Skip to main content
Antioxidants & Redox Signaling logoLink to Antioxidants & Redox Signaling
. 2023 May 3;38(13):1041–1069. doi: 10.1089/ars.2022.0087

Cigarette Smoke Modulates Inflammation and Immunity via Reactive Oxygen Species-Regulated Trained Immunity and Trained Tolerance Mechanisms

Fatma Saaoud 1, Ying Shao 1, William Cornwell 2, Hong Wang 3, Thomas J Rogers 2, Xiaofeng Yang 1,3,
PMCID: PMC10171958  PMID: 36017612

Abstract

Significance:

Cigarette smoke (CS) is a prominent cause of morbidity and death and poses a serious challenge to the current health care system worldwide. Its multifaceted roles have led to cardiovascular, respiratory, immunological, and neoplastic diseases.

Recent Advances:

CS influences both innate and adaptive immunity and regulates immune responses by exacerbating pathogenic immunological responses and/or suppressing defense immunity. There is substantial evidence pointing toward a critical role of CS in vascular immunopathology, but a comprehensive and up-to-date review is lacking.

Critical Issues:

This review aims to synthesize novel conceptual advances on the immunomodulatory action of CS with a focus on the cardiovascular system from the following perspectives: (i) the signaling of danger-associated molecular pattern (DAMP) receptors contributes to CS modulation of inflammation and immunity; (ii) CS reprograms immunometabolism and trained immunity-related metabolic pathways in innate immune cells and T cells, which can be sensed by the cytoplasmic (cytosolic and non-nuclear organelles) reactive oxygen species (ROS) system in vascular cells; (iii) how nuclear ROS drive CS-promoted DNA damage and cell death pathways, thereby amplifying inflammation and immune responses; and (iv) CS induces endothelial cell (EC) dysfunction and vascular inflammation to promote cardiovascular diseases (CVDs).

Future Directions:

Despite significant progress in understanding the cellular and molecular mechanisms linking CS to immunity, further investigations are warranted to elucidate novel mechanisms responsible for CS-mediated immunopathology of CVDs; in particular, the research in redox regulation of immune functions of ECs and their fate affected by CS is still in its infancy.

Keywords: cigarette smoke, morphine, trained immunity, trained tolerance, immunometabolism, cell death

Introduction

Cigarette smoke (CS) accounts for 480,000 adult death annually (1/5 all the deaths/year; 1300 deaths/day) (Warren et al, 2014) and 41,000 deaths from secondhand smoke exposure in the United States (Jamal et al, 2016). Annual economic costs of diseases associated with CS exceed $289 billion (Kohut, 2017). CS contributes significantly to the pathogenesis of several types of major immune-inflammatory diseases including coronary heart disease (CHD) (Centers for Disease Control (CDC), 1989), lung cancers, chronic obstructive pulmonary disease (COPD) (Ishii, 2013), end-stage renal disease (Choi et al, 2019), multiple sclerosis (Alrouji et al, 2019), stroke, and oral inflammatory disease (Table 1).

Table 1.

Cigarette Smoke Promotes Several Types of Major Immunoinflammatory Diseases

Diseases Effect of cigarette smoke PMID
CVD Increased neutrophils, lymphocytes, and monocytes
Increased inflammatory cytokines and chemokines such as IL-1β and TNF-α
Induced endothelial vascular dysfunction and inflammation
Increased oxidative stress and ROS production
Increased total serum cholesterol, VLDL, LDL, and triglyceride concentrations
Increased risk of thrombosis, atherosclerosis, stroke, and CHD
Increased cardiovascular morbidity and mortality
17485580
21285293
15145091
26174518
Respiratory diseases Increased production of immune mediators and ROS by lung macrophages
Increased incidence of COPD and airway inflammation
Increased risk of interstitial lung diseases, bronchial asthma, and lung cancer
22196881
24507834
23631228
CKD Increased production of proinflammatory mediators
Increased EC dysfunction
Increased oxidative stress and ROS production
Increased cardiovascular morbidity and mortality in CKD patients
Increased risk of incident ESRD
18003763
28339863
22158113
31862942
RA Increased risk for development and severity of RA
Increased rheumatoid nodule formation with multiple joint involvements
24594022
25479074
MS Increased incidence and severity of the disease 32905534
SLE Increased inflammatory markers associated with decreased anti-inflammatory IL-10
Increased oxidative stress
Elevated titers of anti-dsDNA
Increased risk of developing SLE
29724134
IBD Impaired function of inflammatory cells
Increased recruitment of CD4+ and CD8+ T cells, and of CD11b+ DCs
Increased proinflammatory chemokines/cytokines (CCR6, CCL20, IL-8)
increased in the expression of MHC-II and costimulatory molecules
Increased incidence of Crohn's disease
Protected against UC by decreased inflammatory mediators and cytokine and decreased gut permeability by nicotine
20333390
21112082
24691114
21537330
32823518
Oral inflammatory diseases Impaired function of inflammatory cells
Increased inflammatory cytokines
Promoted periodontal diseases
20361572

CS increases ROS production, inflammatory cytokine secretion, inflammatory and immune cells, and impairment of inflammatory cells' function. Complex roles of CS have resulted in several diseases, including CVDs, respiratory diseases, CKD, rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, and inflammatory bowel disease.

anti-dsDNA, anti-double-stranded DNA; CCL20, chemokine (C-C motif) ligand 20; CCR6, C-C motif chemokine receptor 6; CHD, coronary heart disease; CKD, chronic kidney disease; COPD, chronic obstructive pulmonary disease; CS, cigarrette smoke; CVD, cardiovascular diseases; DC, dendritic cell; EC, endothelial cell; ESRD, end-stage kidney disease; IBD, inflammatory bowel disease; IL, interleukin; LDL, low-density lipoprotein; MHC-II, MHC class II molecules; MS, multiple sclerosis; RA, rheumatoid arthritis; ROS, reactive oxygen species; SLE, systemic lupus erythematosus; TNF-α, tumor necrosis factor-α; UC, ulcerative colitis; VLDL, very-low-density lipoprotein.

The mechanisms responsible for these effects include increased reactive oxygen species (ROS), membrane receptor (clusters of differentiation)-mediated cell–cell interaction (Horvathova et al, 2009; Xu et al, 2021), noncoding RNA-containing exosome secretion (Maccani and Knopik, 2012; Yang et al, 2017b), and proinflammatory cytokine secretion (Lu et al, 2022; Ni et al, 2021; Zhang et al, 2020a; Zhang et al, 2020b), which lead to both an upregulation in the numbers of leukocytes and an impairment of the function of inflammatory cells. CS induces chronic inflammation in the lungs (Duaso and Duncan, 2012; Wood and Stockley, 2007), which is apparent at the level of tissue damage, and may result in COPD (Jaspers, 2014).

Tobacco smoke (TS) is a carcinogenic and immunomodulatory toxic mixture of more than 5000 chemicals (Talhout et al, 2011). Chronic exposure to CS leads to impaired T cell function (Valiathan et al, 2014), reprogramming of Treg transcriptomes (Shao et al, 2022; Shao et al, 2021b), a heightened level of systemic inflammation (Stämpfli and Anderson, 2009), and depressed antiviral immune responses, particularly in the lungs (Bauer et al, 2013). CS may increase end-organ injury (including atherosclerosis) by activating endothelial cells (ECs), a new innate immune cell type as we proposed (Drummer et al, 2021a; Mai et al, 2013; Shao et al, 2020), and promoting EC–monocyte interactions and increasing leukocyte tissue infiltration. CS induces chronic inflammation by increasing the numbers of neutrophils and macrophages (Jaspers, 2014).

CS administration leads to decreased CD4+ forkhead box P3 (Foxp3)+ T regulatory cells (Tregs, the major anti-inflammation cell type) and reduced expression of anti-inflammatory/immunosuppressive cytokine interleukin (IL)-10 and Treg-specific transcription factor Foxp3. For example, the frequency of Treg cells is lower in CS-exposed mice compared with the control group. More importantly, the frequency of Treg cells is negatively correlated with T helper cell 17 (Th17 cells) and CD8+ IL-17 producing T cells (Tc17 cells) (Duan et al, 2016).

CS is a major reversible risk factor for cardiovascular disease (CVD). CS-induced vascular inflammation and vascular pathology play important roles in the development and progression of subsequent clinical outcomes (Ambrose and Barua, 2004). CS has various cytotoxic effects in a wide range of vascular cell types including vascular ECs and vascular smooth muscle cells (VSMCs), in part, by increasing ROS production, activating nuclear factor kappa B (NF-κB), upregulating adhesion molecules, inducing endothelial activation and endothelial dysfunction, and promoting VSMC phenotypic switching and VSMC matrix degradation. The adhesion of leukocytes on the surface of ECs is an early event in the atherogenesis process and the increased levels of proinflammatory cytokines promote leukocyte-EC adhesion and leukocyte recruitment (Xu et al, 2022; Xu et al, 2021).

CS increases the expression of vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and E-selectin on human umbilical vein endothelial cells (HUVECs), and increased transendothelial migration of monocytes and neutrophils (Bermudez et al, 2002; Shen et al, 1996). In addition, CS induces ferroptosis cell death in VSMCs (Sampilvanjil et al, 2020) and reduces carotid artery VSMC numbers due to increased cell necrosis (Ambalavanan et al, 2001).

Nicotine exposure promotes VSMC migration from the tunica media to atheromatous plaques in the vascular intima and induces VSMC transformation from contractile to synthetic phenotype (VSMC phenotype switching) via nicotinic acetylcholine receptors and G protein-coupled receptors (Yoshiyama et al, 2014). Electronic cigarettes (e-cigarettes), also known as electronic nicotine delivery systems, have become popular as substitutes for conventional tobacco cigarettes. While e-cigarettes contain fewer toxicants than tobacco cigarettes, they still emit detectable levels of volatile organic compounds, aldehydes, and nitrosamines that induce oxidative stress and inflammation (Cheng, 2014). E-cigarette-exposed alveolar macrophages show a significant increase in the production of proinflammatory cytokines/chemokines, apoptosis and necrosis, and ROS production (Scott et al, 2018). Furthermore, e-cigarette exposure significantly increases oxidative stress, oxidized low-density lipoprotein (ox-LDL), and cardiovascular risk (Carnevale et al, 2016; Moheimani et al, 2017).

Chronic e-cigarette exposure induces vascular endothelial dysfunction (Shao et al, 2014), cardiac dysfunction, and atherosclerosis in mice (Espinoza-Derout et al, 2019). In addition, e-cigarette exposure increases the risk for COPD (Bowler et al, 2017) and CVDs (D'Amario et al, 2019).

