L.C.
      • Kandane-Rathnayake R.
      • et al.
      The dose–response association between nitrogen dioxide exposure and serum interleukin-6 concentrations.
      ]
      CAD with the same weight as traditional risk factors such as HTN [
      • Perret J.L.
      • Bowatte G.
      • Lodge C.J.
      • Knibbs L.D.
      • Gurrin L.C.
      • Kandane-Rathnayake R.
      • et al.
      The dose–response association between nitrogen dioxide exposure and serum interleukin-6 concentrations.
      ]

      Levels predictor of worse outcomes [
      • Ridker P.M.
      • Rane M.
      Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease.
      ]
      Interleukin-8 (IL-8)A small cytokine that belongs to the CXC subfamily, which is released primarily from monocytes and macrophages, leading to the attraction of other inflammatory cells towards the inflammatory site as well as activation of monocytes and neutrophils. IL-8 has a central role in the inflammatory cascade at the site of coronary artery plaque formation [
      • Apostolakis S.
      • Vogiatzi K.
      • Amanatidou V.
      • Spandidos D.A.
      Interleukin 8 and cardiovascular disease.
      ].
      PM2.5 [
      • Yan Z.
      • Wang J.
      • Li J.
      • Jiang N.
      • Zhang R.
      • Yang W.
      • et al.
      Oxidative stress and endocytosis are involved in upregulation of interleukin-8 expression in airway cells exposed to PM2.5.
      ] UFP [
      • Kim Y.-M.
      • Reed W.
      • Lenz A.G.
      • Jaspers I.
      • Silbajoris R.
      • Nick H.S.
      • et al.
      Ultrafine carbon particles induce interleukin-8 gene transcription and p38 MAPK activation in normal human bronchial epithelial cells.
      ]

      PM10 [
      • Silbajoris R.
      • Osornio-Vargas Alvaro R.
      • Simmons Steven O.
      • Reed W.
      • Bromberg Philip A.
      • Dailey Lisa A.
      • et al.
      Ambient particulate matter induces interleukin-8 expression through an alternative NF-κB (nuclear factor-kappa b) mechanism in human airway epithelial cells.
      ]

      O3 [
      • Mirowsky J.E.
      • Dailey L.A.
      • Devlin R.B.
      Differential expression of pro-inflammatory and oxidative stress mediators induced by nitrogen dioxide and ozone in primary human bronchial epithelial cells.
      ]

      Iron particles [
      • Smith K.R.
      • Veranth J.M.
      • Hu A.A.
      • Lighty J.S.
      • Aust A.E.
      Interleukin-8 levels in human lung epithelial cells are increased in response to coal fly ash and vary with the bioavailability of iron, as a function of particle size and source of coal.
      ]
      Inflammatory cascade at the site of coronary artery plaque formation [
      • Apostolakis S.
      • Vogiatzi K.
      • Amanatidou V.
      • Spandidos D.A.
      Interleukin 8 and cardiovascular disease.
      ].

      Chemotactic effects on VSMC and inhibition of local TIMP-1 expression contributing to atherosclerosis formation [
      • Apostolakis S.
      • Vogiatzi K.
      • Amanatidou V.
      • Spandidos D.A.
      Interleukin 8 and cardiovascular disease.
      ].
      Interleukin-10 (IL-10)Anti-inflammatory cytokine by inhibiting inflammatory response from macrophages, antigen presentation and proliferation and inhibiting Th1 T cells.PM2.5 [
      • Wan Q.
      • Liu Z.
      • Yang M.
      • Wu J.
      Acceleratory effects of ambient fine particulate matter on the development and progression of atherosclerosis in apolipoprotein E knockout mice by down-regulating CD4+CD25+Foxp3+ regulatory T cells.
      ] ↓

      NOx [
      • Mostafavi N.
      • Vlaanderen J.
      • Chadeau-Hyam M.
      • Beelen R.
      • Modig L.
      • Palli D.
      • et al.
      Inflammatory markers in relation to long-term air pollution.
      ] ↓
      Anti-atherogenic [
      • Welsh P.
      • Murray H.M.
      • Ford I.
      • Trompet S.
      • de Craen A.J.M.
      • Jukema J.W.
      • et al.
      Circulating interleukin-10 and risk of cardiovascular events.
      ]
      Granulocyte macrophage colony-stimulating factor

      (GM-CSF)
      A multifunctional cytokine that controls all phases of the leukocyte life cycle from production to proliferation and differentiation.PM10 [
      • van Eeden S.F.
      • Tan W.C.
      • Suwa T.
      • Mukae H.
      • Terashima T.
      • Fujii T.
      • et al.
      Cytokines involved in the systemic inflammatory response induced by exposure to particulate matter air pollutants (PM10).
      ]

      O3 [
      • Rusznak C.
      • Devalia J.L.
      • Sapsford R.J.
      • Davies R.J.
      Ozone-induced mediator release from human bronchial epithelial cells in vitro and the influence of nedocromil sodium.
      ]

      NO2 [
      • Devalia J.L.
      • Campbell A.M.
      • Sapsford R.J.
      • Rusznak C.
      • Quint D.
      • Godard P.
      • et al.
      Effect of nitrogen dioxide on synthesis of inflammatory cytokines expressed by human bronchial epithelial cells in vitro.
      ]
      MI [
      • Anzai A.
      • Choi J.L.
      • He S.
      • Fenn A.M.
      • Nairz M.
      • Rattik S.
      • et al.
      The infarcted myocardium solicits GM-CSF for the detrimental oversupply of inflammatory leukocytes.
      ]

      Serum levels predictive of the severity of cardiac failure and myocardial remodelling [
      • Anzai A.
      • Choi J.L.
      • He S.
      • Fenn A.M.
      • Nairz M.
      • Rattik S.
      • et al.
      The infarcted myocardium solicits GM-CSF for the detrimental oversupply of inflammatory leukocytes.
      ]
      HistamineDiesel particles [
      • Nemmar A.
      • Nemery B.
      • Hoet P.H.
      • Vermylen J.
      • Hoylaerts M.F.
      Pulmonary inflammation and thrombogenicity caused by diesel particles in hamsters: role of histamine.
      ]

      PM2.5 [
      • Signorelli S.S.
      • Oliveri Conti G.
      • Zanobetti A.
      • Baccarelli A.
      • Fiore M.
      • Ferrante M.
      Effect of particulate matter-bound metals exposure on prothrombotic biomarkers: A systematic review.
      ]

      O3 [
      • Yeates D.B.
      • Mauderly J.L.
      Inhaled environmental/occupational irritants and allergens: mechanisms of cardiovascular and systemic responses. Introduction.
      ]
      Increase thrombogenicity [
      • Nemmar A.
      • Nemery B.
      • Hoet P.H.
      • Vermylen J.
      • Hoylaerts M.F.
      Pulmonary inflammation and thrombogenicity caused by diesel particles in hamsters: role of histamine.
      ]
      Toll-like receptors (TLRs)There are 13 TLRs recognised that are located on the plasma membrane or endolysosomal compartment, with the primary function of detecting ‘danger signals’ and activating a range of inflammatory mediators.Cardiomyopathy, atherosclerosis, MI, and myocardial remodelling in CCF [
      • Vallejo Jesus G.
      Role of Toll-like receptors in cardiovascular diseases.
      ]
      Insulin-like growth factor binding proteins (IGFBPs)Modulate insulin-like growth factors (IGF) binding to their receptors by binding to them and carrying them in plasma as a dimetric complex [
      • Fischer F.
      • Schulte H.
      • Mohan S.
      • Tataru M.-C.
      • Köhler E.
      • Assmann G.
      • et al.
      Associations of insulin-like growth factors, insulin-like growth factor binding proteins and acid-labile subunit with coronary heart disease.
      ].
      PM2.5 [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]
      CAD, IHD, and total mortality [
      • Fischer F.
      • Schulte H.
      • Mohan S.
      • Tataru M.-C.
      • Köhler E.
      • Assmann G.
      • et al.
      Associations of insulin-like growth factors, insulin-like growth factor binding proteins and acid-labile subunit with coronary heart disease.
      ]
      Interferon γ (IFN-γ)A central cytokine in both innate and adaptive immunity, which, by activating Janus kinases (JAKs), modulates close to 2,300 human genes.PM2.5 [
      • Ma Q.-Y.
      • Huang D.-Y.
      • Zhang H.-J.
      • Wang S.
      • Chen X.-F.
      Exposure to particulate matter 2.5 (PM2.5) induced macrophage-dependent inflammation, characterized by increased Th1/Th17 cytokine secretion and cytotoxicity.
      ]

      O3 [

      Larini A, Aldinucci C, Bocci V, editors. Ozone as a modulator of the immune system. Proceedings of the XV Ozone World Congress London; 2001.

      ]
      Atherosclerosis via formation of foam cells, VSM inflammation, the release of adhesion cells, and systemic inflammation [
      • Elyasi A.
      • Voloshyna I.
      • Ahmed S.
      • Kasselman L.J.
      • Behbodikhah J.
      • De Leon J.
      • et al.
      The role of interferon-γ in cardiovascular disease: an update.
      ]
      Transforming growth factor beta (TGF-β)A superfamily of more than 30 proteins involved in cellular growth and modulating different phases of cellular life, such as differentiation, proliferation, and apoptosis [
      • Yue Y.
      • Meng K.
      • Pu Y.
      • Zhang X.
      Transforming growth factor beta (TGF-β) mediates cardiac fibrosis and induces diabetic cardiomyopathy.
      ].
      PM2.5 [
      • Dysart M.M.
      • Galvis B.R.
      • Russell A.G.
      • Barker T.H.
      Environmental particulate (PM2. 5) augments stiffness-induced alveolar epithelial cell mechanoactivation of transforming growth factor beta.
      ]
      Cardiac fibrosis formation via SMAD dependent and independent mechanisms [
      • Yue Y.
      • Meng K.
      • Pu Y.
      • Zhang X.
      Transforming growth factor beta (TGF-β) mediates cardiac fibrosis and induces diabetic cardiomyopathy.
      ]
      Monocyte chemoattractant protein-1 (MCP-1)A very potent chemokine that belongs to the C-C chemokine family and is released in response to a range of pro-inflammatory stimuli, such as oxidative stress and growth factor, which can result in the attraction of monocytes to sub-endothelium [
      • Deshmane S.L.
      • Kremlev S.
      • Amini S.
      • Sawaya B.E.
      Monocyte chemoattractant protein-1 (MCP-1): an overview.
      ].
      PM2.5 [
      • Fu H.
      • Liu X.
      • Li W.
      • Zu Y.
      • Zhou F.
      • Shou Q.
      • et al.
      PM2.5 exposure induces inflammatory response in macrophages via the TLR4/COX-2/NF-κB pathway.
      ]
      Thrombogenicity, CAD and restenosis following balloon angioplasty [
      • Deshmane S.L.
      • Kremlev S.
      • Amini S.
      • Sawaya B.E.
      Monocyte chemoattractant protein-1 (MCP-1): an overview.
      ]
      NLRP3 inflammasomeNLRP3 belongs to the NOD-like receptor (NLR) family with a pyrin domain, which is involved in inflammasome formation, which in turn can initiate an inflammatory cascade [
      • Zhou W.
      • Chen C.
      • Chen Z.
      • Liu L.
      • Jiang J.
      • Wu Z.
      • et al.
      NLRP3: a novel mediator in cardiovascular disease.
      ].
      PM2.5 [
      • Hu T.
      • Zhu P.
      • Liu Y.
      • Zhu H.
      • Geng J.
      • Wang B.
      • et al.
      PM2 .5 induces endothelial dysfunction via activating NLRP3 inflammasome.
      ]
      Atherosclerosis, IHD and cardiomyopathy [
      • Zhou W.
      • Chen C.
      • Chen Z.
      • Liu L.
      • Jiang J.
      • Wu Z.
      • et al.
      NLRP3: a novel mediator in cardiovascular disease.
      ]
      PI3K/Akt/FOXO1 pathwayForkhead transcription factors of the O class (FOXOs), consisting of four members, are expressed in most human tissues, and play a role in various cell cycle stages and inflammation, metabolism, and stress resistance [
      • Wang Y.
      • Zhou Y.
      • Graves D.T.
      FOXO Transcription factors: their clinical significance and regulation.
      ].
      PM2.5 [
      • Su X.
      • Tian J.
      • Li B.
      • Zhou L.
      • Kang H.
      • Pei Z.
      • et al.
      Ambient PM2.5 caused cardiac dysfunction through FoxO1-targeted cardiac hypertrophy and macrophage-activated fibrosis in mice.
      ]
      Ischaemia/reperfusion injury, cardiomyopathy, and insulin insensitivity [
      • Wang Y.
      • Zhou Y.
      • Graves D.T.
      FOXO Transcription factors: their clinical significance and regulation.
      ]
      Abbreviations: PM2.5, particulate matter <2.5 μm; PM10, particulate matter <10 μm; CAD, coronary artery disease; CCF, congestive cardiac failure; ROS, reactive oxygen species; HAP, hazardous air pollutant; IHD, ischaemic heart disease; ACS, acute coronary syndrome; HTN, hypertension; UFP, ultrafine particles; VSM, vascular smooth muscle; TIMP-1, tissue inhibitor matrix metalloproteinase 1; Th1, T helper 1; MI, myocardial infarction.