Signals of Danger-Associated Molecular Pattern Receptors Contribute to the Modulation of Inflammation and Immunity by CS

TS contains several chemicals that are harmful. Among the more than 5000 chemicals identified, at least 250 are reported to be harmful, including hydrogen cyanide, carbon monoxide (CO1), and ammonia (Centers for Disease Control and Prevention et al, 2010; National Center for Chronic Disease Prevention and Health Promotion Office on Smoking and Health, 2014). Based on chemical binding capacities, we tentatively classify them into five groups as follows:

  • (i)

    At least 72 chemicals cause cancer (carcinogens) and some of these covalently bind to DNA directly (Goodson et al, 2015) and form DNA adducts. Twenty-eight of the carcinogenic chemicals including polycyclic aromatic hydrocarbons (benzo[a]pyrene), N-nitrosamines, heavy metals (nickel, cadmium, chromium, and arsenic), alkaloids (nicotine and its major metabolite, cotinine), and aromatic amines have immunomodulatory effects on distinct innate and adaptive immune cells and can lead to impaired immunity. These compounds can either downregulate (anti-inflammatory) or upregulate (proinflammatory) the immune response, leading to either an increased susceptibility to the development of cancers or infectious diseases, or immunopotentiation resulting in an increased secretion of inflammatory mediators (Table 2).

  • (ii)

    The second group consists of more than 400 odorants. There are distinctive and clinically relevant odorants that evokes a response from 144 odorant receptors (ORs) and 3 trace-amine-associated receptors. The recognition of CS is accomplished by a broad receptor response pattern, and 1-pentanethiol (the odorant most critical for perception of the artificial mimic) is responsible for a small subset of the responsive ORs in this combinatorial code (McClintock et al, 2020). The ratios of >400 odorants over 144 ORs suggest significant degeneracy of CS chemicals in binding to their cellular receptors, about 2.7 odorants/receptors in nonhigh odorant-specific manners.

  • (iii)

    Danger-associated molecular pattern (DAMP)/pathogen-associated molecular pattern (PAMP) receptors are the third group. These include toll-like receptors (TLRs), C-type lectin receptors, NOD-like receptors (NLRs including NLRP3), retinoic acid-inducible gene-like receptors, cytosolic DNA sensors, receptor for advanced glycation end products, triggering receptors expressed on myeloid cells, G-protein-coupled receptors, ion channels (Gong et al, 2020), and conditional DAMP receptors (Wang et al, 2016b). Compared with wild-type (WT) mice, CS-induced pulmonary inflammation is unaltered in DAMP/PAMP receptors and TLR2-deficient (Tlr2−/−) (Yang et al, 2008) and TLR4-deficient (Tlr4−/−) mice. CS-induced airway fibrosis, characterized by increased collagen deposition around small airways, is not altered in Tlr2−/− mice, but is attenuated in Tlr4−/− mice compared with CS-exposed WT controls (Haw et al, 2018). In addition, CS amplifies inflammatory reactions and atherogenesis through activation of the H1R-TLR2/4-cyclooxygenase-2 (COX-2) axis (Barua et al, 2015).

  • (iv)

    CS inhibits the NLRP3 inflammasome and leads to caspase-1 activation via the TLR4-toll-like receptor adaptor molecule 1 (TICAM1, TRIF)-caspase-8 axis in human macrophages (Buscetta et al, 2020).

  • (v)

    The compounds induced by CS and shared with endogenous metabolites (Zhang et al, 2022) as well as chemokines induced by morphine (Rogers, 2020) can use their intrinsic receptors [conditional DAMP receptors as we proposed (Wang et al, 2016b)] to drive or modulate inflammatory signals (Muri and Kopf, 2021; Voss et al, 2021).

Table 2.

Carcinogenic Constituents from Cigarette Smoke Induce Reactive Oxygen Species Production and Oxidative DNA Damage, Inhibit Immune Cell Generation and Proliferation, Modulate Cytokine Secretion, and Suppress Immune Functions

Carcinogen Immune cells/tissue Effects Proinflammatory (+) Anti-inflammatory (*) Receptor/pathway PMID
Benz[j]aceanthrylene Macrophages DNA damage     9054606
Lymphocytes DNA damage     9744557
Benz[a]anthracene Lymphocytes Increased ROS production and oxidative DNA damage     21888224
T cells Inhibited T cell proliferation
Inhibited IL-2 production
Induced immunosuppression
* Signal transduction mediated by TCR and IL-2R 8931739
T cells Suppressed helper T cell activation     6219909
Benzo[b]fluoranthene HPBECs Increased innate and adaptive immune signals
Increased expression of oxidative stress genes
+   30090392
Macrophages Increased secretion of inflammatory cytokines +   18830893
Benzo[j]fluoranthene Mouse skin A potent tumor initiator with high tumorigenic activity     3677067
B[k]F Mouse Reduced T cells in thymus and spleen
Reduced CD4+ IL-2+ cells
* Immunosuppression through IL-2 production 16326422
BaP BMDMs Impaired proliferation and decreased number of mature cells
Increased expression of F4/80 and MHC-II
Increased TNF-α and IL-10
+ AhR 30099064
Mouse/mouse epidermal cells Increased neutrophil and macrophage accumulation
Increased secretion of IL-5, IL-13, IL-33, MCP-1
Increased expression of MHC-II and CD86 expression
Increased Th1/Th2 responses and allergic airway inflammation Induced VEGF
PI-3K/AP-1 activation
+   26918773
16461351
T cells Increased expression of AhR and CYP1 expression   Activation of AhR/CYP1-metabolizing enzymes 28461126
24412381
BMDMs Decreased proinflammatory cytokines
Increased expression of IL-10, MHC-II, CD14, Fcγ receptor I (FcγRI/CD64)
Increased NO production and phagocytosis
* AhR activation 30053493
DCs and keratinocytes Increased secretion of cytokines + AhR activation 30748024
T cells Inhibited T cell proliferation
Decreased production of IFN-γ, IL-2, and IL-4
* Ca2+/CaM/NF-κB and Ca2+/CaM/CaN/NFAT signal transduction pathways 28290727
Cyclopenta[c,d]pyrene Leukocytes Increased DNA adducts     8200070
5-Methylchrysene Mouse skin Increased DNA adducts     1643254
BD Mouse Suppressed cytotoxic T cell generation     3787617
2401263
Benzene Human and mouse inhalation Decreased proliferation of B/T cells
Suppressed antibody production by of B cells
Oxidative stress imbalance
Decreased p53 expression
    2941900
29883905
9129168
Naphthalene Macrophages Increased lipid peroxidation
Increased cytochrome c reduction and oxidative stress
+   9667488
Styrene Mouse DNA damage in lymphocytes, liver, bone marrow, and kidney     9783323
Human Reduced proportion of T helper cells
Increased T suppressor cells
Increased NK cells
    1630405
Human Increased expression of adhesion molecules on lymphocytes, monocytes, and granulocytes +   12141393
N-Nitrosodimethylamine Mouse Suppressed cell-mediated immunity and humoral immunity *   1433375
6716271
3156199
PMNs Increased expression of iNOS, phospho-PI3K, phospho-IκBα and NF-κB, phospho-Akt (T308), phospho-Akt (S473), and phospho-IKKαβ, c-Jun and FosB
Increased apoptosis
  PI3K-Akt/PKB pathway 30722700
23971717
N-Nitrosodiethanolamine Lymphocytes Increased mutagenicity and genotoxicity     2808483
3204103
Catechol Human PBMCs Inhibited production of IL-2 and IL-1β *   10932071
NNN Rat oral and esophageal mucosa and keratinocytes Alteration of immune regulation genes (Aire, Ctla4, and CD80) and inflammation (Ephx2 and Inpp5d)     26785143
NNN Rat oral and esophageal mucosa and keratinocytes Alteration of immune regulation genes (Aire, Ctla4, and CD80) and inflammation (Ephx2 and Inpp5d)     26785143
NNK CTLs Increased expression of adhesion molecule CD62L
Impaired expansion capacity of CTLs
Reduced memory programming
    23673295
Epithelial cells/macrophages Inhibited secretion of IL-8, IL-6, MCP-1, and TNF *   15762874
17096151
14764458
Macrophages Increased release of soluble TNF
Decreased IL-10 synthesis
+   11258792
o-Anisidine Hepatocytes/macrophages Increased proinflammatory cytokines
Induced ROS generation
+ eIF2- and Nrf2-mediated oxidative stress response 28089782
Trp-P-1 DCs Increased expression of costimulatory receptors (CD80, CD86, and MHC)
Inhibited DC maturation
Inhibited IL-12 and TNF-α production
Attenuated T cell proliferation/activation induced by DCs
+ signaling pathways mediated through p38 kinase 21078543
Macrophages Inhibited IL-8 expression
Inhibited phosphorylation of p38 MAP kinase
* Intracellular calcium/p38 MAP kinase-dependent 18281166
Benzo[b]furan Cancer cells Increased expression of proapoptotic genes (TNFRSF 10A, TNFRSF 10B, CASP8, BAX, BID, NOXA, APAF1)
Activated JNK and p38 kinase
  PI3K/Akt/mTOR signaling and mitochondrial-mediated apoptosis 30465514
27149364
FA Splenocytes Increased T cell differentiation into regulatory T cells
Suppressed Th1-, Th2-, and Th17-related splenic cytokines
Suppressed effector T cell activity and decreased T cell-related cytokines
* Calcineurin-NFAT signaling 33046725
Human exposure Increased CD19+ B cells and CD56+ NK cells
Increased IL-10 and IL-4
Decreased IL-8 and IFN-γ
*   25157974
Acetaldehyde Splenocytes Decreased cytokine production
Inhibited aerobic glycolysis-related signal in T cells
Inhibited NK activity and CTL-mediated lysis
*   30121625
2690659
Human exposure Degranulation of human mast cells and histamine release
Induced GMCSF production and NF-κB activation
Increased allergic inflammation
+   17590989
PMN/monocytes Increased ROS production
Decreased PMN phagocytic functions
+    
PBMC Inhibited T cell and B cell proliferation
Decreased release of IFN-γ
    8953156
Cadmium Macrophages
Mast cells
Increased ROS production
Increased TNF-α and nitrite production
Decreased mast cell TNF-α and IgE-mediated histamine release
+   30828855
Peripheral blood neutrophils and splenocytes Increased expression of cytochrome P450s enzymes
Increased ROS production
Induced autophagy and apoptosis in splenocytes
  miR-216a-PI3K/AKT 32058096 30985881 32563067
Cobalt HMEC-1
Macrophages
Increased inflammatory markers such as IL-6, IL-8, ICAM-1, and sICAM-1 + TLR4 27835611
Murine macrophages Increased oxidative stress and ROS production
Decreased OCR and induced mitochondrial dysfunction
    30144138
Lead (inorganic) Macrophages Increased cell death
Increased the antioxidant enzymatic activity of catalase
Decreased macrophage phagocytic index, nitric oxide production, endosomal/lysosomal system stability
    16757190
17959351
N-nitrosopiperidine Esophageal epithelium Increased cell death     16816872

Twenty-eight out of 72 carcinogens in cigarette smoke (PMID: 21324834) have been characterized to affect the activity and function of different innate and adaptive immune cells.