      Local Tissue Inflammation

      HAP can induce local tissue inflammation via direct insult as well as indirectly by triggering the systemic inflammatory response. Besides inflammation of the lungs, that is the primary source of systemic cytokine release, the following tissues can also be inflamed following HAP exposure, further releasing harmful mediators:

      Adipose tissue inflammation

      Long-term exposure to PM2.5 can lead to adipose inflammation [
      • Sun Q.
      • Yue P.
      • Deiuliis J.A.
      • Lumeng C.N.
      • Kampfrath T.
      • Mikolaj M.B.
      • et al.
      Ambient air pollution exaggerates adipose inflammation and insulin resistance in a mouse model of diet-induced obesity.
      ], leading to the release of adipokines into the circulation. The mechanism behind this effect is not established but may be secondary to systemic inflammation in a similar manner to that proposed to occur in patients with metabolic syndrome. PM2.5 is known to alter mitochondrial activity and gene expression in brown adipose tissue (BAT) and white adipose tissue (WAT) differently, with more exaggerated changes seen in BAT—resulting in a functional mismatch and contributing to a range of metabolic syndrome features including insulin resistance and atherosclerosis [
      • Xu Z.
      • Xu X.
      • Zhong M.
      • Hotchkiss I.P.
      • Lewandowski R.P.
      • Wagner J.G.
      • et al.
      Ambient particulate air pollution induces oxidative stress and alterations of mitochondria and gene expression in brown and white adipose tissues.
      ].

      Intestinal inflammation

      Air pollution enters the gastrointestinal tract (GIT) via mucociliary clearance of inhaled air, and therefore can directly interact with the GIT epithelium to cause local, and subsequently systemic inflammation by forming reactive oxygen species (ROS) and pro-inflammatory cytokines [
      • Feng J.
      • Cavallero S.
      • Hsiai T.
      • Li R.
      Impact of air pollution on intestinal redox lipidome and microbiome.
      ].

      Perivascular adipose tissue (PVAT) inflammation

      PVAT involves adipocytes surrounding blood vessels and, in normal physiology, has a dominant anticontractile function, but in the pathophysiological state contributes to endothelial dysfunction, atherosclerosis and aortic aneurysm [
      • Haberzettl P.
      • Jin L.
      • Riggs D.W.
      • Zhao J.
      • O’Toole T.E.
      • Conklin D.J.
      Fine particulate matter air pollution and aortic perivascular adipose tissue: oxidative stress, leptin, and vascular dysfunction.
      ]. It is shown to be activated in mice exposed to PM2.5, likely by ROS formation and inflammatory changes [
      • Haberzettl P.
      • Jin L.
      • Riggs D.W.
      • Zhao J.
      • O’Toole T.E.
      • Conklin D.J.
      Fine particulate matter air pollution and aortic perivascular adipose tissue: oxidative stress, leptin, and vascular dysfunction.
      ]. Acrolein is proposed as the primary mediator responsible for upregulation of PVAT and release of PVAT leptin following exposure to PM2.5 [
      • Haberzettl P.
      • Jin L.
      • Riggs D.W.
      • Zhao J.
      • O’Toole T.E.
      • Conklin D.J.
      Fine particulate matter air pollution and aortic perivascular adipose tissue: oxidative stress, leptin, and vascular dysfunction.
      ].
      These processes can result in release of mediators when triggered by HAP, as shown in Table 2.
      Table 2Potential air pollution induced local inflammatory mediators.
      MediatorDescriptionTriggersEffect on CVD
      Visfatin/nicotinamide phosphoribosyl transferase (NAMPT)An intracellular and extracellular adipocytokine found in adipocytes, bone marrow, muscles, and the heart [
      • Dakroub A.
      • Nasser S.A.
      • Kobeissy F.
      • Yassine H.M.
      • Orekhov A.
      • Sharifi-Rad J.
      • et al.
      Visfatin: an emerging adipocytokine bridging the gap in the evolution of cardiovascular diseases.
      ]. Visfatin has a role in releasing inflammatory cytokines, MMPs, and activating inflammatory signalling pathways [
      • Dakroub A.
      • Nasser S.A.
      • Kobeissy F.
      • Yassine H.M.
      • Orekhov A.
      • Sharifi-Rad J.
      • et al.
      Visfatin: an emerging adipocytokine bridging the gap in the evolution of cardiovascular diseases.
      ].
      PM2.5 [
      • Wan Q.
      • Cui X.
      • Shao J.
      • Zhou F.
      • Jia Y.
      • Sun X.
      • et al.
      Beijing ambient particle exposure accelerates atherosclerosis in ApoE knockout mice by upregulating visfatin expression.
      ]

      PM10 [
      • Wan Q.
      • Cui X.
      • Shao J.
      • Zhou F.
      • Jia Y.
      • Sun X.
      • et al.
      Beijing ambient particle exposure accelerates atherosclerosis in ApoE knockout mice by upregulating visfatin expression.
      ]
      Atherosclerosis progression by promoting inflammation and adhesion, vascular remodelling by VEGF modulation, the release of FGF-2 and MMPs and activation of several angiogenic signalling pathways [
      • Dakroub A.
      • Nasser S.A.
      • Kobeissy F.
      • Yassine H.M.
      • Orekhov A.
      • Sharifi-Rad J.
      • et al.
      Visfatin: an emerging adipocytokine bridging the gap in the evolution of cardiovascular diseases.
      ]
      LeptinPM2.5 [
      • Haberzettl P.
      • Jin L.
      • Riggs D.W.
      • Zhao J.
      • O’Toole T.E.
      • Conklin D.J.
      Fine particulate matter air pollution and aortic perivascular adipose tissue: oxidative stress, leptin, and vascular dysfunction.
      ]
      Atherosclerosis by alteration in VSMC [
      • Haberzettl P.
      • Jin L.
      • Riggs D.W.
      • Zhao J.
      • O’Toole T.E.
      • Conklin D.J.
      Fine particulate matter air pollution and aortic perivascular adipose tissue: oxidative stress, leptin, and vascular dysfunction.
      ]
      Plasminogen activator inhibitor-1 (PAI-1)Sourced from inflamed adipose tissue, vascular endothelium and the liver that can be stored in platelets, making them resistant to thrombolysis [
      • Vaughan D.E.
      PAI-1 and atherothrombosis.
      ]. PAI-1's secretion is stimulated by a range of cytokines, growth factors and angiotensin II and IV [
      • Vaughan D.E.
      PAI-1 and atherothrombosis.
      ].
      PM2.5 [
      • Chen R.
      • Zhao Z.
      • Sun Q.
      • Lin Z.
      • Zhao A.
      • Wang C.
      • et al.
      Size-fractionated particulate air pollution and circulating biomarkers of inflammation, coagulation, and vasoconstriction in a panel of young adults.
      ]

      PM10 [
      • Chen R.
      • Zhao Z.
      • Sun Q.
      • Lin Z.
      • Zhao A.
      • Wang C.
      • et al.
      Size-fractionated particulate air pollution and circulating biomarkers of inflammation, coagulation, and vasoconstriction in a panel of young adults.
      ]

      Metal particles [
      • Wu S.
      • Deng F.
      • Wei H.
      • Huang J.
      • Wang H.
      • Shima M.
      • et al.
      Chemical constituents of ambient particulate air pollution and biomarkers of inflammation, coagulation and homocysteine in healthy adults: a prospective panel study.
      ]
      Atherosclerosis and CAD [
      • Vaughan D.E.
      PAI-1 and atherothrombosis.
      ]
      ResistinAn adipokine that has been shown to counteract insulin action leading to insulin resistance.PM2.5 [
      • Sun Q.
      • Yue P.
      • Deiuliis J.A.
      • Lumeng C.N.
      • Kampfrath T.
      • Mikolaj M.B.
      • et al.
      Ambient air pollution exaggerates adipose inflammation and insulin resistance in a mouse model of diet-induced obesity.
      ]
      Atherosclerosis, thrombus formation, angiogenesis, VSM cell function and altered endothelial function [
      • Jamaluddin M.S.
      • Weakley S.M.
      • Yao Q.
      • Chen C.
      Resistin: functional roles and therapeutic considerations for cardiovascular disease.
      ]
      AdiponectinThe most abundant secreted factor by adipocytes, with levels declining following stimulation with insulin, and cytokines such as TNF-α and endothelin-1 increase with IGF-1 stimulation [
      • Szmitko P.E.
      • Teoh H.
      • Stewart D.J.
      • Verma S.
      Adiponectin and cardiovascular disease: state of the art?.
      ].

      Adiponectin's level is declined with increased adipose tissue mass, and its function is opposite of that of leptin and resistin, making it an anti-inflammatory and anti-thrombotic agent [
      • Szmitko P.E.
      • Teoh H.
      • Stewart D.J.
      • Verma S.
      Adiponectin and cardiovascular disease: state of the art?.
      ].
      PM2.5 [
      • Wan Q.
      • Ding T.
      • Xu Y.
      • Zheng C.
      • Tu M.
      • Zhao T.
      Urban fine particulate air pollution exposure promotes atherosclerosis in apolipoprotein E-deficient mice by activating perivascular adipose tissue inflammation via the Wnt5a/Ror2 signaling pathway.
      ] ↓
      Cardioprotective agent by controlling atherosclerotic pathways such as endothelial dysfunction, plaque initiation and progression [
      • Szmitko P.E.
      • Teoh H.
      • Stewart D.J.
      • Verma S.
      Adiponectin and cardiovascular disease: state of the art?.
      ]
      Abbreviations: MMPs, matrix metalloproteinases; PM2.5, particulate matter <2.5 μm; PM10, particulate matter <10 μm; VEGF, vascular endothelial growth factor; FGF-2, fibroblast growth factor 2; VSMC, vascular smooth muscle cells; CAD, coronary artery disease; TNF-α, tumour necrosis factor alpha; IGF-1, insulin-like growth factor 1; VSM, vascular smooth muscle.

      Oxidative Stress

      Oxidative stress refers to the imbalance between the production of reactive oxygen species (ROS) and antioxidants, which play a central role in both local and systemic inflammation. The main active ROS are free radicals (O-) in various forms such as O2-, OH- and H2O2 (superoxide, hydroxyl, and hydrogen peroxide) [
      • D’Oria R.
      • Schipani R.
      • Leonardini A.
      • Natalicchio A.
      • Perrini S.
      • Cignarelli A.
      • et al.
      The role of oxidative stress in cardiac disease: from physiological response to injury factor.
      ]. ROS serve an essential role in vasodilation in controlled quantities. The different types of ROS differ from each other in terms of their production source and structure. These highly reactive components are controlled using various mechanisms collectively referred to as antioxidants. Failure of this control mechanism or excessive ROS production (endogenous or exogenous) results in an adverse reaction locally and systemically.
      In oxidative stress, ROS themselves can be considered mediators that are released in response to exposure to toxins such as PM2.5. Studies have shown that air pollution is a significant source of oxidative stress. PM2.5 comprising metals (iron [Fe], copper [Cu]) and organic species (photochemically aged organics), have the highest oxidative potential (OP) even at moderate concentrations [
      • Gao D.
      • Ripley S.
      • Weichenthal S.
      • Godri Pollitt K.J.
      Ambient particulate matter oxidative potential: chemical determinants, associated health effects, and strategies for risk management.
      ]. ROS can also interact with their surrounding lipids in the membrane, resulting in lipid pre-oxidisation. This effect produces a range of compounds, such as hydroxyl fatty acids and oxidised cholesterol, eventually causing destruction and increased membrane permeability, which further promotes local and systemic inflammation [
      • Feng J.
      • Cavallero S.
      • Hsiai T.
      • Li R.
      Impact of air pollution on intestinal redox lipidome and microbiome.
      ]. Various mediators are implicated in HAP-induced oxidative stress, as shown in Table 3.
      Table 3Potential air pollution induced oxidative stress mediators.
      MediatorDescriptionTriggersEffect on CVD
      Xanthine oxidoreductase (XOR)Expressed as either xanthine dehydrogenase (XDH) or xanthine oxide (XO) which are responsible for the oxidation of xanthine to uric acid resulting in the influx of free radicals [
      • D’Oria R.
      • Schipani R.
      • Leonardini A.
      • Natalicchio A.
      • Perrini S.
      • Cignarelli A.
      • et al.
      The role of oxidative stress in cardiac disease: from physiological response to injury factor.
      ].
      PM2.5 [
      • Xu M.-X.
      • Ge C.-X.
      • Qin Y.-T.
      • Gu T.-T.
      • Lou D.-S.
      • Li Q.
      • et al.
      Prolonged PM2.5 exposure elevates risk of oxidative stress-driven nonalcoholic fatty liver disease by triggering increase of dyslipidemia.
      ]
      Impaired myocardial energy production, especially in CCF [
      • D’Oria R.
      • Schipani R.
      • Leonardini A.
      • Natalicchio A.
      • Perrini S.
      • Cignarelli A.
      • et al.
      The role of oxidative stress in cardiac disease: from physiological response to injury factor.
      ]
      Nitric oxide synthases (NOS)Which consists of three known subtypes neuronal (nNOS), inducible (iNOS) and endothelial (eNOS), stimulates the production of NO both in the physiological state to maintain cardiovascular homeostasis but also in response to pathophysiological stimuli.PM2.5 [
      • Ying Z.
      • Xu X.
      • Chen M.
      • Liu D.
      • Zhong M.
      • Chen L.-c.
      • et al.
      A synergistic vascular effect of airborne particulate matter and nickel in a mouse model.
      ] ↓ (Mainly eNOS)
      Protective effect on the cardiovascular system and anti-thrombotic effect [
      • Shimokawa H.
      • Tsutsui M.
      Nitric oxide synthases in the pathogenesis of cardiovascular disease.
      ]
      NADPH oxidase (NOX)Constitutes seven members, utilise NADPH to donate electrons which results in the production of superoxide (O2) or hydrogen peroxide (H2O2) [
      • Sirker A.
      • Zhang M.
      • Shah A.M.
      NADPH oxidases in cardiovascular disease: insights from in vivo models and clinical studies.
      ].
      PM2.5 [
      • Cáceres L.
      • Paz M.L.
      • Garcés M.
      • Calabró V.
      • Magnani N.D.
      • Martinefski M.
      • et al.
      NADPH oxidase and mitochondria are relevant sources of superoxide anion in the oxinflammatory response of macrophages exposed to airborne particulate matter.
      ]
      HTN, atherosclerosis, IHD and cardiac fibrosis [
      • Sirker A.
      • Zhang M.
      • Shah A.M.
      NADPH oxidases in cardiovascular disease: insights from in vivo models and clinical studies.
      ]
      Cytochrome P450 (CYP)Primarily responsible for the peroxidation and oxidation of elements such as vitamins, steroids, and certain drugs. Outside its physiological role, CYP contributes to amino acid (AA) metabolism and ROS production.PM2.5 [
      • Simkhovich B.Z.
      • Kleinman M.T.
      • Mehrian-Shai R.
      • Hsu Y.-H.
      • Meacher D.
      • Gookin G.
      • et al.
      Chronic exposure to ambient particulate matter alters cardiac gene expression patterns and markers of oxidative stress in rats.
      ]
      HTN, angiogenesis, atherosclerosis, CCF, arrhythmia and cardiomyopathy [
      • Hunter A.L.
      • Cruz R.P.
      • Cheyne B.M.
      • McManus B.M.
      • Granville D.J.
      Cytochrome p450 enzymes and cardiovascular disease.
      ]
      Abbreviations: PM2.5, particulate matter <2.5 μm; CCF, congestive cardiac failure; NADPH, nicotinamide adenine dinucleotide phosphate; HTN, hypertension; IHD, ischaemic heart disease; AA, amino acid; ROS, reactive oxygen species.

      Mitochondrial Dysfunction

      The role of mitochondrial disorders in acquired cardiovascular pathophysiology has become a subject of great activity owing to correlations seen in hereditary mitochondrial diseases and cardiovascular comorbidities. There are several ways that mitochondrial physiology can be perturbed: altered morphology, disrupted mitochondrial energetics, increased oxidative stress, dysregulation of apoptosis and autophagy, and via increased mitochondrial mutations [
      • Dominic E.A.
      • Ramezani A.
      • Anker S.D.
      • Verma M.
      • Mehta N.
      • Rao M.
      Mitochondrial cytopathies and cardiovascular disease.
      ].
      Mitochondrial oxidative stress and DNA damage are shown to result in endothelial dysfunction [
      • Mikhed Y.
      • Daiber A.
      • Steven S.
      Mitochondrial Oxidative Stress, Mitochondrial DNA damage and their role in age-related vascular dysfunction.
      ] and various cardiac complications such as hypertension, atherosclerosis, and cardiomyopathy [
      • West A.P.
      • Shadel G.S.
      Mitochondrial DNA in innate immune responses and inflammatory pathology.
      ]. Mitochondrial DNA (mtDNA) damage has a role in innate immune response and inflammation [
      • West A.P.
      • Shadel G.S.
      Mitochondrial DNA in innate immune responses and inflammatory pathology.
      ] which can explain these cardiovascular complications [
      • Nakayama H.
      • Otsu K.
      Mitochondrial DNA as an inflammatory mediator in cardiovascular diseases.
      ]. More specifically, some of the pathophysiological processes attributed to cardiovascular injury include increased arterial pressure and vascular dysfunction [
      • West A.P.
      • Shadel G.S.
      Mitochondrial DNA in innate immune responses and inflammatory pathology.
      ], Incomplete degradation of mtDNA by autophagy [
      • Ding Z.
      • Liu S.
      • Wang X.
      • Khaidakov M.
      • Dai Y.
      • Mehta J.L.
      Oxidant stress in mitochondrial DNA damage, autophagy and inflammation in atherosclerosis.
      ] and accumulation of mtDNA–LL37 complexes in atherosclerotic plaques [
      • Zhang Z.
      • Meng P.
      • Han Y.
      • Shen C.
      • Li B.
      • Hakim M.A.
      • et al.
      Mitochondrial DNA-LL-37 Complex promotes atherosclerosis by escaping from autophagic recognition.
      ].
      Exposure to air pollution, especially PM2.5 and PM10, result in mitochondrial damage resulting in several adverse cardiovascular outcomes such as reduced HRV [
      • Byun H.M.
      • Colicino E.
      • Trevisi L.
      • Fan T.
      • Christiani D.C.
      • Baccarelli A.A.
      Effects of air pollution and blood mitochondrial DNA methylation on markers of heart rate variability.
      ]. This is owing to diverse mitochondrial functions in energy production, regulation of metabolism and homeostasis of elements such as iron and copper and even cellular death mechanisms [
      • Boovarahan S.R.
      • Kurian G.A.
      Mitochondrial dysfunction: a key player in the pathogenesis of cardiovascular diseases linked to air pollution.
      ]. Pathways leading to mitochondrial damage are currently poorly understood, and, at present, ROS are the central identified mediators following exposure to toxins such as air pollution, as shown in Table 4.
      Table 4Potential air pollution induced mediators of mitochondrial dysfunction.
      MediatorDescriptionTriggersEffect on CVD
      Mitochondrial reactive oxygen species (m)Raised ROS due to various internal or exogenous factors are shown to cause mtDNA damage [
      • Suematsu N.
      • Tsutsui H.
      • Wen J.
      • Kang D.
      • Ikeuchi M.
      • Ide T.
      • et al.
      Oxidative stress mediates tumor necrosis factor-α–induced mitochondrial DNA damage and dysfunction in cardiac myocytes.
      ,
      • Ballinger S.W.
      • Patterson C.
      • Knight-Lozano C.A.
      • Burow D.L.
      • Conklin C.A.
      • Hu Z.
      • et al.
      Mitochondrial integrity and function in atherogenesis.
      ].
      PM2.5 [
      • Byun H.M.
      • Colicino E.
      • Trevisi L.
      • Fan T.
      • Christiani D.C.
      • Baccarelli A.A.
      Effects of air pollution and blood mitochondrial DNA methylation on markers of heart rate variability.
      ]

      PM10 [
      • Byun H.M.
      • Colicino E.
      • Trevisi L.
      • Fan T.
      • Christiani D.C.
      • Baccarelli A.A.
      Effects of air pollution and blood mitochondrial DNA methylation on markers of heart rate variability.
      ]
      Myositis and dilated cardiomyopathy [
      • Oka T.
      • Hikoso S.
      • Yamaguchi O.
      • Taneike M.
      • Takeda T.
      • Tamai T.
      • et al.
      Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure.
      ]

      Atherosclerosis and VSM dysfunction [
      • Ballinger S.W.
      • Patterson C.
      • Knight-Lozano C.A.
      • Burow D.L.
      • Conklin C.A.
      • Hu Z.
      • et al.
      Mitochondrial integrity and function in atherogenesis.
      ,
      • Ballinger S.W.
      • Patterson C.
      • Yan C.-N.
      • Doan R.
      • Burow D.L.
      • Young C.G.
      • et al.
      Hydrogen peroxide–and peroxynitrite-induced mitochondrial DNA damage and dysfunction in vascular endothelial and smooth muscle cells.
      ]
      Abbreviations: ROS, reactive oxygen species; mtDNA, mitochondrial DNA; PM2.5, particulate matter <2.5 μm; PM10, particulate matter <10 μm; VSM, vascular smooth muscle.

      Results, Part B: Non-Inflammatory Mediators

      Endothelial Dysfunction

      Endothelial dysfunction has a critical role in developing atherosclerotic plaques as the initial step in this pathological cascade which is often followed by increased vascular permeability to lipoproteins, enhanced leucocyte adhesion, platelet aggregation, and the production of various inflammatory cytokines. Endothelial dysfunction is a result of various pathophysiological pathways, most notably: oxidative stress, inflammation, leucocyte adhesion and transmigration, apoptosis and cell death, endothelial NO synthase uncoupling, endothelial-to-mesenchymal transition, and endothelial cell senescence [
      • Xu S.
      • Ilyas I.
      • Little P.J.
      • Li H.
      • Kamato D.
      • Zheng X.
      • et al.
      Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies.
      ].
      Acute exposure to PM2.5 is shown to elevate the circulating von Willebrand factor (vWF) level, which directly measures endothelial injury [
      • Liang Q.
      • Sun M.
      • Wang F.
      • Ma Y.
      • Lin L.
      • Li T.
      • et al.
      Short-term PM2.5 exposure and circulating von Willebrand factor level: a meta-analysis.
      ]. Liang et al. [
      • Liang Q.
      • Sun M.
      • Wang F.
      • Ma Y.
      • Lin L.
      • Li T.
      • et al.
      Short-term PM2.5 exposure and circulating von Willebrand factor level: a meta-analysis.
      ] demonstrated that the effect of PM2.5 on endothelial dysfunction is likely via inflammation and oxidative stress, whereas, in another study [
      • Bo L.
      • Jiang S.
      • Xie Y.
      • Kan H.
      • Song W.
      • Zhao J.
      Effect of vitamin E and omega-3 fatty acids on protecting ambient PM2.5-induced inflammatory response and oxidative stress in vascular endothelial cells.
      ], direct cytotoxicity and autophagy were proposed as the likely mechanism. Air pollution mediated factors that can lead to endothelial dysfunction are show in Table 5.
      Table 5Potential air pollution induced mediators of endothelial dysfunction.
      MediatorDescriptionTriggersEffect on CVD
      Endothelial microparticles (EMP)Result from endothelial shedding and are thought to have both beneficial and detrimental effects on the endothelium [
      • Dignat-George F.
      • Boulanger C.M.
      The many faces of endothelial microparticles.
      ].
      PM2.5 [
      • Pope C.A.
      • Bhatnagar A.
      • McCracken J.P.
      • Abplanalp W.
      • Conklin D.J.
      • O’Toole T.
      Exposure to fine particulate air pollution is associated with endothelial injury and systemic inflammation.
      ]
      Endothelial injury, inflammation, thrombosis, and angiogenesis [
      • Dignat-George F.
      • Boulanger C.M.
      The many faces of endothelial microparticles.
      ]
      Intercellular adhesion molecule 1 (ICAM-1)Expressed on a range of cells, including cardiomyocyte, which is upregulated in response to cytokine, ROS, and apoptosis, amongst other stimuli [
      • Niessen H.W.M.
      • Krijnen P.A.J.
      • Visser C.A.
      • Meijer C.J.L.M.
      • Hack C.E.
      Intercellular adhesion molecule-1 in the heart.
      ]. As the name implies, it mediates the adhesion of circulating inflammatory molecules to the vascular wall and trans-endothelial migration to vascular intima.
      PM2.5 [
      • Xie W.
      • You J.
      • Zhi C.
      • Li L.
      The toxicity of ambient fine particulate matter (PM2.5) to vascular endothelial cells.
      ] PM10 [
      • Yatera K.
      • Hsieh J.
      • Hogg J.C.
      • Tranfield E.
      • Suzuki H.
      • Shih C.-H.
      • et al.
      Particulate matter air pollution exposure promotes recruitment of monocytes into atherosclerotic plaques.
      ]

      NO2 [
      • Li H.
      • Han M.
      • Guo L.
      • Li G.
      • Sang N.
      Oxidative stress, endothelial dysfunction and inflammatory response in rat heart to NO2 inhalation exposure.
      ]
      Atherosclerosis and the progression of CAD [
      • Niessen H.W.M.
      • Krijnen P.A.J.
      • Visser C.A.
      • Meijer C.J.L.M.
      • Hack C.E.
      Intercellular adhesion molecule-1 in the heart.
      ]
      Vascular adhesion molecule 1 (VCAM-1)Belongs to the superfamily of immunoglobulins, expressed in the endothelium of blood vessels in most vital organs, including the brain and heart, induced by pro-inflammatory cytokines, and following expression leading to accumulation and transmigration of monocytes and other innate inflammatory cells [
      • Troncoso M.F.
      • Ortiz-Quintero J.
      • Garrido-Moreno V.
      • Sanhueza-Olivares F.
      • Guerrero-Moncayo A.
      • Chiong M.
      • et al.
      VCAM-1 as a predictor biomarker in cardiovascular disease.
      ].
      PM2.5 [
      • Xie W.
      • You J.
      • Zhi C.
      • Li L.
      The toxicity of ambient fine particulate matter (PM2.5) to vascular endothelial cells.
      ] PM10 [
      • Yatera K.
      • Hsieh J.
      • Hogg J.C.
      • Tranfield E.
      • Suzuki H.
      • Shih C.-H.
      • et al.
      Particulate matter air pollution exposure promotes recruitment of monocytes into atherosclerotic plaques.
      ]
      Atherosclerosis, HTN, AF, IHD and HFpEF [
      • Troncoso M.F.
      • Ortiz-Quintero J.
      • Garrido-Moreno V.
      • Sanhueza-Olivares F.
      • Guerrero-Moncayo A.
      • Chiong M.
      • et al.
      VCAM-1 as a predictor biomarker in cardiovascular disease.
      ]
      Lectin-like oxidised low-density lipoprotein 1 receptor (LOX-1 receptor)A prominent scavenger receptor that is expressed on ECs, inflammatory cells and cardiomyocytes. LOX-1 receptors located on macrophages can bind to modified LDL (e.g., oxidised carbamylated or acetylated) and non-self or modified self-targets that result in uptake of the modified lipids. Similarly, in endothelial cells, they behave as sensors that can respond to external stimuli and alter their cellular phenotype from anti-inflammatory to pro-inflammatory.