AhR, aryl hydrocarbon receptor; B[k]F, benzo[k]fluoranthene; BaP, benzo[a]pyrene; BD, 1,3-butadiene; BMDMs, bone marrow-derived macrophages; CTLs, CD8+ cytotoxic T lymphocytes; CYP1, cytochrome P450 family 1; FA, formaldehyde; GMCSF, granulocyte/macrophage-colony-stimulating factor; HMEC-1, human microvascular endothelial cell 1; HPBECs, human primary bronchial epithelial cells; ICAM-1, intercellular adhesion molecule 1; IFN-γ, interferon-gamma; JNK, c-Jun N-terminal kinase; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor kappa B; NK, natural killer; NNK, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone; NNN, N′-nitrosonornicotine; NO, nitric oxide; Nrf2, nuclear factor erythroid 2-related factor 2; OCR, oxygen consumption rate; PBMCs, peripheral blood mononuclear cells; PHA, phytohemagglutinin; PI3-AKT, phosphoinositide-3-kinase–protein kinase B; PMNs, polymorphonuclear neutrophils; sICAM-1, soluble intercellular adhesion molecule 1; Th, T helper cell; TLR, toll-like receptor; VEGF, vascular endothelial growth factor.

Trained immunity is a relatively new concept where signaling from DAMP receptors can modulate immunity. TLR agonists have been shown to trigger trained immunity through metabolic reprogramming and epigenetic modifications, which drive profound augmentation of antimicrobial functions (Owen et al, 2020). In addition, inflammasome NLRP3 was reported to mediate trained immunity following the use of Western diet (high-fat diet) and could thereby mediate the potentially deleterious effects of trained immunity in inflammatory diseases. Taken together, although we do not know whether signals triggered by all the DAMPs/PAMPs modulate trained immunity, CS activates signaling of TLRs and inflammasomes, which could lead to the establishment of chronic inflammation via trained immunity (Christ et al, 2018).

Cigarette Smoke Reprograms Immunometabolism and Trained Immunity-Related Metabolic Pathways in Innate Immune Cells and T Cells, Which Can Be Sensed by the Cytoplasmic (Cytosolic and Non-Nucleus Organelles) ROS System

CS influences both innate and adaptive immunity and plays dual functions in regulating immunity by promoting antipathogen immune responses or suppressing defensive immunity. Noncarcinogenic DAMPs from CS activate both innate and adaptive immune cells including macrophages, neutrophils, dendritic cells (DCs), natural killer (NK) cells, B cells, CD4+ T helper cells (T helper cell 1, Th1) (Th1/Th2/Th17), CD4+Foxp3+ Tregs, CD8+ T cells, and memory T/B lymphocytes (Table 3).

Table 3.

Noncarcinogenic Danger-Associated Molecular Patterns from Cigarette Smoke Activate Both Innate and Adaptive Immune Cells

Cell type Effects PMID
Macrophages Increased cell activation
Increased expression of TLR4
Increased secretion of proinflammatory mediators (TNF-α)
Increased ROS production
Impaired bactericidal and phagocytotic processes
Activation of IL-1R-associated kinase, p38, and NF-κB
12033743
17630319
16004610
16620395
19409098
17947684
Neutrophils Increased cell number
Increased IL-8 production
Increased ROS production
Enhanced degradation of IκB-α/β proteins and activity of p65 and p50
6556892
12960242
DCs Increased number of immature DCs and decreased number of mature DCs
Decreased T cell stimulatory capacity and Th1-cell polarization
Increased expression of MHC-II molecules and costimulatory molecules CD40 and CD86
Increased CD4+ cells rather than CD8+ cells
18337593
25338516
16055867
NK cells Decreased NK cell numbers and activity
Decreased cytotoxic activity and cytokine production
Decreased IFN-γ, TNF-α secretions, and perforin expression
26201093
18055568
B lymphocytes Increased levels of memory B cells and memory IgG+ B cells
Increased levels of circulating IgE
Decreased IgG, IgM, and IgA
Increased expression of nicotinic receptors
19909533
24502245
14500745
25011477
T cells Increased lung CD3+, CD8+, and CD4+ T cells
Increased the percentage of CD8+ NKG2D+ cells
Enhanced percentage of CD8+ CD69+ cells and cell surface expression of CD69
Increased IFN-γ and CCR6 expression
Increased Th17 cells and Th17-related cytokines (IL-17A, IL-6, and IL-23)
Increased percentages of Treg cells
Increased activation of cytotoxic CD8+ T cells
Increased production of IL-1β, IL-6, IL-10, IL-12p70, TNF-α, and IFN-γ
Decreased CD4+ CD25+ Treg cells, Foxp3, TGF-β, and IL-10
18706444
21763119
20863413
20646637
23044435
23319833
28745532
22070100

CS affects both innate and adaptive immunity and plays dual functions in regulating immunity by promoting pathogenic immune responses or suppressing defensive immunity. Innate immune cells affected by CS include macrophages, neutrophils, DCs, and NK cells, while adaptive immune cells affected by CS mainly include T helper cells (Th1/Th2/Th17), CD4+CD25+ regulatory T cells, CD8+ T cells, B cells, and memory T/B lymphocytes.

Foxp3, forkhead box P3; TGF-β, transforming growth factor-β.

Innate immune cells can develop exacerbated long-term immune responses and inflammatory phenotype following brief exposure to endogenous or exogenous DAMPs, which results in a primed and significantly enhanced inflammatory response toward a second challenge after the return to a nonactivated state. This phenomenon is known as innate immune memory or trained immunity (Drummer et al, 2021a; Lu et al, 2019; Zhong et al, 2020). As shown in Figure 1, the establishment of trained immunity involves metabolic reprogramming and epigenetic modification of the innate immune cells, allowing qualitatively and quantitatively adjusted responses of innate immune cells to subsequent time-delayed heterologous stimulations (Netea et al, 2020; Netea et al, 2019; Netea et al, 2016).

FIG. 1.

FIG. 1.

The concept of trained immunity and trained tolerance. Trained immunity involves metabolic reprogramming and epigenetic modification of the innate immune cells, allowing qualitatively and quantitatively adjusted responses of innate immune cells to subsequent time-delayed heterologous stimulation. Inappropriate trained immunity response can contribute to disease progression, resulting in either a chronic hyperinflammatory state or a persistent state of immunological tolerance.

Trained immunity is not only important for host defense and vaccine responses but also for promotion of the pathogenesis of chronic inflammation including metabolic CVDs such as atherosclerosis (Shao et al, 2021a) and synergies among the disease risk factors (Drummer et al, 2021b; Fagenson et al, 2020).

In contrast to the memory function in adaptive immune system with specialized cell subsets to carry out memory function (i.e., memory T cells and memory B cells) (Shen et al, 2019), trained immunity can occur in innate immune cells, including monocytes/macrophages (Lai et al, 2019), DCs (Netea et al, 2020), NK cells, aortic cells (Lu et al, 2022), innate immune functions of T cells, and Treg cells (Lee et al, 2020; Ni et al, 2021; Seyda et al, 2016; Zhang et al, 2020b), and nontraditional immune cells, such as ECs (Lu et al, 2022; Lu et al, 2019; Mai et al, 2013; Shao et al, 2020), VSMCs (Flores-Gomez et al, 2021; Lu et al, 2022; Schnack et al, 2019), fibroblasts (Drummer et al, 2021a), and hepatocytes (Drummer et al, 2021b; Fagenson et al, 2020). Furthermore, after the second challenge, these innate cells can alternatively respond less strongly than to the primary response, and the anti-inflammatory mechanisms are promoted resulting in a state of innate immunological tolerance to prevent tissue damage and limit the inflammatory response (Netea et al, 2020).

It has been reported that T cells play important roles in innate immunity and in antigen nonspecific protection (Berg and Forman, 2006). On the contrary, as adaptive immune cells, T cells are regulated by cytokines and cell–cell communication signals of the innate immune system (Kwesi-Maliepaard et al, 2021; Stäger and Kaye, 2004). Along the same line, we recently reported that CD4+Foxp3+ Treg cells have many active innate immune pathways (Ni et al, 2021; Zhang et al, 2020b), and Tregs can sustain their immunosuppressive functions (Shao et al, 2021b) although Treg cell plasticity in atherosclerosis with a chronic inflammatory environment has been reported (Shao et al, 2021b; Xu et al, 2018).

Our reports indicate that Treg cells and other adaptive immune cells not only respond to antigen stimulation (Yan et al, 2008) but also respond to the stimulation by DAMPs/PAMPs similar to that of innate immune cells (Ke et al, 2008; Lopez-Pastrana et al, 2015; Ni et al, 2021; Yang et al, 2015; Yin et al, 2013). Evidence supporting this new model is that DAMPs/PAMPs sensor inflammasome-dependent release of cytokines, and antigen can activate, shape, or even inhibit adaptive immune responses (Deets and Vance, 2021). TLRs, cytokine receptors, and T cell and B cell antigen receptors have been shown to activate trained immunity-related metabolic pathways, which include phosphoinositide-3-kinase–protein kinase B, mammalian target of rapamycin (Bekkering et al, 2021), liver kinase B1 (also known as serine/threonine kinase 11) (Timilshina et al, 2019), and AMP-activated protein kinase signaling pathways (Cheng et al, 2014; Chou et al, 2022).

CS constituents including nicotine can bind to the intracellular and outer membrane receptors and induce metabolic reprograming and increase disease risk (Fig. 2). CS exposure in several disease conditions, which would include COPD (Pauwels et al, 2011), hyperlipidemia (high-fat diet feeding) (Wu et al, 2018), and CHD (Mao et al, 2021), enhances the innate immune response and promotes disease progression (Table 4). It has been reported that increased energy metabolism and the electron transport chain (Yin and O'Neill, 2021) via (i) glycolysis, (ii) acetyl-CoA generation, (iii) mevalonate synthesis (part of cholesterol biosynthesis), (iv) glutaminolysis (converting glutamine into tricarboxylic acid [TCA] cycle metabolites through the activity of multiple enzymes), and (v) epigenetic modification (Ferreira et al, 2022; Lu et al, 2019) contribute significantly to the establishment of trained immunity. Trained immunity can contribute to disease progression, resulting in a chronic hyperinflammatory state (Shao et al, 2021a).

FIG. 2.

FIG. 2.