      In the normal physiological state, these actions eliminate harmful or degraded substances. However, these responses can result in endothelial dysfunction in a pathophysiological state.
      PM2.5 [
      • Liu J.
      • Sun Q.
      • Sun M.
      • Lin L.
      • Ren X.
      • Li T.
      • et al.
      Melatonin alleviates PM2.5-triggered macrophage M1 polarization and atherosclerosis via regulating NOX2-mediated oxidative stress homeostasis.
      ]
      Atherosclerosis [
      • Akhmedov A.
      • Sawamura T.
      • Chen C.-H.
      • Kraler S.
      • Vdovenko D.
      • Lüscher T.F.
      Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1): a crucial driver of atherosclerotic cardiovascular disease.
      ]
      Angiotensin-converting enzyme/angiotensin II/angiotensin type 1 receptor axis (ACE/ANGII/AT1R)Renin-angiotensin system (RAS) with the end-product of angiotensin II has long been identified as an important agent in the pathophysiology of cardiovascular diseases and cardiac remodelling. The development of angiotensin ACE inhibitors and angiotensin receptor blockers (ARBs) have revolutionised the management of hypertension, CCF and IHD [
      • Ferrario C.M.
      Role of Angiotensin II in Cardiovascular Disease — Therapeutic implications of more than a century of research.
      ].
      PM2.5 [
      • Xu X.
      • Qimuge A.
      • Wang H.
      • Xing C.
      • Gu Y.
      • Liu S.
      • et al.
      IRE1α/XBP1s branch of UPR links HIF1α activation to mediate ANGII-dependent endothelial dysfunction under particulate matter (PM) 2.5 exposure.
      ]
      Cardiac remodelling, HTN, CCF and IHD
      Small proline-rich repeat protein 3 (SPRR3)Usually found in the foregut and oesophagus; however, it is also expressed in VSMCs of the atheroma of large arteries in a pathological state in response to mechanic cyclic stress [
      • Saraswati S.
      • Lietman C.D.
      • Li B.
      • Mathew S.
      • Zent R.
      • Young P.P.
      Small proline-rich repeat 3 is a novel coordinator of PDGFRβ and integrin β1 crosstalk to augment proliferation and matrix synthesis by cardiac fibroblasts.
      ].
      PM2.5 [
      • Bae J.-E.
      • Choi H.
      • Shin D.W.
      • Na H.-W.
      • Park N.Y.
      • Kim J.B.
      • et al.
      Fine particulate matter (PM2.5) inhibits ciliogenesis by increasing SPRR3 expression via c-Jun activation in RPE cells and skin keratinocytes.
      ]
      Cardiac fibrosis by augmentation of fibroblast proliferation [
      • Saraswati S.
      • Lietman C.D.
      • Li B.
      • Mathew S.
      • Zent R.
      • Young P.P.
      Small proline-rich repeat 3 is a novel coordinator of PDGFRβ and integrin β1 crosstalk to augment proliferation and matrix synthesis by cardiac fibroblasts.
      ]
      Abbreviations: PM2.5, particulate matter <2.5 µm; PM10, particulate matter <10 µm; VSMCs, vascular smooth muscle cells; CCF, congestive cardiac failure; HTN, hypertension; IHD, ischaemic heart disease; AF, atrial fibrillation; HFpEF, heart failure with preserved ejection fraction; ECs, endothelial cells.

      Atherosclerotic Plaque Vulnerability

      Atherosclerotic plaque rupture is considered the leading cause of myocardial infarction and ischaemic stroke. Plaque vulnerability is shown to be increased after exposure to HAPs, likely by upregulation of MMPs and TIMPs. Key mediators are metalloproteases and proteins that modulate these moieties, as shown in Table 6.
      Table 6Potential air pollution induced mediators of increased atherosclerotic plaque vulnerability.
      MediatorDescriptionTriggersEffect on CVD
      Matrix-degrading metalloproteinases (MMPS)Present in macrophages, endothelial cells, and smooth muscle cells at low levels [
      • Chistiakov D.A.
      • Sobenin I.A.
      • Orekhov A.N.
      Vascular extracellular matrix in atherosclerosis.
      ], playing an essential role in regulating inflammatory response and thrombogenesis following plaque rupture [
      • Mosevoll K.A.
      • Johansen S.
      • Wendelbo Ø.
      • Nepstad I.
      • Bruserud Ø.
      • Reikvam H.
      Cytokines, adhesion molecules, and matrix metalloproteases as predisposing, diagnostic, and prognostic factors in venous thrombosis.
      ].
      PM2.5, NO2, CO and SO2 [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]
      Alteration in vascular anatomy and atherosclerotic plaque vulnerability [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]
      Tissue inhibitor of metalloproteinases (TIMPS)The proteins responsible for modulating MMPs.PM2.5, NO2, CO and SO2 [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]
      Serum level is associated with Framingham Risk Score and inversely with LV systolic function [
      • Messerli F.H.
      TIMPs, MMPs and cardiovascular disease.
      ]
      Abbreviations: PM2.5, particulate matter <2.5 µm; PM10, particulate matter <10 µm; CVD, cardiovascular disease; MMPs, matrix metalloproteinases; LV, left ventricle.

      Thrombogenesis

      High HAP levels are associated with increased pro-thrombogenicity markers, as shown in Table 7 [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]. Obese patients are shown to be primarily impacted by the pro-thrombogenic effect of PM2.5, which is thought to be modulated by adipose inflammation leading to platelet activation and aggregation [
      • Hu D.
      • Jia X.
      • Cui L.
      • Liu J.
      • Chen J.
      • Wang Y.
      • et al.
      Exposure to fine particulate matter promotes platelet activation and thrombosis via obesity-related inflammation.
      ].
      Table 7Potential air pollution induced thrombogenic mediators.
      MediatorDescriptionTriggersEffect on CVD
      Soluble CD40 ligand (SCD40L)A pro-inflammatory and pro-thrombotic protein that belongs to the superfamily of TNF superfamily with receptors on B cells. sCD40L is predominantly sourced from platelets and can enhance platelet activation and aggregation [
      • Yacoub D.
      • Hachem A.
      • Théorêt J.-F.
      • Gillis M.-A.
      • Mourad W.
      • Merhi Y.
      Enhanced levels of soluble CD40 ligand exacerbate platelet aggregation and thrombus formation through a CD40-dependent tumor necrosis factor receptor-associated factor-2/Rac1/p38 mitogen-activated protein kinase signaling pathway.
      ].
      HAP [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]
      Thrombus formation [
      • Yacoub D.
      • Hachem A.
      • Théorêt J.-F.
      • Gillis M.-A.
      • Mourad W.
      • Merhi Y.
      Enhanced levels of soluble CD40 ligand exacerbate platelet aggregation and thrombus formation through a CD40-dependent tumor necrosis factor receptor-associated factor-2/Rac1/p38 mitogen-activated protein kinase signaling pathway.
      ]
      Soluble P-selectin (SCD62P)A cell adhesion protein that belongs to the lectin family, which is stored in platelets and endothelial cells. Once activated, it gets translocated to the cell surface and released as a soluble form to circulation. Once the soluble form is bounded to its receptor PSGL-1, it starts a pro-coagulant cascade [
      • Frenette P.S.
      • Denis C.V.
      • Weiss L.
      • Jurk K.
      • Subbarao S.
      • Kehrel B.
      • et al.
      P-Selectin glycoprotein ligand 1 (PSGL-1) is expressed on platelets and can mediate platelet-endothelial interactions in vivo.
      ].
      HAP [
      • Xu H.
      • Wang T.
      • Liu S.
      • Brook R.D.
      • Feng B.
      • Zhao Q.
      • et al.
      Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
      ]
      Thrombus formation [
      • Frenette P.S.
      • Denis C.V.
      • Weiss L.
      • Jurk K.
      • Subbarao S.
      • Kehrel B.
      • et al.
      P-Selectin glycoprotein ligand 1 (PSGL-1) is expressed on platelets and can mediate platelet-endothelial interactions in vivo.
      ]
      Abbreviations: HAP, hazardous air pollutants; TNF, tumour necrosis factors; CVD, cardiovascular disease; PSGL-1, P-selectin glycoprotein ligand 1.

      Autonomic Dysfunction

      Autonomic dysregulation, which results from the accentuated sympathetic drive and diminished parasympathetic activity, is considered a major risk factor for adverse cardiovascular outcomes such as cardiac arrest and AMI [
      • Peçanha T.
      • Silva-Júnior N.D.
      • Forjaz CLdM.
      Heart rate recovery: autonomic determinants, methods of assessment and association with mortality and cardiovascular diseases.
      ]. Alterations in cardiac autonomic tone are thought to be the most immediate effects of exposure to air pollutants [
      • Perez C.M.
      • Hazari M.S.
      • Farraj A.K.
      Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
      ]. This is thought to be due to a shift from sympatho-inhibition to sympatho-excitation as evidenced by studies showing reduced high-frequency heart rate variability (HRV) [
      • Huang W.
      • Zhu T.
      • Pan X.
      • Hu M.
      • Lu S.-E.
      • Lin Y.
      • et al.
      Air pollution and autonomic and vascular dysfunction in patients with cardiovascular disease: interactions of systemic inflammation, overweight, and gender.
      ] and the observed protective effect of β-adrenoceptor inhibitors on pollution-evoked CV events [
      • Taylor-Clark T.E.
      Air pollution-induced autonomic modulation.
      ]. There is no study directly assessing autonomic nerve activity in response to air pollution, and most of our understanding of the pathophysiology is theoretical.

      Inflammatory mediators

      Inflammatory mediators can have a direct effect on modulation of autonomic system. These include:
      • Toll-like receptor 2 (TLR2) Methylation: secondary to PM2.5 is shown to result in activation of autonomic system [
        • Zhong J.
        • Colicino E.
        • Lin X.
        • Mehta A.
        • Kloog I.
        • Zanobetti A.
        • et al.
        Cardiac autonomic dysfunction: particulate air pollution effects are modulated by epigenetic immunoregulation of Toll-like receptor 2 and dietary flavonoid intake.
        ].

      Airway receptors

      Airway receptors include sensors located throughout the upper and lower airways (C-nerve fibres, Rapidly adapting pulmonary receptors [RARs] and Slowly adapting pulmonary receptors [SARs]), designed to sense environmental irritants, such as cigarette smoke and air pollutants, leading to a range of reflexes such as coughing and dyspnoea [
      • Perez C.M.
      • Hazari M.S.
      • Farraj A.K.
      Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
      ].
      Air pollution-induced mediators of C-nerve fibres activation are as follows:
      • Transient receptor potential ankyrin 1 (TRPA1) channel is a small Ca2+-permeant non-selective cation channel activated by cold exposure, environmental irritants, and reactive oxides [
        • Grace M.S.
        • Belvisi M.G.
        TRPA1 receptors in cough.
        ]. Its activation results in inflammatory hyperalgesia and neurogenic inflammation, and, being located on nociceptive neurons, causes the perception of noxious stimuli [
        • Grace M.S.
        • Belvisi M.G.
        TRPA1 receptors in cough.
        ]. Following exposure to diesel exhaust, these receptors are shown to increase cardiac arrhythmogenesis and autonomic activation [
        • Hazari M.S.
        • Haykal-Coates N.
        • Winsett D.W.
        • Krantz Q.T.
        • King C.
        • Costa D.L.
        • et al.
        TRPA1 and sympathetic activation contribute to increased risk of triggered cardiac arrhythmias in hypertensive rats exposed to diesel exhaust.
        ].
      • Transient receptor potential vanilloid 1 (TRPV1) channel is present on the nasal mucosa, trachea, bronchi, and alveoli, which senses toxins such as capsaicin, extracellular protons (released during tissue acidosis and ischaemia) as well as excess heat [
        • Bessac B.F.
        • Jordt S.-E.
        Breathtaking TRP channels: TRPA1 and TRPV1 in airway chemosensation and reflex control.
        ].
      • Purinergic P2X channel receptors are present throughout the body with physiological roles such as neurotransmission, inflammation, and cell death [
        • Barth K.
        • Kasper M.
        Membrane compartments and purinergic signalling: occurrence and function of P2X receptors in lung.
        ]. In the lungs, subtypes of P2X receptors can be found on most microvascular endothelial cells.
      Activation of C-nerve fibres result in production of substance P, which is also a potent biological mediator, as shown in Table 8.
      Table 8Activated airway receptors following exposure to HAP with the potential to cause cardiovascular damage.
      MediatorDescriptionTriggersEffect on CVD
      Substance P (SP)Widely expressed in both central and peripheral nervous systems with functions such as regulating pulmonary and cardiovascular function, emetic reflux, noxious stimuli, and modulating autonomic reflexes [
      • Kraneveld A.D.
      • Nijkamp F.P.
      Tachykinins and neuro-immune interactions in asthma.
      ]. SP is also released by inflammatory cells [
      • O'Connor T.M.
      • O'Connell J.
      • O'Brien D.I.
      • Goode T.
      • Bredin C.P.
      • Shanahan F.
      The role of substance P in inflammatory disease.
      ].

      Local SP is shown to result in coronary artery vasodilation by releasing NO [
      • Quyyumi A.A.
      • Mulcahy D.
      • Andrews N.P.
      • Husain S.
      • Panza J.A.
      • Cannon R.O.
      Coronary vascular nitric oxide activity in hypertension and hypercholesterolemia.
      ] and have a protective effect against endothelial dysfunction [
      • Kim D.Y.
      • Piao J.
      • Hong H.S.
      Substance-P inhibits cardiac microvascular endothelial dysfunction caused by high glucose-induced oxidative stress.
      ]. In contrast, over the long term, elevated SP is shown to have a detrimental effect on the heart as it plays a vital role in cardiac mast cell activation [
      • Levick S.P.
      • Brower G.L.
      • Janicki J.S.
      Substance P-mediated cardiac mast cell activation: an in vitro study.
      ]. Cardiac mast cell activation does so via three pathways: the release of pro-inflammatory mediators such as TNFα and MMPs; the release of renin; and the production of vascular endothelial growth factor (VEGF) [
      • Feickert M.
      • Burckhardt B.B.
      Substance P in cardiovascular diseases—a bioanalytical review.
      ].
      O3 [
      • Barker J.S.
      • Wu Z.
      • Hunter D.D.
      • Dey R.D.
      Ozone exposure initiates a sequential signaling cascade in airways involving interleukin-1beta release, nerve growth factor secretion, and substance P upregulation.
      ,
      • Hazbun M.E.
      • Hamilton R.
      • Holian A.
      • Eschenbacher W.L.
      Ozone-induced Increases in substance P and 8-epi-prostaglandin F2α in the airways of human subjects.
      ]
      Cardiac fibrosis and hypertrophy by upregulating Endothelin-1 [
      • Dehlin H.M.
      • Manteufel E.J.
      • Monroe A.L.
      • Reimer M.H.
      • Levick S.P.
      Substance P acting via the neurokinin-1 receptor regulates adverse myocardial remodeling in a rat model of hypertension.
      ]
      Abbreviations: TNFα, tumour necrosis factor alpha; MMP, matrix metalloproteinases.

      Baroreceptors

      Baroreceptors are stretch-sensitive sensors located in the carotid sinus and aortic arch to adjust blood pressure in response to alterations in systemic BP by inhibiting the sympathetic or activating the parasympathetic system [
      • Perez C.M.
      • Hazari M.S.
      • Farraj A.K.
      Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
      ]. Baroreflex desensitisation can result from chronic hypertension and CVD [
      • La Rovere M.T.
      • Pinna G.D.
      • Raczak G.
      Baroreflex sensitivity: measurement and clinical implications.
      ]. Exposure to major air pollutants such as SO2 reduces baroreflex sensitivity (BRS). At the same time, many studies have also shown BP instability following exposure to air pollutants (lowered BP in the acute setting and hypertension in the long term), which can indirectly result in reduced BRS [
      • Perez C.M.
      • Hazari M.S.
      • Farraj A.K.
      Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
      ].

      Chemoreceptors

      Chemoreceptors are located centrally in the brainstem and peripherally in the aorta and carotid bodies, maintaining homeostasis [
      • Perez C.M.
      • Hazari M.S.
      • Farraj A.K.
      Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
      ]. Carotid body chemoreceptors act as sensors for variations in O2, CO2, pH and temperature and activate sympathetic tone in response to hypoxia [
      • Perez C.M.
      • Hazari M.S.
      • Farraj A.K.
      Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
      ]. Thus far, no study has directly examined the effect of air pollution on carotid body chemoreceptors. However, given that there is evidence of hypoxia following exposure to air pollution, it is safe to conclude that these sensors play a role in air pollution-induced autonomic dysregulation [
      • Perez C.M.
      • Hazari M.S.
      • Farraj A.K.
      Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
      ].

      Impaired Lipid Metabolism

      An important site of lipid metabolism that is also impacted by exposure to air pollution is the intestine. The intestine is a crucial player in lipid metabolism by controlling dietary or biliary cholesterol absorption and synthesising endogenous cholesterol such as apolipoprotein A-1 (apoA-I) and high-density lipoprotein (HDL) via intestine enterocytes [
      • Feng J.
      • Cavallero S.
      • Hsiai T.
      • Li R.
      Impact of air pollution on intestinal redox lipidome and microbiome.
      ]. Recent evidence shows that air pollution can directly alter intestinal lipid metabolism by changes in lipid intestinal redox lipidome [
      • Feng J.
      • Cavallero S.
      • Hsiai T.
      • Li R.
      Impact of air pollution on intestinal redox lipidome and microbiome.
      ].
      Another vital source of dysregulation in lipid metabolism is the liver. Air pollutants can result in lipid peroxidation leading to oxidation of a range of lipids, including low-density lipoprotein (LDL) and Apo [
      • Feng J.
      • Cavallero S.
      • Hsiai T.
      • Li R.
      Impact of air pollution on intestinal redox lipidome and microbiome.
      ]. The oxidisation of lipids via intestinal and hepatic routes has a range of adverse health impacts, such as diabetes, atherosclerosis, and systemic inflammation [
      • Feng J.
      • Cavallero S.
      • Hsiai T.
      • Li R.
      Impact of air pollution on intestinal redox lipidome and microbiome.
      ]. Air pollution induced mediators causing impaired lipid metabolism are shown in Table 9.
      Table 9Air pollution induced mediators causing impaired lipid metabolism.
      MediatorDescriptionTriggersEffect on CVD
      Fatty acid translocase (FAT; CD36)A member of the scavenger receptor family that is expressed on endothelial cells (ECs), cardiomyocytes, and adipocytes amongst other tissues, which has a crucial role in immune regulation, platelet activation, and regulation of metabolism by regulating fatty acid (FA) uptake in ECs and myocardial tissue and FA metabolism in the liver [
      • Shu H.
      • Peng Y.
      • Hang W.
      • Nie J.
      • Zhou N.
      • Wang D.W.
      The role of CD36 in cardiovascular disease.
      ]. CD36 is stimulated by insulin, hyperglycaemia, and hyperlipidaemia [
      • Shu H.
      • Peng Y.
      • Hang W.
      • Nie J.
      • Zhou N.
      • Wang D.W.
      The role of CD36 in cardiovascular disease.
      ].
      PM2.5 [
      • Du X.
      • Jiang S.
      • Zeng X.
      • Zhang J.
      • Pan K.
      • Zhou J.
      • et al.
      Air pollution is associated with the development of atherosclerosis via the cooperation of CD36 and NLRP3 inflammasome in ApoE-/- mice.
      ]

      O3 [
      • Robertson S.
      • Colombo E.S.
      • Lucas S.N.
      • Hall P.R.
      • Febbraio M.
      • Paffett M.L.
      • et al.
      CD36 mediates endothelial dysfunction downstream of circulating factors induced by O3 exposure.
      ]
      IHD, cardiomyopathy, cardiac hypertrophy, and atherosclerosis by altering myocardial energy supply and the activation of platelets. [
      • Shu H.
      • Peng Y.
      • Hang W.
      • Nie J.
      • Zhou N.
      • Wang D.W.
      The role of CD36 in cardiovascular disease.
      ]
      OX-LDLExcessive ox-LDL stimulate the expression of the LOX-1 receptor, thereby reducing the protective autophagy response, leading to EC dysfunction [
      • Mollace V.
      • Gliozzi M.
      • Musolino V.
      • Carresi C.
      • Muscoli S.
      • Mollace R.
      • et al.
      Oxidized LDL attenuates protective autophagy and induces apoptotic cell death of endothelial cells: role of oxidative stress and LOX-1 receptor expression.
      ].
      PM2.5 [
      • Tian M.
      • Zhao J.
      • Mi X.
      • Wang K.
      • Kong D.
      • Mao H.
      • et al.
      Progress in research on effect of PM2.5 on occurrence and development of atherosclerosis.
      ]
      Atherosclerosis [
      • Mollace V.
      • Gliozzi M.
      • Musolino V.
      • Carresi C.
      • Muscoli S.
      • Mollace R.
      • et al.
      Oxidized LDL attenuates protective autophagy and induces apoptotic cell death of endothelial cells: role of oxidative stress and LOX-1 receptor expression.
      ]
      LR4/MYD88/NF-ΚB signalling pathwayPM2.5 [
      • Geng J.
      • Liu H.
      • Ge P.
      • Hu T.
      • Zhang Y.
      • Zhang X.
      • et al.
      PM2.5 promotes plaque vulnerability at different stages of atherosclerosis and the formation of foam cells via TLR4/MyD88/NFκB pathway.
      ]

      O3 [
      • Fakhrzadeh L.
      • Laskin J.D.
      • Laskin D.L.
      Ozone-induced production of nitric oxide and TNF-α and tissue injury are dependent on NF-κB p50.
      ]
      Promote the transformation of macrophages into foam cells [
      • Geng J.
      • Liu H.
      • Ge P.
      • Hu T.
      • Zhang Y.
      • Zhang X.
      • et al.
      PM2.5 promotes plaque vulnerability at different stages of atherosclerosis and the formation of foam cells via TLR4/MyD88/NFκB pathway.
      ]

      Excessive expression of inflammatory factors such as TNF-α, CRP and IL-6 [
      • Fakhrzadeh L.
      • Laskin J.D.
      • Laskin D.L.
      Ozone-induced production of nitric oxide and TNF-α and tissue injury are dependent on NF-κB p50.
      ]
      High-density lipoprotein (HDL)A critical cardioprotective mediator with a primary anti-atherosclerotic function by reverse cholesterol transport and efflux of cholesterol from macrophages into lipid-free ApoA-I, which results in healthy NO synthesis [
      • Li J.
      • Zhou C.
      • Xu H.
      • Brook R.D.
      • Liu S.
      • Yi T.
      • et al.
      Ambient air pollution is associated with HDL (high-density lipoprotein) dysfunction in healthy adults.
      ].
      PM2.5 [
      • Li J.
      • Zhou C.
      • Xu H.
      • Brook R.D.
      • Liu S.
      • Yi T.
      • et al.
      Ambient air pollution is associated with HDL (high-density lipoprotein) dysfunction in healthy adults.
      ] ↓
      HDL dysfunction is linked with CAD, atherosclerosis and increased cardiac mortality [
      • Li J.
      • Zhou C.
      • Xu H.
      • Brook R.D.
      • Liu S.
      • Yi T.
      • et al.
      Ambient air pollution is associated with HDL (high-density lipoprotein) dysfunction in healthy adults.
      ]
      Low-density lipids (LDL)In contrast to HDL, increased LDL levels are directly correlated with increased atherosclerosis burden and related CVD.PM2.5 [
      • Kim H.-J.
      • Kwon H.
      • Yun J.M.
      • Cho B.
      • Park J.-H.
      Interaction between visceral adiposity and ambient air pollution on LDL cholesterol level in Korean adults.
      ]
      Atherosclerosis [
      • Kim H.-J.
      • Kwon H.
      • Yun J.M.
      • Cho B.
      • Park J.-H.
      Interaction between visceral adiposity and ambient air pollution on LDL cholesterol level in Korean adults.
      ]
      Abbreviations: PM2.5, particulate matter <2.5 µm; IHD, ischaemic heart disease; TNF-α, tumour necrosis factor alpha; CRP, C-reactive protein; IL-6, Interleukin 6; HDL, high-density lipoprotein; LDL, low-density lipoprotein; CAD, coronary artery disease; CVD, cardiovascular disease.

      10 Other Mediators

      The emergence of new mediators that do not fall under any of the traditional pathogenic pathways is shining a new light in our understanding of the missing links between interaction of HAPs with airway tissues and adverse cardiovascular outcomes (Table 10).
      Table 10Novel mediators linking air pollution and cardiovascular injury.
      MediatorDescriptionTriggersEffect on CVD
      Aryl hydrocarbon receptor (AHR)Plays a central role in regulating the toxicity of environmental pollutants, and their upregulation results in inflammation, oxidative stress, and lipid infiltration [
      • Wang C.
      • Petriello M.C.
      • Zhu B.
      • Hennig B.
      PCB 126 induces monocyte/macrophage polarization and inflammation through AhR and NF-κB pathways.
      ].
      PM2.5 [
      • Tian M.
      • Zhao J.
      • Mi X.
      • Wang K.
      • Kong D.
      • Mao H.
      • et al.
      Progress in research on effect of PM2.5 on occurrence and development of atherosclerosis.
      ]
      Inflammation, oxidative stress, and lipid infiltration [
      • Wang C.
      • Petriello M.C.
      • Zhu B.
      • Hennig B.
      PCB 126 induces monocyte/macrophage polarization and inflammation through AhR and NF-κB pathways.
      ].
      Gut microbiomeBy far the largest reservoir of micro-organisms, containing 90% of the human body, which have myriad roles in the body's homeostasis, including their role in the immune system and cell-signalling processes [
      • Nicholson J.K.
      • Holmes E.
      • Kinross J.
      • Burcelin R.
      • Gibson G.
      • Jia W.
      • et al.
      Host-gut microbiota metabolic interactions.
      ].
      HAP [
      • Fouladi F.
      • Bailey M.J.
      • Patterson W.B.
      • Sioda M.
      • Blakley I.C.
      • Fodor A.A.
      • et al.
      Air pollution exposure is associated with the gut microbiome as revealed by shotgun metagenomic sequencing.
      ]
      Associated with increased CVD comorbidities such as IHD and CAD
      Stress hormonesActivation of the Hypothalamus-Pituitary-Adrenal (HPA) axis results in stress hormones, including glucocorticoids and catecholamines. These compounds result in the so-called “fight or flight” by modulating various body functions such as inflammation, metabolism, CNS function and cardiovascular function [
      • McEwen B.S.
      Central effects of stress hormones in health and disease: understanding the protective and damaging effects of stress and stress mediators.
      ]. In a pathological state caused by excess production either due to continuous stress stimuli, abnormal stress response or malignant tumours, these excess stress hormones can result in metabolic syndrome, excess immune response, EC damage, and atherosclerosis [
      • McEwen B.S.
      Central effects of stress hormones in health and disease: understanding the protective and damaging effects of stress and stress mediators.
      ].
      PM2.5 [
      • Miller D.B.
      • Ghio A.J.
      • Karoly E.D.
      • Bell L.N.
      • Snow S.J.
      • Madden M.C.
      • et al.
      Ozone exposure increases circulating stress hormones and lipid metabolites in humans.
      ]

      O3 [
      • Miller D.B.
      • Ghio A.J.
      • Karoly E.D.
      • Bell L.N.
      • Snow S.J.
      • Madden M.C.
      • et al.
      Ozone exposure increases circulating stress hormones and lipid metabolites in humans.
      ]
      HTN, AMI, and chronic CAD [
      • Steptoe A.
      • Kivimäki M.
      Stress and cardiovascular disease: an update on current knowledge.
      ]
      Abbreviations: PM2.5, particulate matter <2.5 µm; HAP, hazardous air pollutants; HPA, hypothalamus-pituitary-adrenal; IHD, ischaemic heart disease; CAD, coronary artery disease; CNS, central nervous system; EC, endothelial cells; AMI, acute myocardial infarction; HTN, hypertension.