Several DAMP receptor pathways induce innate immune memory (trained immunity) and sustained inflammation, tissue remodeling, and damage. CS promotes disease risk factors by binding to its receptors and induces bioenergetic metabolic reprogramming to induce trained immunity pathology. CS, cigarette smoke; DAMP, danger-associated molecular pattern.

Table 4.

Cigarette Smoke Exacerbates Inflammatory and Immune Responses

Smoke component Cell type/tissue/disease condition Effect Receptor/pathway PMID
CS-exposed patients Lung tissue of COPD patients Increased expression of IL-1α and IL-1β
Promoted inflammation and activated NLRP3-related pyroptosis pathway
IL-1α and Nlrp3/caspase-1/IL-1β 21622588
Nicotine HFD-fed ApoE−/− mice Increased inflammatory cytokine secretion
Increased cleavage of caspase-1, IL-1β, and IL-18 secretion
Increased atherosclerotic plaques
Increased activation of NLRP3-ASC inflammasome
Increased pyroptosis in HAECs
ROS-NLRP3-mediated EC pyroptosis 29416034
Nicotine BMDM/ECs from CHD patients Increased plasma IL-1β and IL-18
Increased TXNIP expression
Increased mitochondrial ROS production
Increased activation of NLRP3 and caspase-1 inflammasome
Increased GSDMD expression
Increased monocyte/macrophage dysfunction
TXNIP/NLRP3-mediated pyroptotic pathway 33626512
Nicotine Macrophages from HFD-fed ApoE/ mice Increased expression of cleaved caspase1, NLRP3, IL-1β, IL-18
Increased LDH release
Induced pyroptosis
HDAC6/NF-κB/NLRP3 signaling pathway 33321327
Smoke exposure Pseudomonas aeruginosa infected mouse lung Delayed rate of bacterial clearance
Increased neutrophils and mononuclear cells infiltration
Increased proinflammatory cytokines and chemokines
  15317669
CS Angiotensin-II induced hypertension in mice Induced mitochondrial oxidative stress and endothelial dysfunction
Induced oxidation of cardiolipin (mitochondrial oxidative stress biomarker)
Increased blood pressure
  30608177
Chronic nicotine exposure Renal IRI in mice Exacerbated renal IRI and oxidative stress-induced injury
Aggravate acute renal ischemia- or oxidative stress-induced stress kinase signaling
  21511693
23892062
22552933
CSE Human bronchial epithelial cell Increased expression of TLR4
Increased binding of LPS
Increased release of IL-8 by LPS stimulated cells
Increased neutrophil chemotaxis
Amplification of inflammation
Activation of ERK pathway 18217953
CS Mice with poly (I:C) administration Increased infiltration of macrophages, neutrophils, and eosinophils in the bronchoalveolar lavage
Induced inflammatory response
  28468623
CS NTHI Exacerbated NTHI-mediated chronic respiratory inflammation
Increased proinflammatory IL-1β, IL-6, and TNF-α
Decreased IFN-γ and IL-4 secretion
  24752444

CSE, cigarette smoke extract; GSDMD, gasdermin D; HAECs, human aortic endothelial cells; HDAC6, histone deacetylase 6; HFD, high-fat diet; IRI, ischemia–reperfusion injury; LDH, lactate-dehydrogenase; LPS, lipopolysaccharide; NLRP3, NOD-, LRR- and pyrin domain-containing protein 3; NTHI, nontypeable Haemophilus influenzae; TXNIP, thioredoxin-interacting protein.

We previously reported that the anti-inflammatory cytokine IL-35 is an inflammation-induced cytokine (Li et al, 2018; Li et al, 2012), in contrast to housekeeping anti-inflammatory cytokines such as transforming growth factor-β (Li et al, 2012). We reported that anti-inflammatory cytokines IL-35 and IL-10 can inhibit EC activation and vascular inflammation but spare the modulation of trained immunity gene expression (Li et al, 2020). Similarly, concomitant to a proinflammatory response, anti-inflammatory mechanisms such as trained innate immune tolerance, with a feature of upregulated immune responsive gene 1 (IRG1) itaconate synthesis enzyme (Domínguez-Andrés et al, 2019), and homeostasis molecular patterns (Wang et al, 2016b) are induced to prevent excessive inflammation and tissue damage and to limit the inflammatory response in time.

Overall, CS has the capacity to induce opposing effects on the immune system, with chronic exposure increasing inflammation on the one hand, and potentially reducing inflammation by trained immune tolerance on the other (Table 5).

Table 5.

Cigarette Smoke Can Induce Trained Innate Immune Tolerance

  Cell type/disease Effect Receptor/pathway PMID
CSE MDMs and THP-1 cells Inhibited LPS- and LPS plus ATP-induced IL-1β and IL-18 release and pro-IL-1β, pro-IL-18, and NLRP3 expression
Decreased LPS-induced lactate release and basal glycolytic flux and impaired glycolytic burst
NLRP3-independent and TLR4-TRIF-caspase-8-dependent pathway 31914643
A single injection of nicotine Renal I/R injury in rats Attenuated renal dysfunction and tubular necrosis induced by I/R injury
Ameliorated acute tubular damage following renal I/R injury
Reduced TNF-α protein expression and leukocyte infiltration of the kidney
α7nAChR 18401335
A single injection of nicotine Renal I/R injury in rats Reduced the α7nAChR protein after I/R injury
Inhibited renal I/R-induced STAT3 activation
α7nAChR 18614620
Nicotine (500 μM) LPS-stimulated primary human macrophages Inhibited TNF release from LPS-stimulated macrophages   12508119
Nicotine IL-18-stimulated PBMC Inhibited IL-18-enhanced expression of ICAM-1 and CD40
Inhibited production of IL-12, TNF-α, and IFN-γ by IL-18-stimulated cells
α7nAChR 16966384
Pretreatment with CSE LPS stimulated bronchial epithelial cells Inhibited LPS-induced GM-CSF and IL-8 protein release
Suppressed neutrophil chemotaxis induced by LPS
Downregulated the activity of LPS-induced transcription factor AP-1
Attenuated inflammatory response induced by LPS
Suppression of AP-1 activation 15356167
Pretreatment with CSE Anti-CD3 and PMA stimulated PBMCs Suppressed production of IL-1β, TNF-α, IL-2, and IFN-γ   10932071
CS Lung macrophages of Apoe/ mice Increased itaconate metabolite
Increased expression of IRG1
Prevents overshooting macrophage activation in the lungs
May counteract the oxidative challenge from the activated immune system as well as directly from CS exposure.
  32419906

CS increases the expression of anti-inflammatory cytokines and decreases the expression of proinflammatory cytokines and chemokines.

α7nAChR, α7 nicotinic acetylcholine receptor; IRG1, immune responsive gene 1; MDMs, monocyte-derived macrophages; PMA, phorbol myristate acetate; STAT3, signal transducer and activator of transcription 3.

Trained innate immune tolerance leads to a persistent state of innate immunological tolerance, a new mechanism that dampens the inflammatory response of the host to maintain homeostasis and prevent tissue damage and organ failure, but with the subsequent risk of secondary infections and other diseases related to decreased activity of the immune system (Netea et al, 2020). Itaconate accumulation has been shown to inhibit the expression of proinflammatory mediators such as IL-6, IL-1β, and IL-12p70 in lipopolysaccharide (LPS)-stimulated macrophages (Lampropoulou et al, 2016).

As shown in Figure 3, itaconate is upregulated in macrophages activated by LPS via increased cis-aconitate decarboxylase/IRG1 transcription. Overproduction of itaconate activates the antioxidant transcription factor nuclear factor erythroid 2–related factor 2 (Nrf2) pathway by alkylation of Kelch-like ECH-associated protein 1, which induces the transcription of various Nrf2-dependent antioxidant and anti-inflammatory genes. This would include heme oxygenase-1, glutamate cysteine synthase, and glutathione synthetase (for glutathione biosynthesis). Itaconate can also inhibit succinate dehydrogenase and reduce ROS generation and IL-1β secretion. Itaconate promotes the transcription of activating transcription factor 3, which directly inhibits the inhibitor of nuclear factor kappa B zeta (IκBζ) expression and leads to decreased expression of IL-6. In addition, itaconate directly alkylates the cysteine residues of glyceraldehyde-3-phosphate dehydrogenase, aldolase, fructose-bisphosphate A (leading to reduced glycolysis), and downregulates IL-1β secretion, thereby depressing the inflammatory response (O'Neill and Artyomov, 2019).

FIG. 3.

FIG. 3.

The concept of trained immune tolerance. Upon LPS stimulation, itaconate production is increased by CAD/IRG1 transcription. Overproduction of itaconate activates the antioxidant transcription factor Nrf2 by alkylation Keap1, which induces the transcription of various Nrf2-dependent antioxidant and anti-inflammatory genes. Itaconate can also inhibit SDH and reduce ROS generation and IL-1β secretion. Itaconate promotes the transcription of ATF3, which directly inhibits the IκBζ expression to reduce IL-6 secretion. In addition, itaconate directly alkylates the cysteine residue 22 of GAPDH and ALDOA to inhibit glycolysis, and reduces IL-1β secretion, thereby alleviating the inflammatory response. CS exposure can significantly increase the expression of IRG1 and the abundance of itaconate metabolites. ALDOA, aldolase, fructose-bisphosphate A; ATF3, activating transcription factor 3; CAD, cis-aconitate decarboxylase; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; IκBζ, inhibitor of nuclear factor-kappa B zeta; IL, interleukin; IRG1, immune responsive gene 1; LPS, lipopolysaccharide; Keap1, kelch-like ECH-associated protein 1; Nrf2, nuclear factor erythroid 2-related factor 2; ROS, reactive oxygen species; SDH, succinate dehydrogenase.

CS can increase the anti-inflammatory gene expression and decreases the expression of proinflammatory cytokines and chemokines, leading to an immunosuppressive effect. CS exposure significantly increases the abundance of itaconate metabolites and the expression of IRG1 (Table 5) (Titz et al, 2020).

Recent progress on trained innate immune tolerance may lead to the development of a new anti-inflammatory therapy such as ItaCORMs (Krause et al, 2021). Although it is unclear whether CS promotes functionally trained immunity or trained immune tolerance, CS has divergent effects on innate immunity in promoting inflammatory bowel disease but inhibiting ulcerative colitis (AlQasrawi et al, 2020). These findings demonstrate that innate immune cells and adaptive immune cells are equipped with innate immune signaling pathways, presumably including both innate immune memory (trained immunity) and innate immune tolerance, in response to stimulation by various DAMPs, PAMPs, and other environmental stimuli derived from CS.