      Discussion

      There is convergent evidence connecting CVD with acute and chronic exposure to HAP, identifying HAP as an independent CVD risk factor [
      • Al-Kindi S.G.
      • Brook R.D.
      • Biswal S.
      • Rajagopalan S.
      Environmental determinants of cardiovascular disease: lessons learned from air pollution.
      ]. Through our advancements in household heating methods, we have seen an enormous reduction in mortality attributable to indoor air pollution. Despite this success, industrialisation has resulted in increases in large-scale ambient outdoor air pollution, meaning the impact of air pollution is no longer confined to the place of the pollutant’s origin.
      Advancements in molecular biology have enabled our understanding of the complex nature of air pollution—highlighting numerous pathways and mediators of myocardial and vascular injury. In this paper we have demonstrated that local and systemic inflammation get triggered in response to most HAPs via activation of cytokines such as interleukins and TNF-α. Inflammation is usually accompanied by oxidative stress and mitochondrial damage. In most scenarios, pro-atherosclerosis mechanisms, such as endothelial dysfunction, lipid metabolism dysfunction, and increased plaque vulnerability, are also activated. Other biological processes that influence the cardiovascular system, such as autonomic dysfunction and alteration of the gut microbiome, may also be triggered. By providing a big-picture view of the currently accepted HAP-related mediators, we can start to build a physiological framework of HAP activity in vivo beyond the alveoli. We can see that, not only is the widely hypothesised local and systemic inflammatory response indeed being activated, but also alternate non-inflammatory pathways, such as endothelial damage and autonomic dysfunction, are involved, and this may point toward a common, yet-to-be-discovered, ‘pre-inflammatory’ mechanism. The systems biology model put forward in this paper can also be used to improve our understanding of the other environmental triggers leading to CVD. It is noteworthy that the focus of most of the human studies used in this paper was short-term exposure to traffic-related air pollution (TRAP). As such, there is a knowledge gap in our understanding of how these proposed bio-mediators differ in response to other sources of air pollution, such as bush fires or sandstorms. Similarly, although, historically, most of our public health knowledge regarding morbidities and mortality attributable to air pollution comes from cohort studies of the long-term effects of HAPs, a great degree of further research needs to be done on long-term effects on mediators of pathophysiological pathways.
      Future directions for researchers should also include examining common pathways for the mediators described in this paper in large-cohort studies. Further, by utilising multiomics (a biological analysis approach in which the data sets are multiple “omes” such as the genome, proteome, transcriptome, epigenome, metabolome, and microbiome) technologies and novel machine-learning techniques, researchers can aim to expand our understanding of pathways leading to activation or deactivation of these mediators. The sub-molecular omic view of diseases, from genomics to metabolomics, equips us to understand ‘pre-inflammatory’ mechanisms that are common between various pathological states. This will be important for our future drug-discovery efforts. Finally, to identify vulnerable or resilient individuals, future studies need to utilise the latest air-quality monitoring technologies and especially high temporal resolution and granular resolution satellite-based data to design long-term assessment of participants’ short-term HAP exposure. This will enable us to risk stratify patients based on their short-term exposomic profile rather than a large-scale long-term estimation of their exposure.
      Beyond laying grounds for future directions of HAP-related research, this paper should also be seen as part of the series of ‘wake-up call’ publications in recent years aimed at policymakers and clinicians [
      • Landrigan P.J.
      • Fuller R.
      • Acosta N.J.R.
      • Adeyi O.
      • Arnold R.
      • Basu N.
      • et al.
      The Lancet Commission on pollution and health.
      ,
      • Rajagopalan S
      • Al-Kindi SG
      • Brook RD
      Air pollution and cardiovascular disease: JACC State-of-the-Art Review.
      ]. Public understanding of the dangers of inhaling HAPs tends to be centred around respiratory implications: the cardiovascular damage may supersede respiratory injuries in susceptible individuals, and this needs to be considered in public health policies, as well as in the day-to-day assessment and education of patients.

      Conclusions

      Despite many studies showing mediators of inflammation and vascular dysfunction in response to air pollution, we are far from being able to apply this to clinical decision making. Whilst we all see distinct individual differences in lung responses to air pollution, host cardiovascular response to air pollution is less evident. If a marker were shown to be causally involved and could be reliably measured as an integrated marker of exposure and host response, it may have potential as a risk measure that could guide management, both in regard to extra vigilance to avoid exposure, as well as more aggressive primary or secondary prevention strategies. Also, whilst all the above is seen to increase in response to air pollution, many are common mediators of inflammation, and the exact mechanisms are often not known. Is it particulates activating systemic factors in the lungs, or are particles carried in cells or vesicles to organs where damage is then observed (e.g., vasculature, heart)? Improved understanding of the activation points may provide insights to novel drug targets.

      Sources of Funding

      The authors report the following financial support for the research, authorship and/or publication of this article: GAF reports grants from National Health and Medical Research Council (Australia), Heart Research Australia and the NSW Office of Health and Medical Research; SMG acknowledges the support of the Parker-Hughes Bequest, a NSW Senior Research Fellowship and the Frecker Family.

      Disclosures and Acknowledgments

      GAF reports personal consulting fees from CSL Ltd, Amgen and Janssen, and grants from Abbott Diagnostic and Sanofi outside the submitted work. The remaining authors have nothing to disclose.
      We would like to thank Dr Zara S. Ali for her generous assistance in completing this review.

      Appendices. Supplementary Data

      References

        • Fowler D.
        • Brimblecombe P.
        • Burrows J.
        • Heal M.R.
        • Grennfelt P.
        • Stevenson D.S.
        • et al.
        A chronology of global air quality.
        Philos Trans A Math Phys Eng Sci. 2020; 37820190314
        • Daiber A.
        • Lelieveld J.
        • Steven S.
        • Oelze M.
        • Kröller-Schön S.
        • Sørensen M.
        • et al.
        The “exposome” concept—how environmental risk factors influence cardiovascular health.
        Acta Biochimica Polonica. 2019; 66: 269-283
        • Cohen A.J.
        • Brauer M.
        • Burnett R.
        • Anderson H.R.
        • Frostad J.
        • Estep K.
        • et al.
        Estimates and 25-year trends of the global burden of disease attributable to ambient air pollution: an analysis of data from the Global Burden of Diseases Study 2015.
        Lancet. 2017; 389: 1907-1918
        • Chan S.H.
        • Van Hee V.C.
        • Bergen S.
        • Szpiro A.A.
        • DeRoo L.A.
        • London S.J.
        • et al.
        Long-term air pollution exposure and blood pressure in the sister study.
        Environ Health Perspect. 2015; 123: 951-958
        • Link M.S.
        • Dockery D.W.
        Air pollution and the triggering of cardiac arrhythmias.
        Curr Opin Cardiol. 2010; 25: 16-22
        • Gold D.R.
        • Samet J.M.
        Air pollution, climate, and heart disease.
        Circulation. 2013; 128: e411-e414
        • Newby D.E.
        • Mannucci P.M.
        • Tell G.S.
        • Baccarelli A.A.
        • Brook R.D.
        • Donaldson K.
        • et al.
        Expert position paper on air pollution and cardiovascular disease.
        Eur Heart J. 2015; 36: 83-93
        • Beelen R.
        • Stafoggia M.
        • Raaschou-Nielsen O.
        • Andersen Z.J.
        • Xun W.W.
        • Katsouyanni K.
        • et al.
        Long-term exposure to air pollution and cardiovascular mortality: an analysis of 22 European cohorts.
        Epidemiology. 2014; 25: 368-378
        • Wang X.
        • Kindzierski W.
        • Kaul P.
        Air pollution and acute myocardial infarction hospital admission in Alberta, Canada: a three-step procedure case-crossover study.
        PLoS One. 2015; 10e0132769
        • Su C.
        • Breitner S.
        • Schneider A.
        • Liu L.
        • Franck U.
        • Peters A.
        • et al.
        Short-term effects of fine particulate air pollution on cardiovascular hospital emergency room visits: a time-series study in Beijing, China.
        Int Arch Occup Environ Health. 2016; 89: 641-657
        • Weichenthal S.
        • Lavigne E.
        • Evans G.
        • Pollitt K.
        • Burnett R.T.
        Ambient PM2.5 and risk of emergency room visits for myocardial infarction: impact of regional PM2.5 oxidative potential: a case-crossover study.
        Environ Health. 2016; 15: 46
        • Hamra G.B.
        • Guha N.
        • Cohen A.
        • Laden F.
        • Raaschou-Nielsen O.
        • Samet J.M.
        • et al.
        Outdoor particulate matter exposure and lung cancer: a systematic review and meta-analysis.
        Environ Health Perspect. 2014; 122: 906-911
        • Meo S.A.
        • Memon A.N.
        • Sheikh S.A.
        • Rouq F.A.
        • Usmani A.M.
        • Hassan A.
        • et al.
        Effect of environmental air pollution on type 2 diabetes mellitus.
        Eur Rev Med Pharmacol Sci. 2015; 19: 123-128
        • Stafoggia M.
        • Cesaroni G.
        • Peters A.
        • Andersen Z.J.
        • Badaloni C.
        • Beelen R.
        • et al.
        Long-term exposure to ambient air pollution and incidence of cerebrovascular events: results from 11 European cohorts within the ESCAPE project.
        Environ Health Perspect. 2014; 122: 919-925
        • Landrigan P.J.
        • Fuller R.
        • Acosta N.J.R.
        • Adeyi O.
        • Arnold R.
        • Basu N.
        • et al.
        The Lancet Commission on pollution and health.
        Lancet. 2018; 391: 462-512
        • Vineis P.
        What Is the exposome and how it can help research on air pollution.
        Emiss. Control Sci. Technol. 2019; 5: 31-36
        • Pozzi R.
        • De Berardis B.
        • Paoletti L.
        • Guastadisegni C.
        Inflammatory mediators induced by coarse (PM2.5–10) and fine (PM2.5) urban air particles in RAW 264.7 cells.
        Toxicology. 2003; 183: 243-254
        • van Eeden S.F.
        • Tan W.C.
        • Suwa T.
        • Mukae H.
        • Terashima T.
        • Fujii T.
        • et al.
        Cytokines involved in the systemic inflammatory response induced by exposure to particulate matter air pollutants (PM10).
        Am J Respir Crit Care Med. 2001; 164: 826-830
        • Fakhrzadeh L.
        • Laskin J.D.
        • Laskin D.L.
        Ozone-induced production of nitric oxide and TNF-α and tissue injury are dependent on NF-κB p50.
        Am J Physiol Lung Cell Mol Physiol. 2004; 287: L279-L285
        • Li H.
        • Han M.
        • Guo L.
        • Li G.
        • Sang N.
        Oxidative stress, endothelial dysfunction and inflammatory response in rat heart to NO2 inhalation exposure.
        Chemosphere. 2011; 82: 1589-1596
        • Tuomisto K.
        • Jousilahti P.
        • Sundvall J.
        • Pajunen P.
        • Salomaa V.
        C-reactive protein, interleukin-6 and tumor necrosis factor alpha as predictors of incident coronary and cardiovascular events and total mortality: a population-based, prospective study.
        Thromb Haemost. 2006; 95: 511-518
        • Vasan R.S.
        • Sullivan L.M.
        • Roubenoff R.
        • Dinarello C.A.
        • Harris T.
        • Benjamin E.J.
        • et al.
        Inflammatory markers and risk of heart failure in elderly subjects without prior myocardial infarction: the Framingham Heart Study.
        Circulation. 2003; 107: 1486-1491
        • Yuan S.
        • Carter P.
        • Bruzelius M.
        • Vithayathil M.
        • Kar S.
        • Mason A.M.
        • et al.
        Effects of tumour necrosis factor on cardiovascular disease and cancer: a two-sample Mendelian randomization study.
        EBioMedicine. 2020; 59102956
        • Xu H.
        • Wang T.
        • Liu S.
        • Brook R.D.
        • Feng B.
        • Zhao Q.
        • et al.
        Extreme levels of air pollution associated with changes in biomarkers of atherosclerotic plaque vulnerability and thrombogenicity in healthy adults.
        Circ Res. 2019; 124: e30-e43
        • Barker J.S.
        • Wu Z.
        • Hunter D.D.
        • Dey R.D.
        Ozone exposure initiates a sequential signaling cascade in airways involving interleukin-1beta release, nerve growth factor secretion, and substance P upregulation.
        J Toxicol Environ Health A. 2015; 78: 397-407
        • Qamar A.
        • Rader D.J.
        Effect of interleukin 1β inhibition in cardiovascular disease.
        Curr Opin Lipidol. 2012; 23
        • Tomei G.
        • Ciarrocca M.
        • Bernardini A.
        • Capozzella A.
        • Fortunato B.R.
        • Pimpinella B.
        • et al.
        Plasma IL-2, NK, IFN-γ, and C3 in male workers exposed to traffic pollutants.
        Environ Toxicol Pharmacol. 2006; 22: 131-135
        • Mostafavi N.
        • Vlaanderen J.
        • Chadeau-Hyam M.
        • Beelen R.
        • Modig L.
        • Palli D.
        • et al.
        Inflammatory markers in relation to long-term air pollution.
        Environ Int. 2015; 81: 1-7
        • Ding R.
        • Gao W.
        • Ostrodci D.H.
        • He Z.
        • Song Y.
        • Ma L.
        • et al.
        Effect of interleukin-2 level and genetic variants on coronary artery disease.
        Inflammation. 2013; 36: 1225-1231
        • Simon A.D.
        • Yazdani S.
        • Wang W.
        • Schwartz A.
        • Rabbani L.E.
        Elevated plasma levels of interleukin-2 and soluble il-2 receptor in ischemic heart disease.
        Clin Cardiol. 2001; 24: 253-256
        • Mirowsky J.E.
        • Dailey L.A.
        • Devlin R.B.
        Differential expression of pro-inflammatory and oxidative stress mediators induced by nitrogen dioxide and ozone in primary human bronchial epithelial cells.
        Inhal Toxicol. 2016; 28: 374-382
        • Panasevich S.
        • Leander K.
        • Rosenlund M.
        • Ljungman P.
        • Bellander T.
        • de Faire U.
        • et al.
        Associations of long- and short-term air pollution exposure with markers of inflammation and coagulation in a population sample.
        Occup Environ Med. 2009; 66: 747
        • Perret J.L.
        • Bowatte G.
        • Lodge C.J.
        • Knibbs L.D.
        • Gurrin L.C.
        • Kandane-Rathnayake R.
        • et al.
        The dose–response association between nitrogen dioxide exposure and serum interleukin-6 concentrations.
        Int J Mol Sci. 2017; 18: 1015
        • Ridker P.M.
        • Rane M.
        Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease.
        Circ Res. 2021; 128: 1728-1746
        • Apostolakis S.
        • Vogiatzi K.
        • Amanatidou V.
        • Spandidos D.A.
        Interleukin 8 and cardiovascular disease.
        Cardiovasc Res. 2009; 84: 353-360
        • Yan Z.
        • Wang J.
        • Li J.
        • Jiang N.
        • Zhang R.
        • Yang W.
        • et al.
        Oxidative stress and endocytosis are involved in upregulation of interleukin-8 expression in airway cells exposed to PM2.5.
        Environ Toxicol. 2016; 31: 1869-1878
        • Kim Y.-M.
        • Reed W.
        • Lenz A.G.
        • Jaspers I.
        • Silbajoris R.
        • Nick H.S.
        • et al.
        Ultrafine carbon particles induce interleukin-8 gene transcription and p38 MAPK activation in normal human bronchial epithelial cells.
        Am J Physiol Lung Cell Mol Physiol. 2005; 288: L432-L441
        • Silbajoris R.
        • Osornio-Vargas Alvaro R.
        • Simmons Steven O.
        • Reed W.
        • Bromberg Philip A.
        • Dailey Lisa A.
        • et al.
        Ambient particulate matter induces interleukin-8 expression through an alternative NF-κB (nuclear factor-kappa b) mechanism in human airway epithelial cells.
        Environ Health Perspect. 2011; 119: 1379-1383
        • Smith K.R.
        • Veranth J.M.
        • Hu A.A.
        • Lighty J.S.
        • Aust A.E.
        Interleukin-8 levels in human lung epithelial cells are increased in response to coal fly ash and vary with the bioavailability of iron, as a function of particle size and source of coal.
        Chem Res Toxicol. 2000; 13: 118-125
        • Wan Q.
        • Liu Z.
        • Yang M.
        • Wu J.
        Acceleratory effects of ambient fine particulate matter on the development and progression of atherosclerosis in apolipoprotein E knockout mice by down-regulating CD4+CD25+Foxp3+ regulatory T cells.
        Toxicol Lett. 2019; 316: 27-34
        • Welsh P.
        • Murray H.M.
        • Ford I.
        • Trompet S.
        • de Craen A.J.M.
        • Jukema J.W.
        • et al.
        Circulating interleukin-10 and risk of cardiovascular events.
        Arterioscler Thromb Vasc Biol. 2011; 31: 2338-2344
        • Rusznak C.
        • Devalia J.L.
        • Sapsford R.J.
        • Davies R.J.
        Ozone-induced mediator release from human bronchial epithelial cells in vitro and the influence of nedocromil sodium.
        Eur Respir J. 1996; 9: 2298
        • Devalia J.L.
        • Campbell A.M.
        • Sapsford R.J.
        • Rusznak C.
        • Quint D.
        • Godard P.
        • et al.
        Effect of nitrogen dioxide on synthesis of inflammatory cytokines expressed by human bronchial epithelial cells in vitro.
        Am J Respir Cell Mol Biol. 1993; 9: 271-278
        • Anzai A.
        • Choi J.L.
        • He S.
        • Fenn A.M.
        • Nairz M.
        • Rattik S.
        • et al.
        The infarcted myocardium solicits GM-CSF for the detrimental oversupply of inflammatory leukocytes.
        J Exp Med. 2017; 214: 3293-3310
        • Nemmar A.
        • Nemery B.
        • Hoet P.H.
        • Vermylen J.
        • Hoylaerts M.F.
        Pulmonary inflammation and thrombogenicity caused by diesel particles in hamsters: role of histamine.
        Am J Respir Crit Care Med. 2003; 168: 1366-1372
        • Signorelli S.S.
        • Oliveri Conti G.
        • Zanobetti A.
        • Baccarelli A.
        • Fiore M.
        • Ferrante M.
        Effect of particulate matter-bound metals exposure on prothrombotic biomarkers: A systematic review.
        Environ Res. 2019; 177108573
        • Yeates D.B.
        • Mauderly J.L.
        Inhaled environmental/occupational irritants and allergens: mechanisms of cardiovascular and systemic responses. Introduction.
        Environ Health Perspect. 2001; 109: 479-481
        • Vallejo Jesus G.
        Role of Toll-like receptors in cardiovascular diseases.
        Clin Sci. 2011; 121: 1-10
        • Fischer F.
        • Schulte H.
        • Mohan S.
        • Tataru M.-C.
        • Köhler E.
        • Assmann G.
        • et al.
        Associations of insulin-like growth factors, insulin-like growth factor binding proteins and acid-labile subunit with coronary heart disease.
        Clin Endocrinol. 2004; 61: 595-602
        • Ma Q.-Y.
        • Huang D.-Y.
        • Zhang H.-J.
        • Wang S.
        • Chen X.-F.
        Exposure to particulate matter 2.5 (PM2.5) induced macrophage-dependent inflammation, characterized by increased Th1/Th17 cytokine secretion and cytotoxicity.
        Int Immunopharmacol. 2017; 50: 139-145
      1. Larini A, Aldinucci C, Bocci V, editors. Ozone as a modulator of the immune system. Proceedings of the XV Ozone World Congress London; 2001.

        • Elyasi A.
        • Voloshyna I.
        • Ahmed S.
        • Kasselman L.J.
        • Behbodikhah J.
        • De Leon J.
        • et al.
        The role of interferon-γ in cardiovascular disease: an update.
        Inflamm Res. 2020; 69: 975-988
        • Yue Y.
        • Meng K.
        • Pu Y.
        • Zhang X.
        Transforming growth factor beta (TGF-β) mediates cardiac fibrosis and induces diabetic cardiomyopathy.
        Diabetes Res Clin Pract. 2017; 133: 124-130
        • Dysart M.M.
        • Galvis B.R.
        • Russell A.G.
        • Barker T.H.
        Environmental particulate (PM2. 5) augments stiffness-induced alveolar epithelial cell mechanoactivation of transforming growth factor beta.
        PLoS One. 2014; 9e106821
        • Deshmane S.L.
        • Kremlev S.
        • Amini S.
        • Sawaya B.E.
        Monocyte chemoattractant protein-1 (MCP-1): an overview.
        J Interferon Cytokine Res. 2009; 29: 313-326
        • Fu H.
        • Liu X.
        • Li W.
        • Zu Y.
        • Zhou F.
        • Shou Q.
        • et al.
        PM2.5 exposure induces inflammatory response in macrophages via the TLR4/COX-2/NF-κB pathway.
        Inflammation. 2020; 43: 1948-1958
        • Zhou W.
        • Chen C.
        • Chen Z.
        • Liu L.
        • Jiang J.
        • Wu Z.
        • et al.
        NLRP3: a novel mediator in cardiovascular disease.
        J Immunol Res. 2018; 20185702103
        • Hu T.
        • Zhu P.
        • Liu Y.
        • Zhu H.
        • Geng J.
        • Wang B.
        • et al.
        PM2 .5 induces endothelial dysfunction via activating NLRP3 inflammasome.
        Environ Toxicol. 2021; 36: 1886-1893
        • Wang Y.
        • Zhou Y.
        • Graves D.T.
        FOXO Transcription factors: their clinical significance and regulation.
        BioMed Res Int. 2014; 2014925350
        • Su X.
        • Tian J.
        • Li B.
        • Zhou L.
        • Kang H.
        • Pei Z.
        • et al.
        Ambient PM2.5 caused cardiac dysfunction through FoxO1-targeted cardiac hypertrophy and macrophage-activated fibrosis in mice.
        Chemosphere. 2020; 247125881
        • Sun Q.
        • Yue P.
        • Deiuliis J.A.
        • Lumeng C.N.
        • Kampfrath T.
        • Mikolaj M.B.
        • et al.
        Ambient air pollution exaggerates adipose inflammation and insulin resistance in a mouse model of diet-induced obesity.
        Circulation. 2009; 119: 538-546
        • Xu Z.
        • Xu X.
        • Zhong M.
        • Hotchkiss I.P.
        • Lewandowski R.P.
        • Wagner J.G.
        • et al.
        Ambient particulate air pollution induces oxidative stress and alterations of mitochondria and gene expression in brown and white adipose tissues.
        Part Fibre Toxicol. 2011; 8: 20
        • Feng J.
        • Cavallero S.
        • Hsiai T.
        • Li R.
        Impact of air pollution on intestinal redox lipidome and microbiome.
        Free Radic Biol Med. 2020; 151: 99-110
        • Haberzettl P.
        • Jin L.
        • Riggs D.W.
        • Zhao J.
        • O’Toole T.E.
        • Conklin D.J.
        Fine particulate matter air pollution and aortic perivascular adipose tissue: oxidative stress, leptin, and vascular dysfunction.
        Physiol Rep. 2021; 9e14980
        • Dakroub A.
        • Nasser S.A.
        • Kobeissy F.
        • Yassine H.M.
        • Orekhov A.
        • Sharifi-Rad J.
        • et al.
        Visfatin: an emerging adipocytokine bridging the gap in the evolution of cardiovascular diseases.
        J Cell Physiol. 2021; 236: 6282-6296
        • Wan Q.
        • Cui X.
        • Shao J.
        • Zhou F.
        • Jia Y.
        • Sun X.
        • et al.
        Beijing ambient particle exposure accelerates atherosclerosis in ApoE knockout mice by upregulating visfatin expression.
        Cell Stress Chaperones. 2014; 19: 715-724
        • Vaughan D.E.
        PAI-1 and atherothrombosis.
        J Thromb Haemost. 2005; 3: 1879-1883
        • Chen R.
        • Zhao Z.
        • Sun Q.
        • Lin Z.
        • Zhao A.
        • Wang C.
        • et al.
        Size-fractionated particulate air pollution and circulating biomarkers of inflammation, coagulation, and vasoconstriction in a panel of young adults.
        Epidemiology. 2015; 26: 328-336
        • Wu S.
        • Deng F.
        • Wei H.
        • Huang J.
        • Wang H.
        • Shima M.
        • et al.
        Chemical constituents of ambient particulate air pollution and biomarkers of inflammation, coagulation and homocysteine in healthy adults: a prospective panel study.
        Part Fibre Toxicol. 2012; 9: 49
        • Jamaluddin M.S.
        • Weakley S.M.
        • Yao Q.
        • Chen C.
        Resistin: functional roles and therapeutic considerations for cardiovascular disease.
        B J Pharmacol. 2012; 165: 622-632
        • Szmitko P.E.
        • Teoh H.
        • Stewart D.J.
        • Verma S.
        Adiponectin and cardiovascular disease: state of the art?.
        Am J Physiol Heart Circ Physiol. 2007; 292: H1655-H1663
        • Wan Q.
        • Ding T.
        • Xu Y.
        • Zheng C.
        • Tu M.
        • Zhao T.
        Urban fine particulate air pollution exposure promotes atherosclerosis in apolipoprotein E-deficient mice by activating perivascular adipose tissue inflammation via the Wnt5a/Ror2 signaling pathway.
        Ecotoxicol Environ Saf. 2021; 227112912
        • D’Oria R.
        • Schipani R.
        • Leonardini A.
        • Natalicchio A.
        • Perrini S.
        • Cignarelli A.
        • et al.
        The role of oxidative stress in cardiac disease: from physiological response to injury factor.
        Oxid Med Cell Longev. 2020; 20205732956
        • Gao D.
        • Ripley S.
        • Weichenthal S.
        • Godri Pollitt K.J.
        Ambient particulate matter oxidative potential: chemical determinants, associated health effects, and strategies for risk management.
        Free Radic Biol Med. 2020; 151: 7-25
        • Xu M.-X.
        • Ge C.-X.
        • Qin Y.-T.
        • Gu T.-T.
        • Lou D.-S.
        • Li Q.
        • et al.
        Prolonged PM2.5 exposure elevates risk of oxidative stress-driven nonalcoholic fatty liver disease by triggering increase of dyslipidemia.
        Free Radic Biol Med. 2019; 130: 542-556
        • Ying Z.
        • Xu X.
        • Chen M.
        • Liu D.
        • Zhong M.
        • Chen L.-c.
        • et al.
        A synergistic vascular effect of airborne particulate matter and nickel in a mouse model.
        Toxicol Sci. 2013; 135: 72-80
        • Shimokawa H.
        • Tsutsui M.
        Nitric oxide synthases in the pathogenesis of cardiovascular disease.
        Pflügers Arch. 2010; 459: 959-967
        • Sirker A.
        • Zhang M.
        • Shah A.M.
        NADPH oxidases in cardiovascular disease: insights from in vivo models and clinical studies.
        Basic Res Cardiol. 2011; 106: 735-747
        • Cáceres L.
        • Paz M.L.
        • Garcés M.
        • Calabró V.
        • Magnani N.D.
        • Martinefski M.
        • et al.
        NADPH oxidase and mitochondria are relevant sources of superoxide anion in the oxinflammatory response of macrophages exposed to airborne particulate matter.
        Ecotoxicol Environ Saf. 2020; 205111186
        • Simkhovich B.Z.
        • Kleinman M.T.
        • Mehrian-Shai R.
        • Hsu Y.-H.
        • Meacher D.
        • Gookin G.
        • et al.
        Chronic exposure to ambient particulate matter alters cardiac gene expression patterns and markers of oxidative stress in rats.
        Air Qual Atmos Health. 2011; 4: 15-25
        • Hunter A.L.
        • Cruz R.P.
        • Cheyne B.M.
        • McManus B.M.
        • Granville D.J.
        Cytochrome p450 enzymes and cardiovascular disease.
        Can J Physiol Pharmacol. 2004; 82: 1053-1060
        • Dominic E.A.
        • Ramezani A.
        • Anker S.D.
        • Verma M.
        • Mehta N.
        • Rao M.
        Mitochondrial cytopathies and cardiovascular disease.
        Heart. 2014; 100: 611-618
        • Mikhed Y.
        • Daiber A.
        • Steven S.
        Mitochondrial Oxidative Stress, Mitochondrial DNA damage and their role in age-related vascular dysfunction.
        Int J Mol Sci. 2015; 16: 15918-15953
        • West A.P.
        • Shadel G.S.
        Mitochondrial DNA in innate immune responses and inflammatory pathology.
        Nat Rev Immunol. 2017; 17: 363-375
        • Nakayama H.
        • Otsu K.
        Mitochondrial DNA as an inflammatory mediator in cardiovascular diseases.
        Biochem J. 2018; 475: 839-852
        • Ding Z.
        • Liu S.
        • Wang X.
        • Khaidakov M.
        • Dai Y.
        • Mehta J.L.
        Oxidant stress in mitochondrial DNA damage, autophagy and inflammation in atherosclerosis.
        Sci Rep. 2013; 3
        • Zhang Z.
        • Meng P.
        • Han Y.
        • Shen C.
        • Li B.
        • Hakim M.A.
        • et al.
        Mitochondrial DNA-LL-37 Complex promotes atherosclerosis by escaping from autophagic recognition.
        Immunity. 2015; 43: 1137-1147
        • Byun H.M.
        • Colicino E.
        • Trevisi L.
        • Fan T.
        • Christiani D.C.
        • Baccarelli A.A.
        Effects of air pollution and blood mitochondrial DNA methylation on markers of heart rate variability.
        J Am Heart Assoc. 2016; 5e003218
        • Boovarahan S.R.
        • Kurian G.A.
        Mitochondrial dysfunction: a key player in the pathogenesis of cardiovascular diseases linked to air pollution.
        Rev Environ Health. 2018; 33: 111-122
        • Suematsu N.
        • Tsutsui H.
        • Wen J.
        • Kang D.
        • Ikeuchi M.
        • Ide T.