Based on extensive analysis, we recently proposed that ROS systems are a new integrated network for sensing homeostasis and alarming stress in organelle metabolic processes (Sun et al, 2020). In addition to critical functions in signaling (Li et al, 2016a; Li et al, 2013) and damaging DNAs (nuclear ROS), ROS contribute to a large number of human diseases (Hybertson et al, 2011) and aging (Kowalska et al, 2020). Thus, cytoplasmic ROS and reactive nitric species (RNS) (Michel and Vanhoutte, 2010) can be used as the indicators of metabolic reprogramming. CS contributes to metabolic dysregulation and a shift of metabolism in innate immune cells (Table 6). CS inhibits alveolar macrophage mitochondrial respiration while inducing glycolysis and ROS (Aridgides et al, 2019).

Table 6.

Metabolic Reprogramming Induced by Cigarette Smoke May Be the Novel Mechanisms Underlying the Establishment of Trained Innate Immunity and Trained Tolerance

Smoke component Cell type/tissue/species Effect PMID
Chronic CS Lung cells Increased expression of TCA cycle enzymes
Increased expression of enzymes involved in glutamine metabolism, fatty acid degradation, and lactate synthesis
Induced mitochondrial metabolic reprogramming
29042306
CS and electronic cigarette vaping   Increased sphingolipid metabolites
Decreased TCA cycle metabolites
34072305
Short-term CS Alveolar type-II cells Increased glycolysis
Increased mitochondria β-oxidation
Decreases levels of phosphatidylcholine
24625219
Short-term CS Mouse lung (mitochondria) Increased pentose phosphate pathway
Increased expression and activity of complexes II, III, IV, and V
Upregulation of genes involved in glycolysis, TCA cycle, mitochondrial fatty acid oxidation, and redox regulation
23064950
CSE Pulmonary microvascular ECs Increased glycolysis
Decreased mitochondrial respiration
Decreased fatty acid oxidation
31555131
CS Alveolar macrophages Increased glycolysis
Decreased mitochondrial respiration
Increased ROS and metabolic dysfunction
31270372
Chewing tobacco Epithelial cells Increased oxidative phosphorylation
Decreased expression of enzymes involved in the glycolytic pathway
Increased expression of mitochondrial proteins involved in the electron transport chain and enzymes of the TCA cycle
31438645
CS Human lymphocytes Decreased mitochondrial complex IV activity
Increased lipid peroxidation
10383908
CSE Monocyte-derived macrophages and THP-1 cells Decreased basal glycolytic flux
Impaired glycolytic burst in response to LPS
31914643

CS induces metabolic reprogramming depending on many variables, including the type of tobacco and duration of use, and type of exposed cells.

TCA, tricarboxylic acid.

Furthermore, short-term (4 and 8 weeks) exposure of A/J mice to CS (a model for lung tumorigenesis caused by TS) (Witschi, 2005) induces upregulation of genes encoding glycolysis, TCA cycle, mitochondrial fatty acid oxidation pathway, and redox regulation (Agarwal et al, 2014). Increased expression of nuclear genes in the glycolytic pathway and decreased levels of mitochondrial genes following exposure to either the main stream extract or side stream smoke extract support the notion that CS significantly contributes to the transformation of nonmalignant esophageal epithelial cells into a tumorigenic phenotype via metabolic reprogramming (Kim et al, 2010). In addition, acute CS exposure leads to a reversible airspace enlargement in A/J mice, indicative of alveolar damage. A decrease in respiration rates while metabolizing glucose is observed in the CS-exposed group, indicating altered glycolysis that is compensated by an increase in fatty acid palmitate utilization.

Fatty acid palmitate utilization is accompanied by an increase in the expression of CD36 and carnitine palmitoyl transferase 1 in type II alveolar cells for the transport of palmitate into the cells and into mitochondria, respectively (Agarwal et al, 2014). As we and others reported, fatty acid oxidation is also essential for inflammasome activation in proinflammatory type 1 macrophages (M1) (Batista-Gonzalez et al, 2019; Lai et al, 2019).

Epigenetic alteration, including DNA methylation (Jamaluddin et al, 2007) and histone posttranslational modifications (Shao et al, 2016), has been shown to play a significant role in the progression of CS-related diseases (Table 7). DNA methylation is catalyzed by a protein family known as DNA methyltransferases (DNMTs), which transfer methyl groups from S-adenosyl-l-methionine to the 5-carbon position of cytosine residues in DNA. Several studies demonstrate the associations between smoking and altered DNA methylation at multiple cytosine-phosphateguanine (CpG) sites (Breitling et al, 2011; Shenker et al, 2013; Wan et al, 2012). Some DNA methylation sites associated with tobacco smoking have also been localized to genes related to CHD (Breitling et al, 2012) and pulmonary disease (Wauters et al, 2015). Some studies have found different DNA methylation associated CpGs in smokers versus nonsmokers (Zeilinger et al, 2013). CS increases DNA methylation and inflammation in macrophages (Yu et al, 2016) and CS-exposed individuals (Siedlinski et al, 2012). DNMTs are significantly overexpressed in the lung tissues of the smokers compared with the nonsmokers (Kwon et al, 2007). Histone acetylation, methylation, phosphorylation, and ubiquitination are the most broadly studied and extensively characterized histone posttranslational modifications in terms of the regulation of chromatin structure and transcriptional activity (Li et al, 2018; Shao et al, 2016).

Table 7.

Cigarette Smoke Induces Epigenetic Innate Immune Memory in the Form of Histone Acetylation and Methylation and Provides a Foundation for Sustained Inflammation, in Which Acetyl and Methyl Donations Are Generated by CS-Induced Metabolic Reprogramming

HATs/HDACs Smoking Tissues/cells/species Changes by CS PMID
I: DNA methylation
 DNMT1 CS Macrophages Increased DNMT1 expression
Increased proinflammatory cytokine production
Decreased PPAR-γ
17053888
27530451
 JAK3
 KRT1
 RUNX3
CS CS-exposed individuals Hypermethylation of RUNX3, JAK3, and KRT1 genes associated with CRP in COPD increased inflammation 22617718
II: Histone posttranslational modification: HATs and HDACs
 HAT, CBP/p300 CSE Bronchial epithelial cells Increased expression of HAT, p300/CBP
Increase acetylation of histones (H3/H4) and NF-κB in COPD
Increased inflammation
27925185
19811389
 HDAC1 CSE/CS Rat lung tissue and macrophages Decreased expression of HDAC1
Increased MCP-1, IL-8, TNF-α, and MMP9 expressions increased level of acetylated H3K9,
Increased inflammation
26033389
 HDAC2 CS Lung tissue of COPD smokers Decreased cytoplasmic expression of HDAC2
Increased acetylation of histones H3 and H4
Increased expression of IL-12p40
Decreased IκBα expression
Increased inflammation
30659954
16574938
 HDAC3 CSE Bronchial epithelial cells
Alveolar macrophages
Decreased SIRT1, HDAC2, HDAC3, and FoxO3
Increased TLR4 and survivin, LPS binding, and ERK ½ activation
Increased IL8 and IL1β production
Increased inflammation
30659954
22613758
 HDAC4 Chronic CS Mouse lung and lung tissue of COPD smokers Decreased expression of HDAC4
Repressed c-Jun and IL-17A in COPD
27793800
 HDAC5/8 CS Lung tissue and alveolar macrophages of COPD smokers Decreased HDAC5 expression
Increased IL-8 expression
Increased histone-4 acetylation
15888697
 HDAC6 CSE Mouse epithelial cells and lung microvascular ECs/chronic smokers with COPD Increased HDAC6 expression 24200693
26452072
 HDAC7 CS Human exposure Decreased HDAC7 expression
Increased HIF1α
22172637
 HDAC10 CS Lung tissues of smokers with COPD Decreased HDAC10 expression 30214182
 SIRT1 Smoker with COPD PBMCs Decreased expression and activity of SIRT1 29861836
27167200
 SIRT3 CS WT mice Decreased sirtuin-3 expression
Increased mitochondrial oxidative stress
Induced endothelial dysfunction
30608177
 SIRT4 CS Human pulmonary microvascular ECs Decreased SIRT4 expression
Induced mononuclear cell adhesion
Induced VCAM-1, E-selectin, and NF-κB activation
Increased proinflammatory cytokines IL-1β, TNF-α, and IL-6
24603126
 SIRT5 CSE Lung epithelial cells Increased SIRT5 expression and FOXO3 acetylation 25981116
 SIRT6 CSE Human bronchial epithelial cells Decreased SIRT6 expression
Induced cellular senescence
24367027

CS induces hyperacetylation and increases H3 and H4acetylation by increasing HATs and decreasing HDACs as well as modification of DNA methylation.

CS, cigarette smoke; DNMT, DNA methyltransferase; HATs, histone acetyltransferases; VCAM-1, vascular cell adhesion molecule 1; WT, wild type.

Histone posttranslational modifications play a critical role in gene expression by adding or removing the acetyl or methyl group, which is catalyzed by various enzymes (Bannister and Kouzarides, 2011). Also, lysine residues in histones can be oxidized by the lysyl oxidase family of proteins (Serra-Bardenys and Peiroó, 2021). Furthermore, epigenetic modification in the form of histone acetylation and methylation, which are catalyzed by histone acetyltransferases (HATs), histone demethylases, and histone deacetylases (HDACs), is associated with trained immunity. For example, there are increases inmarkers of open chromatin such as histone 3 lysine 4 trimethylation (H3K4me3), H3K4me1, and histone 3 lysine 27 acetylation (H3K27ac). Simultaneously, there is a decrease in histone markers depicting closed chromatin such as histone 3 lysine 9 dimethylation (H3K9me2). Epigenetic alterations include histone methylation and acetylation that work in concert to regulate gene transcription in a heritable manner.

The enzymes that regulate these epigenetic modifications can be activated by smoking, which further mediates the expression of multiple inflammatory genes (van der Heijden et al, 2018). Finally, CS has been shown to increase histone acetylation by increasing the expression of HATs and decreasing the expression of HDACs (Ito et al, 2005; Sundar et al, 2014).

Transgenerational epigenetic inheritance occurs when developmental programming is transmitted across generations that were not exposed to the initial environment, which triggered the change. The transgenerational programming has been described for metabolic and CVD risk (Krauss-Etschmann et al, 2015). A previous study showed significant hypomethylation in the placentas of babies born to mothers who smoked during pregnancy compared with that of nonsmoking mothers. This hypomethylation was found to correlate with increased placental cytochrome P450 family 1 subfamily A member 1 (CYP1A1) expression, which may have implications for xenobiotic metabolism in the offspring (Suter et al, 2011). Moreover, hypermethylation of the brain-derived neurotrophic factor may be responsible for its decreased expression with subsequent behavioral consequences in infants, children, and adolescents exposed in utero to maternal cigarette smoke (Knopik et al, 2012).