        • et al.
        Oxidative stress mediates tumor necrosis factor-α–induced mitochondrial DNA damage and dysfunction in cardiac myocytes.
        Circulation. 2003; 107: 1418-1423
        • Ballinger S.W.
        • Patterson C.
        • Knight-Lozano C.A.
        • Burow D.L.
        • Conklin C.A.
        • Hu Z.
        • et al.
        Mitochondrial integrity and function in atherogenesis.
        Circulation. 2002; 106: 544-549
        • Oka T.
        • Hikoso S.
        • Yamaguchi O.
        • Taneike M.
        • Takeda T.
        • Tamai T.
        • et al.
        Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure.
        Nature. 2012; 485: 251-255
        • Ballinger S.W.
        • Patterson C.
        • Yan C.-N.
        • Doan R.
        • Burow D.L.
        • Young C.G.
        • et al.
        Hydrogen peroxide–and peroxynitrite-induced mitochondrial DNA damage and dysfunction in vascular endothelial and smooth muscle cells.
        Circ Res. 2000; 86: 960-966
        • Xu S.
        • Ilyas I.
        • Little P.J.
        • Li H.
        • Kamato D.
        • Zheng X.
        • et al.
        Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies.
        Pharmacol Rev. 2021; 73: 924-967
        • Liang Q.
        • Sun M.
        • Wang F.
        • Ma Y.
        • Lin L.
        • Li T.
        • et al.
        Short-term PM2.5 exposure and circulating von Willebrand factor level: a meta-analysis.
        Sci Total Environ. 2020; 737140180
        • Bo L.
        • Jiang S.
        • Xie Y.
        • Kan H.
        • Song W.
        • Zhao J.
        Effect of vitamin E and omega-3 fatty acids on protecting ambient PM2.5-induced inflammatory response and oxidative stress in vascular endothelial cells.
        PL0S One. 2016; 11e0152216
        • Dignat-George F.
        • Boulanger C.M.
        The many faces of endothelial microparticles.
        Arterioscler Thromb Vasc Biol. 2011; 31: 27-33
        • Pope C.A.
        • Bhatnagar A.
        • McCracken J.P.
        • Abplanalp W.
        • Conklin D.J.
        • O’Toole T.
        Exposure to fine particulate air pollution is associated with endothelial injury and systemic inflammation.
        Circ Res. 2016; 119: 1204-1214
        • Niessen H.W.M.
        • Krijnen P.A.J.
        • Visser C.A.
        • Meijer C.J.L.M.
        • Hack C.E.
        Intercellular adhesion molecule-1 in the heart.
        Ann N Y Acad Sci. 2002; 973: 573-585
        • Xie W.
        • You J.
        • Zhi C.
        • Li L.
        The toxicity of ambient fine particulate matter (PM2.5) to vascular endothelial cells.
        J Appl Toxicol. 2021; 41: 713-723
        • Yatera K.
        • Hsieh J.
        • Hogg J.C.
        • Tranfield E.
        • Suzuki H.
        • Shih C.-H.
        • et al.
        Particulate matter air pollution exposure promotes recruitment of monocytes into atherosclerotic plaques.
        Am J Physiol Heart Circ Physiol. 2008; 294: H944-H953
        • Troncoso M.F.
        • Ortiz-Quintero J.
        • Garrido-Moreno V.
        • Sanhueza-Olivares F.
        • Guerrero-Moncayo A.
        • Chiong M.
        • et al.
        VCAM-1 as a predictor biomarker in cardiovascular disease.
        Biochim Biophys Acta Mol Basis Dis. 2021; 1867166170
        • Liu J.
        • Sun Q.
        • Sun M.
        • Lin L.
        • Ren X.
        • Li T.
        • et al.
        Melatonin alleviates PM2.5-triggered macrophage M1 polarization and atherosclerosis via regulating NOX2-mediated oxidative stress homeostasis.
        Free Radic Biol Med. 2022; 181: 166-179
        • Akhmedov A.
        • Sawamura T.
        • Chen C.-H.
        • Kraler S.
        • Vdovenko D.
        • Lüscher T.F.
        Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1): a crucial driver of atherosclerotic cardiovascular disease.
        Eur Heart J. 2020; 42: 1797-1807
        • Ferrario C.M.
        Role of Angiotensin II in Cardiovascular Disease — Therapeutic implications of more than a century of research.
        J Renin Angiotensin Aldosterone Syst. 2006; 7: 3-14
        • Xu X.
        • Qimuge A.
        • Wang H.
        • Xing C.
        • Gu Y.
        • Liu S.
        • et al.
        IRE1α/XBP1s branch of UPR links HIF1α activation to mediate ANGII-dependent endothelial dysfunction under particulate matter (PM) 2.5 exposure.
        Sci Rep. 2017; 7
        • Saraswati S.
        • Lietman C.D.
        • Li B.
        • Mathew S.
        • Zent R.
        • Young P.P.
        Small proline-rich repeat 3 is a novel coordinator of PDGFRβ and integrin β1 crosstalk to augment proliferation and matrix synthesis by cardiac fibroblasts.
        FASEB J. 2020; 34: 7885-7904
        • Bae J.-E.
        • Choi H.
        • Shin D.W.
        • Na H.-W.
        • Park N.Y.
        • Kim J.B.
        • et al.
        Fine particulate matter (PM2.5) inhibits ciliogenesis by increasing SPRR3 expression via c-Jun activation in RPE cells and skin keratinocytes.
        Sci Rep. 2019; 9
        • Chistiakov D.A.
        • Sobenin I.A.
        • Orekhov A.N.
        Vascular extracellular matrix in atherosclerosis.
        Cardiol Rev. 2013; 21: 270-288
        • Mosevoll K.A.
        • Johansen S.
        • Wendelbo Ø.
        • Nepstad I.
        • Bruserud Ø.
        • Reikvam H.
        Cytokines, adhesion molecules, and matrix metalloproteases as predisposing, diagnostic, and prognostic factors in venous thrombosis.
        Front Med (Lausanne). 2018; 5: 147
        • Messerli F.H.
        TIMPs, MMPs and cardiovascular disease.
        Eur Heart J. 2004; 25: 1475-1476
        • Hu D.
        • Jia X.
        • Cui L.
        • Liu J.
        • Chen J.
        • Wang Y.
        • et al.
        Exposure to fine particulate matter promotes platelet activation and thrombosis via obesity-related inflammation.
        J Hazard Mater. 2021; 413125341
        • Yacoub D.
        • Hachem A.
        • Théorêt J.-F.
        • Gillis M.-A.
        • Mourad W.
        • Merhi Y.
        Enhanced levels of soluble CD40 ligand exacerbate platelet aggregation and thrombus formation through a CD40-dependent tumor necrosis factor receptor-associated factor-2/Rac1/p38 mitogen-activated protein kinase signaling pathway.
        Arterioscler Thromb Vasc Biol. 2010; 30: 2424-2433
        • Frenette P.S.
        • Denis C.V.
        • Weiss L.
        • Jurk K.
        • Subbarao S.
        • Kehrel B.
        • et al.
        P-Selectin glycoprotein ligand 1 (PSGL-1) is expressed on platelets and can mediate platelet-endothelial interactions in vivo.
        J Exp Med. 2000; 191: 1413-1422
        • Peçanha T.
        • Silva-Júnior N.D.
        • Forjaz CLdM.
        Heart rate recovery: autonomic determinants, methods of assessment and association with mortality and cardiovascular diseases.
        Clin Physiol Funct Imaging. 2014; 34: 327-339
        • Perez C.M.
        • Hazari M.S.
        • Farraj A.K.
        Role of autonomic reflex arcs in cardiovascular responses to air pollution exposure.
        Cardiovasc Toxicol. 2015; 15: 69-78
        • Huang W.
        • Zhu T.
        • Pan X.
        • Hu M.
        • Lu S.-E.
        • Lin Y.
        • et al.
        Air pollution and autonomic and vascular dysfunction in patients with cardiovascular disease: interactions of systemic inflammation, overweight, and gender.
        Am J Epidemiol. 2012; 176: 117-126
        • Taylor-Clark T.E.
        Air pollution-induced autonomic modulation.
        Physiology. 2020; 35: 363-374
        • Zhong J.
        • Colicino E.
        • Lin X.
        • Mehta A.
        • Kloog I.
        • Zanobetti A.
        • et al.
        Cardiac autonomic dysfunction: particulate air pollution effects are modulated by epigenetic immunoregulation of Toll-like receptor 2 and dietary flavonoid intake.
        J Am Heart Assoc. 2015; 4e001423
        • Grace M.S.
        • Belvisi M.G.
        TRPA1 receptors in cough.
        Pulm Pharmacol Ther. 2011; 24: 286-288
        • Hazari M.S.
        • Haykal-Coates N.
        • Winsett D.W.
        • Krantz Q.T.
        • King C.
        • Costa D.L.
        • et al.
        TRPA1 and sympathetic activation contribute to increased risk of triggered cardiac arrhythmias in hypertensive rats exposed to diesel exhaust.
        Environ Health Perspect. 2011; 119: 951-957
        • Bessac B.F.
        • Jordt S.-E.
        Breathtaking TRP channels: TRPA1 and TRPV1 in airway chemosensation and reflex control.
        Physiology (Bethesda). 2008; 23: 360-370
        • Barth K.
        • Kasper M.
        Membrane compartments and purinergic signalling: occurrence and function of P2X receptors in lung.
        FEBS J. 2009; 276: 341-353
        • Kraneveld A.D.
        • Nijkamp F.P.
        Tachykinins and neuro-immune interactions in asthma.
        Int Immunopharmacol. 2001; 1: 1629-1650
        • O'Connor T.M.
        • O'Connell J.
        • O'Brien D.I.
        • Goode T.
        • Bredin C.P.
        • Shanahan F.
        The role of substance P in inflammatory disease.
        J Cell Physiol. 2004; 201: 167-180
        • Quyyumi A.A.
        • Mulcahy D.
        • Andrews N.P.
        • Husain S.
        • Panza J.A.
        • Cannon R.O.
        Coronary vascular nitric oxide activity in hypertension and hypercholesterolemia.
        Circulation. 1997; 95: 104-110
        • Kim D.Y.
        • Piao J.
        • Hong H.S.
        Substance-P inhibits cardiac microvascular endothelial dysfunction caused by high glucose-induced oxidative stress.
        Antioxidants. 2021; 10
        • Levick S.P.
        • Brower G.L.
        • Janicki J.S.
        Substance P-mediated cardiac mast cell activation: an in vitro study.
        Neuropeptides. 2019; 74: 52-59