Taken together, these results have demonstrated that CS may modulate trained immunity and trained tolerance pathways via regulating energy metabolisms and epigenetic modifications. Since CS has divergent pro- and anti-inflammatory activities (AlQasrawi et al, 2020), we suggest that CS modulates inflammation and immunity via a linked trained innate immunity and trained innate immune tolerance process.

Five Nucleus-Localized ROS Activate CS-Promoted DNA Damage and Cell Death Pathways to Potentially Amplify Inflammation and Immune Responses via Releasing Alarmins

The toxicity of CS is largely attributed to its ability to generate ROS. CS contains a number of highly unstable free radicals, and these free radicals enhance ROS and RNS production leading to oxidative/nitrosative stress. Increased oxidative/nitrosative stress plays an important role in the pathogenesis of several diseases such as diabetes, lung cancer, atherosclerosis, and COPD. Accumulating evidence suggests a key role for smoke-induced ROS and the resulting oxidative stress in inflammation and cancer. CS is also known to stimulate ROS production by activating ROS sources, such as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondria (Fetterman et al, 2017; Kim et al, 2014; Li et al, 2016a).

Five ROS are localized in the nucleus including superoxide (O2), nitrogen dioxide (NO2), hydroxyl radical (OH), hydrogen peroxide (H2O2), and hypochlorous acid (HOCl) (Lu et al, 2021; Sun et al, 2020), and it has been reported that CS extract produces superoxide by reacting with bicarbonate (Park et al, 2021). Nitrogen oxides (NOx), most notably nitrogen dioxide (NO2), are among the most hazardous forms of air pollution. TS is a major indoor source of NOx. Substantial increases in NOx concentrations are found when smoking only one cigarette (Braun et al, 2021), suggesting that NOx exposure occurs for secondhand smokers following smoking in indoor rooms. In addition, cigarette tar in aqueous solution contains a quinone–hydroquinone–semiquinone complex that can reduce oxygen to produce superoxide and hence hydrogen peroxide and the hydroxyl radical.

The cigarette tar radical can penetrate viable cells, bind to DNA, and induce nicks (Pryor, 1997; Zeng et al, 2018). Moreover, nicotine mediates hypochlorous acid-induced nuclear protein damage in mammalian cells (Salama et al, 2014). Chronic CS exposure results in increased oxidative stress leading to diaphragm muscle dysfunction (Barreiro et al, 2012), and increased expression of pro-oxidant genes and proinflammatory markers (Khanna et al, 2013). CS also increases intracellular ROS, cytokine expression, basal mitochondrial ROS, and oxidative stress in alveolar macrophages (Table 8) (Lugg et al, 2022).

Table 8.

Cigarette Smoke Induces Reactive Oxygen Species and Oxidative Stress, Which Promote Inflammation, Inflammatory Cell Death (Pyroptosis), Oxidative DNA Damage, and Cancers

  Cell type/tissue/species Effect PMID
CS-exposed humans, mice, and pig Skeletal muscles
Diaphragm
Induced oxidative stress and inflammation leading to muscle dysfunction 20413628
22349133
CS-exposed rat Serum/brain Increased expression of pro-oxidant genes (iNOS, NOX4, dual oxidase 1, and p22[phox]) leading to oxidative stress
Increased expression of proinflammatory markers (IFN-γ, TNF-α, IL-1α, IL-1β, IL-23, IL-6, IL-23, IL-17, IL-10, TGF-β, T-bet, and FoxP3) and increased inflammation
23031832
CSE Type II alveolar epithelial cells Decreased cell viability
Decreased expression of VEGF
Increased ROS production and oxidative stress
25607112
CSE Mouse β cells Increased β cell endoplasmic reticulum and oxidative stress
Reduced insulin secretion
Impaired β cell survival
32283079
CS Mouse lung Increased MDA and MPO activities
Decreased SOD activity
Induced oxidative stress
Reduced the expression of the mitochondrial fusion protein OPA1 and fission protein MFF
Induced mitochondrial dysfunction
34221235
Acute CS exposure Human alveolar epithelial cell Increased cellular oxidative stress and autophagy 31016633
Electronic cigarette Rats Induced oxidative stress 34515107
CS exposure Alveolar macrophages Increased intracellular ROS
Increased IL-8 cytokine production
Increased basal mitochondrial ROS
Increased oxidative stress
33986144
Nicotine Mouse VSMCs/Apoe/ mice Induced autophagy and VSMC phenotype switching
Increased ROS production and NF-κB signaling pathway
Increased aortic atherosclerotic plaque
30856513

iNOS, inducible nitric oxide synthase; MDA, malondialdehyde; MFF, mitochondrial fission factor; MPO, myeloperoxidase; OPA1, optic atrophy protein 1; SOD, superoxide dismutase; VSMCs, vascular smooth muscle cells.

Several new cell death pathways (Xiong et al, 2009; Yan et al, 2008; Yang et al, 2005; Yang et al, 2002; Yang et al, 1997) have been recently reported including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition-driven necrosis, necroptosis, ferroptosis, pyroptosis (inflammatory cell death), parthanatos (excessive oxidative damage to DNA leading to overactivation of poly(ADP-ribose), polymerase [PARP]), entotic cell death, neutrophil extracellular trap (NETotic) cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence, mitotic catastrophe (Galluzzi et al, 2018; Shao et al, 2021a; Wang et al, 2019), and alkaliptosis and oxelptosis (Tang et al, 2019).

Among the 16 cell death types, most cell death types are involved in inflammation (Galluzzi et al, 2018; Wang et al, 2019; Yan et al, 2008; Yang et al, 2021). Moreover, a new pathway for proinflammatory programmed cell death, PANoptosis, has been described, which is controlled by a recently identified cytoplasmic multimeric protein complex named the PANoptosome. The PANoptosome can engage, in parallel, three key modes of programmed cell death—pyroptosis, apoptosis, and necroptosis (Samir et al, 2020). The cells undergoing death emit numerous signals that interact with the host to dictate the immunological signature of cellular stress and death. In the absence of reactive (nontolerated) antigenic determinants (which is generally the case for healthy cells), such signals may drive inflammation but cannot engage adaptive immunity.

Conversely, when cells exhibit sufficient reactive nontolerated antigenicity, as in the case of infected or malignant cells with tumor antigens (Yang and Yang, 2005; Yang et al, 2006), their death can culminate with adaptive immune responses that are executed by cytotoxic T lymphocytes and elicit immunological memory (Kroemer et al, 2022; Shen et al, 2019).

In addition to carcinogenic factors of CS exposure, oxidative stress plays significant roles in mediating CS-triggered cell death (Aoshiba and Nagai, 2003). CS exposure induces cellular senescence and seven different types of cell death including apoptosis, necrosis, necroptosis, ferroptosis, pyroptosis (Lopez-Pastrana et al, 2015; Wang et al, 2016a; Yin et al, 2015), parthanatos pathway of cell death, and autophagic cell death (Table 9) (Liu et al, 2021). CS exposure induces mitochondria-to-nuclear translocation of apoptosis-inducing factor and endonuclease G within the first 3 h characteristic of the parthanatos pathway.

Table 9.

Cigarette Smoke Induces Seven Types of Inflammation-Related Cell Death and Senescence

Smoke components Model/cell type Effects PMID
CSE Bronchial epithelial cells Induced cell death via repressing PRMT6/AKT signaling 33260152
CS Bronchial epithelial cells Activated parthanatos pathway of cell death 31396404
CSS Olfactory epithelium Induced apoptotic cell death and TNF expression 29950987
CS Alveolar epithelial cells Enhanced oxidative stress-induced apoptosis and/or necrosis 19570263
TS Human premonocytic Induced apoptosis and necrosis 9755110
CS Human bronchial epithelial cell Induced necroptotic cell death and release of DAMP and proinflammatory cytokines 26719146
CS Human umbilical vein ECs Induced prolonged endoplasmic reticulum stress and autophagic cell death 21676957
CSE Bronchial epithelial cells Reduced cell proliferation
Increased β-galactosidase activity
Increased cellular senescence
27237816
CSE Mice and human bronchial epithelial cells Increased expression of NLRP3
Increased caspase-1 activity and cleaved GSDMD
Increased release of IL-1β and IL-18
Induced pyroptosis
33465393
CSE Human bronchial epithelial cells Induced ferroptosis cell death 31316058
Acute cigarette smoke exposure Normal and COPD human bronchial epithelial cells Induced apoptosis and CXCL8/IL8 production
Induced epithelial to mesenchymal transition
28468623
CSE Human bronchial epithelial cells/bronchoalveolar lavage fluid of CS-exposed mice Induced necroptotic cell death
Induced release of DAMPs
Induced release of proinflammatory cytokines via MyD88 signaling
26719146
CS Bovine pulmonary artery ECs and rat lung microvascular ECs/mice Increased EC apoptosis 24853558
CSE Rat VSMCs Induced ferroptosis and cell death
Increased inflammatory responses
31975626

CS induces seven types of newly characterized cell death pathways in epithelial cells and ECs including apoptosis, necrosis, ferroptosis, pyroptosis, necroptosis, parthanatos pathway cell death, autophagic cell death, and cellular senescence (PMID:29362479; 31355136).

CSS, cigarette smoke solution; CXCL8, C-X-C motif chemokine ligand 8; DAMP, danger-associated molecular pattern; PRMT6, protein arginine methyltransferase 6; TS, tobacco smoke.

These diseases cause oxidative stress in neurons that also produce peroxynitrite (ONOO−, also a nuclear ROS) from the reaction of superoxide anions and nitric oxide causing DNA damage with subsequent PARP-1 activation (Sun et al, 2020). The use of a specific PARP-1 inhibitor, BMN673, abrogates the effect of smoke-induced activation of the parthanatos pathway. Smoke-mediated activation of the parthanatos pathway is increased in human bronchial epithelial cells obtained from chronic smokers (Künzi and Holt, 2019).

Inflammatory cell death can amplify inflammation via at least three pathways: (i) caspase-1-caspase-4 (human)/caspase-11 (mouse) facilitated N-gasdermin D membrane pore to release big proinflammatory secretomes as we previously reported (Lu et al, 2022; Lu et al, 2021; Ni et al, 2021; Xu et al, 2021); (ii) release of alarmins (DAMPs) from dying cells (Yang et al, 2017a); and (iii) NETotic cell death (Delgado-Rizo et al, 2017).

High-mobility group box protein 1 (HMGB1), a highly conserved nonhistone nuclear protein, binds to DNA to regulate the structure of chromosomes and maintain transcription, replication, DNA repair, and nucleosome assembly. HMGB1 is actively or passively released into the extracellular space during cell activation or necrosis. Extracellular HMGB1 as an alarmin can initiate immune responses alone or combined with other substances such as nucleic acid. It has been reported that HMGB1 is involved in various inflammatory responses, the inflammatory-reparative response (Foglio et al, 2022), and autoimmunity (Dong et al, 2022). In addition to HMGB1, alarmins also include a surprising number of chromatin-binding moieties, such as HMGN1, IL-1α, and IL-33, as well as heat shock proteins, S100 proteins, ATP, and uric acid crystals (Yang et al, 2017a). It has been reported that CS can silence innate lymphoid cell function and facilitates an exacerbated type I IL33-dependent inflammatory response to infection (Kearley et al, 2015).

Taken together, five nucleus-localized ROS can activate CS-promoted DNA damage (Zeng et al, 2018) and potentially trigger cell death. CS-induced cell death can amplify inflammation and immune responses. In the future, more detailed work is needed to determine whether combinations of several cell death pathways mediate trained immunity-promoted CS-triggered cell death.

Cigarette Smoke Increases ROS and EC Activation/Dysfunction to Induce Vascular Inflammation and Subsequently Promote CVDs

Vascular endothelium plays a fundamental role in the regulation of vascular tone, inflammatory response, vascular growth, and thrombotic balance by producing important vasodilators with antiatherosclerotic and antiaggregatory properties such as nitric oxide (NO) and prostacyclin (Vane et al, 1990). Endothelium is among the first line of the body's defense system. The normal vascular endothelium is taken as a gatekeeper of cardiovascular health, whereas abnormality of vascular endothelium is a major contributor to CVDs, such as atherosclerosis, aging, hypertension, obesity, and diabetes. Based on the capacities of ECs in carrying out 11 important innate immune functions that are originally considered to be carried out by prototypic innate immune cell-type macrophages, in 2013, we proposed a novel concept that ECs are innate immune cells (Drummer et al, 2021a; Mai et al, 2013; Shao et al, 2021a; Shao et al, 2020).

To consolidate this new concept, we reported that ECs are equipped with innate immune sensing function including canonical inflammasome and caspase-1 (Li et al, 2017; Li et al, 2016b; Lopez-Pastrana et al, 2015; Lu et al, 2021; Wang et al, 2016a; Yin et al, 2015), immune checkpoint receptor upregulation in response to proinflammatory cytokine stimulation (Shen et al, 2019), upregulation of six types of canonical and noncanonical secretomes (Lu et al, 2022), upregulation of a list of new CDs (clusters of differentiation) (Xu et al, 2021), and upregulation of innate immune regulatome and myelopoiesis factor (Shao et al, 2021a).

The five cardinal signs of inflammation are redness, swelling, heat, pain, and loss of function. ECs play a major role in the initiation of inflammatory process. Endothelial activation encompasses a range of endothelial responses to inflammatory signals including changes in thromboresistance, altered vasomotor tone, and loss of barrier function. Once activated, the EC facilitates cellular trafficking. Leukocyte activation and transmigration are important for innate and adaptive immunity. Endothelial dysfunction is widely used to describe the nonphysiologic activity of ECs. The pathophysiology of endothelial dysfunction is complex and multifactorial (Shao et al, 2014). These include imbalanced vasodilation and vasoconstriction, decreased bioavailability of NO, reduced activity of endothelial nitric oxide synthase (eNOS), and increased production of ROS, increased proinflammatory factors. The resulting oxidative stress caused by these factors within the vascular wall contributes significantly to the pathophysiology of endothelial dysfunction and, subsequently, vascular inflammation and CVDs (Shao et al, 2014).

Endothelial dysfunction is a central early event in the pathogenesis of most CVDs and provides an important link between diseases such as hypertension, chronic renal failure, and diabetes. Previous reports showed that inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interact with ECs or VSMCs to induce endothelial nitric oxide dysfunction, ROS production, and VSMC proliferation, resulting in endothelial dysfunction and promotion of CVD. Indications regarding endothelial function are mainly assessed using flow-mediated dilation (FMD). FMD is a noninvasive measurement of brachial artery diameter changes following an increase in shear stress induced by reactive hyperemia (Corretti et al, 2002). Brachial FMD showed a significant predictive value for cardiovascular events (Ras et al, 2013). CS is the most important modifiable risk factor and plays a critical role in the pathogenesis and development of atherosclerotic CVDs by, at least in part, endothelial dysfunction.

CS causes increased oxidative stress by several mechanisms, including (i) direct damage by radical species and (ii) the inflammatory response caused by CS. CS-induced oxidative stress and ROS induce expression of cell adhesion molecules to promote leukocytes—EC adhesion and VSMC proliferation (Table 10) (Teasdale et al, 2014). CS, e-cigarette, and nicotine administration have all been shown to reduce NO bioavailability, which is the central mechanism in the pathophysiology of endothelial dysfunction (DiGiacomo et al, 2018; He et al, 2017).

Table 10.

Cigarette Smoke Induces Endothelial Cell Activation/Dysfunction and Vascular Inflammation via Increasing Reactive Oxygen Species Production, Activating Nuclear Factor Kappa B, and Upregulating Adhesion Molecules

Smoke components Model/cell type Effects PMID
CSE Mouse cerebral microvascular ECs Decreased cell viability
Increased paracellular permeability
Increased proinflammatory cytokines
Increased ROS production
31666540
CSE Pulmonary ECs Increased endothelial monolayer permeability
Induced EC activation and promoted lung inflammation
29351435
CSE HMVEC-L Increased PAF which promotes transendothelial migration
Increased cell surface expression of adhesion molecules such as P-selectin, E-selectin, ICAM-1, and VCAM-1
Increased polymorphonuclear leukocytes adherence
21984569
CSE Carotid arteries of rats Impaired vascular relaxations and induced endothelial dysfunction
Increased ROS production
Increased expression of adhesion molecules (ICAM-1, VCAM-1, and E-selectin) and monocyte adhesion.
Increased expression of inflammatory markers (iNOS, TNF-α, IL-1β, and IL-6)
Induced activation of NF-κB
17213480
CSC HUVEC Increased expression of ICAM-1 and VCAM-1
Induced activation of NF-κB
Increased transendothelial migration of monocytes and neutrophils
Induced activation of protein kinase C
8928867
21651795
CSE HLMVECs/mouse lung tissue cells Increased expression of CXCR3
Increased responsiveness to EMAP II and IP-10 to induce apoptosis
22936405
CS and e-cigarette Male C57BL/6 mice Increases superoxide radical and NADPH oxidase in mouse aorta
Decreases eNOS, p-eNOS Ser1177, and BH4 expression in aorta
Impaired ACh-induced endothelium-dependent relaxation in mouse aortic segments
32412791
35089811
CSE HAECs Decrease in eNOS expression
Increase in expression of iNOS, NLRP3, caspase-1p20, and IL-1β
34768128
CS and e-cigarette Healthy smokers and nonsmoker adults Increased soluble NOX2-derived peptide and oxidative stress
Decrease in NO bioavailability and FMD
27108682

Ach, acetylcholine; CSC, cigarette smoke condensate; CXCR3, C-X-C motif chemokine receptor 3; EMAP II, endothelial monocyte-activating polypeptide II; eNOS, endothelial nitric oxide synthase; FMD, flow-mediated dilation; HLMVECs, human lung microvascular endothelial cells; HuAoSMCs, human aorta primary smooth muscle cells; HUVEC, human umbilical vein endothelial cells; IP-10, interferon-inducible protein–10; NADPH, nicotinamide adenine dinucleotide phosphate; PAF, platelet-activating factor.

eNOS is an enzyme that is responsible to produce NO in ECs. Exposure to CS in EC can reduce the expression of eNOS genes and proteins, resulting EC dysfunction (He et al, 2017; Su et al, 1998). A decrease of eNOS and NO levels will increase vascular tone, increase expression of adhesion molecules, trigger coagulation cascade and inflammation (Kaur et al, 2018), and increases the risk of atherosclerotic CVDs (Fig. 4). Measurement of FMD represents a useful tool to assess the impacts of smoking on the vascular wall and the efficacy of treatment options for early smoking-induced proatherogenic changes in the vasculature. CS showed a significant decrease in FMD (Esen et al, 2004). A possible mechanism for lowering FMD induced by CS is reduced production of NO and increased expression of adhesion molecules, leading to impaired endothelial function.

FIG. 4.

FIG. 4.

The schematic figure showed that CS and nicotine induce endothelial dysfunction by binding to different types of DAMP receptors and causing direct damage to endothelial cells, decreasing eNOS and NO bioavailability leading to increased oxidative stress and adhesion molecules, therefore increasing inflammatory response and vascular inflammation to promote initiation and progression of cardiovascular diseases. eNOS, endothelial nitric oxide synthase; NO, nitric oxide.

Conclusion

Cigarette smoke (CS) has a major impact on health issues worldwide. Many of the health care consequences of cigarette smoking could be related to its ability to depress the immune system although the mechanisms by which CS alters immunity are not completely understood. The toxic chemicals present in CS have broad immunomodulatory consequences that include altered innate and adaptive immunity and interrupt immunological homeostasis. This leads to several diseases and exerts paradoxical effects on inflammation. We published one such inflammation paradox between proinflammatory cytokines and microRNAs (Liu et al, 2020). We observed increased ROS and oxidative stress generation, increased DNA damage and cell death, as well as metabolic reprogramming and epigenetic modification. In particularly, CS acts as a double-edged sword that either exacerbates pathological immune responses or attenuates the defensive function of the immune system, possibly due to the complexities and functional diversities of CS components.

Even though CS exerts diverse effects on immune responses, the net effect is deleterious rather than beneficial. We have reviewed experimental evidence that showed (i) the signals of DAMP receptors contribute to CS modulation of inflammation and immunity; (ii) CS reprograms immunometabolism and trained immunity-/trained innate immune tolerance-related metabolic pathways in innate immune cells and T cells; (iii) five nucleus-localized ROS activate CS-promoted DNA damage and cell death pathways to potentially amplify inflammation and immune responses; and (iv) CS increases ROS and EC activation/dysfunction to induce vascular inflammation and subsequently promotes CVDs.

Cigarette smoking attributes significantly to CHD deaths and doubles the risk of ischemic stroke. Smoking acts synergistically with other disease risk factors, substantially increasing the risk of CHD, peripheral vascular disease, cancer, chronic lung disease, and many other chronic diseases (Shinton and Beevers, 1989). In the presence of a high-fat diet (hyperlipidemia), a known trained immunity inducer (Christ et al, 2018), nicotine exposure significantly increased vascular inflammation and atherosclerosis in apolipoprotein E-deficient (ApoE−/−) mice (Wu et al, 2018). CS in angiotensin II-induced hypertension increased mitochondrial oxidative stress, which contributes to endothelial dysfunction and CVDs (Dikalov et al, 2019). Furthermore, smoking substantially increases the risk of CVD in patients with type 2 diabetes (Yang et al, 2022).

Cigarette smoking is known to both increase susceptibility to infection and drives inflammation. Exposure to CS impaired host defense response and immune suppressive effects by suppressing multiple host defense mechanisms such as epithelial cell responses and recruitment and activation of innate immune cells including monocytes/macrophages, neutrophils, and NK cells. Recently, smokers have been found to be at a higher risk of developing severe forms of coronavirus disease 2019 (COVID-19) (Masso-Silva et al, 2021). E-cigarette aerosol inhalation (vaping) has been associated with several inflammatory lung disorders. Previous study demonstrated that mice exposed to e-cigarette aerosol showed a marked reduction in IgA immunoglobulins and CD4 T cells associated with increased neutrophil activation, indicating that e-cigarette vapers may be at a higher risk of increasing the immune response and developing inflammatory disorders of the lungs (Masso-Silva et al, 2021). E-cigarette-exposed alveolar macrophages show a significant increase in the production of proinflammatory cytokines/chemokines, apoptosis and necrosis, and ROS production (Scott et al, 2018), as well as oxidative stress, ox-LDL, and cardiovascular risk (Carnevale et al, 2016; Moheimani et al, 2017).

Chronic e-cigarette exposure induces vascular endothelial dysfunction, cardiac dysfunction, and atherosclerosis in mice (El-Mahdy et al, 2021; Espinoza-Derout et al, 2019) and increases risk for COPD (Bowler et al, 2017) and CVDs (D'Amario et al, 2019). On the contrary, another clinical study has found that CS or vaping e-cigarettes showed a reduction in the expression of immune-related genes. Also, vaping e-cigarettes was associated with suppression of a large number of unique genes and e-cigarette users showed greater suppression of genes common with those changed in cigarette smokers.

Furthermore, vaping e-cigarettes suppressed the expression of transcription factors, such as early growth response 1, which was functionally associated with decreased expression of 5 target genes in cigarette smokers and 18 target genes in e-cigarette users (Martin et al, 2016).

CS modulates the immune functions of ECs. CS exposure increases pulmonary endothelial barrier permeability and causes endothelial activation/dysfunction. CS decreased eNOS and inhibited eNOS activity. In addition, CS increased ROS production, and oxidative stress-mediated reduction in eNOS-NO signaling is a significant contributor to CS-induced vascular endothelial dysfunction. CS stimulates the expression of P-selectin, E-selectin, ICAM-1, and VCAM-1, as well as cytokines and chemokines, including TNF-α, IL-6, and IL-1β, via NADPH oxidase-dependent NF-κB transcriptional activation. CS also increases adherence of monocytes to the endothelium and transendothelial migration, as well as neutrophil transmigration across HUVECs and induced upregulation of the CXCR3 receptor in ECs. CS synergizes with the inflammatory cytokine IL-1β to increase vascular permeability and endothelial dysfunction via ROS/p38/phosphatase and tensin homolog-mediated tyrosinephosphorylation of vascular endothelial cadherin and β-catenin and subsequent β-catenin nuclear translocation and expression of inflammatory genes, such as COX-2 in cardiac ECs.

In our working model (Fig. 5), we showed that after an initial exposure to the first stimulus, innate immune cells with “memory” traits respond more rapidly and with a high magnitude to secondary stimulation. After the secondary stimulus, cells can respond by increasing the proinflammatory mediator, mediated by (i) metabolic reprogramming involving increased aerobic glycolysis, increased acetyl CoA generation, increased mevalonate synthesis, glutaminolysis, and increased production of lactate and fumarate; and (ii) epigenetic modification including increased H3K27ac and H3K4me3. Also, cell can respond with decreasing the magnitude of immune response (innate immune tolerance) characterized by decreased proinflammatory and increased anti-inflammatory mediators mediated by metabolic reprogramming including decreased aerobic glycolysis, increased production of itaconate metabolites, as well as epigenetic rewiring including increased H3K9me3.

FIG. 5.

FIG. 5.

Our working model. Representative model of innate immune memory and immune tolerance response. After initial exposure to the first stimulus, innate immune cells with “memory” traits respond rapidly with a high magnitude of immune response to the secondary stimulation, increased proinflammatory mediators mediated by metabolic reprogramming such as increased aerobic glycolysis, increased acetyl CoA generation, increased mevalonate synthesis, glutaminolysis, and increased production of fumarate and lactate as well as epigenetic modification such as increased H3K27ac and H3K4me3. Also, after exposure to the first stimulus, cells respond with a decreased magnitude of innate immune response (tolerance) characterized by decreased proinflammatory and increased anti-inflammatory mediators mediated by metabolic reprogramming such as decreased aerobic glycolysis, increased production of itaconate metabolites, as well as epigenetic rewiring such as increased and H3K9me3. H3K4me3, histone 3 lysine 4 trimethylations; H3K9me3, histone 3 lysine 9 trimethylation; H3K27ac, histone 3 lysine 27 acetylations.

The carcinogenic and immunomodulatory toxins in CS bind to several receptors on the cell membrane, cytosol, and nucleus leading to increases in the cytosolic ROS and five nuclear ROS expression. This leads to the induction of seven types of cell death and subsequent alarmins and NET release, which amplify inflammation and induced trained immunity. Metabolic reprogramming induced by CS can result in the generation of acetyl and methyl groups and induce histone remodeling and exacerbate immune responses to induce trained immunity. CS exposure significantly upregulates the IRG1 expression and increases the abundance of itaconate metabolites (Titz et al, 2020) resulting in immunosuppression.

The anti-inflammatory effect of CS is counteracted by the proinflammatory effect of other CS constituents, resulting in reduced immunosuppression effects and increased inflammation and trained immunity in a phenomenon known as inflammation paradox (Liu et al, 2020; Virtue et al, 2017) or second inflammation wave as we proposed (Johnson et al, 2020; Liu et al, 2020) (Fig. 6).

FIG. 6.

FIG. 6.

Our working model. The carcinogenic and immunomodulatory toxins in CS bind to different receptors on the cell membrane, cytosol, and nucleus leading to increased cytoplasmic (cytosolic and non-nucleus organelle) ROS, which further induces five different nuclear ROS. Increased ROS production results in the induction of seven types of cell death and subsequent alarmins and neutrophil extracellular trap release, which amplify inflammation and induce trained immunity. Metabolic reprogramming induced by CS can induce epigenetic remodeling and exacerbate immune repose to induce trained immunity. CS exposure significantly upregulates the IRG1 expression and increases the abundance of itaconate metabolites resulting in immunosuppression. The anti-inflammatory effect of CS is counteracted by the proinflammatory effect of other CS constituents resulting in reduced immunosuppression effects and increased inflammation and trained immunity in a phenomenon known as inflammation paradox1 or second inflammation wave. AChR, acetylcholine receptor; CLRs, C-type lectin receptors; GPCRs, G-protein-coupled receptors; NLRs, NOD-like receptors; PAMPs, pathogen-associated molecular patterns; RAGE, receptor for advanced glycation end products; RLRs, retinoic acid-inducible gene-like receptors; TREMs, triggering receptors expressed on myeloid cells; TLRs, toll-like receptors.

In conclusion, the effects of CS on immune responses are wide-ranging and complex, and both potentiation (proinflammatory/trained immunity) and attenuation (anti-inflammatory/trained tolerance) may be induced. The net effect of CS on the immune response and inflammation depends on many variables, including the dose and duration of CS use, cell types, tissue types, and contexture, and the presence of other factors at the time of immune cell stimulation. The recognition of specific mechanisms by which CS affects immune response is an important step toward elucidating mechanisms of CS-induced diseases and may identify novel therapeutic approaches for the management of diseases that afflict smokers.

Acknowledgments

F.S. carried out the primary literature search and drafted the article. Others provided material input and helped in revising the article. X.F.Y. conceived the study and provided field expertise. All the authors read and approved the final article.

Abbreviations Used

α7nAChR

α7 nicotinic acetylcholine receptor

Ach

acetylcholine

AChR

acetylcholine receptor

AhR

aryl hydrocarbon receptor

ALDOA

aldolase, fructose-bisphosphate A

ApoE−/−

apolipoprotein E deficient

ATF3

activating transcription factor 3

B[k]F

benzo[k]fluoranthene

BaP

benzo[a]pyrene

BD

1,3-butadiene

BMDMs

bone marrow-derived macrophages

CAD

cis-aconitate decarboxylase

CCL20

chemokine (C-C motif) ligand 20

CCR6

C-C motif chemokine receptor 6

CDs

clusters of differentiation

CHD

coronary heart disease

CKD

chronic kidney diseases

CLRs

C-type lectin receptors

COPD

chronic obstructive pulmonary disease

COVID-19

coronavirus disease 2019

COX-2

cyclooxygenase-2

CpG

cytosine-phosphate-guanine

CS

cigarette smoke

CSC

cigarette smoke condensate

CSE

cigarette smoke extract

CTLs

CD8+ cytotoxic T lymphocytes

CVD

cardiovascular disease

CXCL8

C-X-C motif chemokine ligand 8

CXCR3

C-X-C motif chemokine receptor 3

CYP1A1

cytochrome P450 family 1 subfamily A member 1

DAMPs

danger-associated molecular pattern

DCs

dendritic cells

DNMTs

DNA methyltransferases

dsDNA

double-stranded DNA

e-cigarettes

electronic cigarettes

ECs

endothelial cells

EMAP II

endothelial monocyte-activating polypeptide II

eNOS

endothelial nitric oxide synthase

ESRD

end-stage kidney disease

FA

formaldehyde

FMD

flow-mediated dilation

Foxp3

forkhead box P3

GAPDH

glyceraldehyde-3-phosphate dehydrogenase

GMCSF

granulocyte/macrophage-colony-stimulating factor

GPCRs

G-protein-coupled receptors

GSDMD

gasdermin D

H1R

histamine receptor 1

H2O2

hydrogen peroxide

H3K27ac

histone 3 lysine 27 acetylation

H3K4me3

histone 3 lysine 4 trimethylation

H3K9me2

histone 3 lysine 9 dimethylation

H3K9me3

histone 3 lysine 9 trimethylation

HAECs

human aortic endothelial cells

HATs

histone acetyltransferases

HDACs

histone deacetylases

HFD

high-fat diet

HLMVECs

human lung microvascular endothelial cells

HMEC-1

human microvascular endothelial cell 1

HMGB1

high-mobility group box protein 1

HMGN1

high-mobility group nucleosome binding domain 1

HOCl

hypochlorous acid

HPBECs

human primary bronchial epithelial cells

HuAoSMCs

human aorta primary smooth muscle cells

HUVECs

human umbilical vein endothelial cells

IBD

inflammatory bowel disease

ICAM-1

intercellular adhesion molecule 1

IFN-γ

interferon-gamma

IκBζ

inhibitor of nuclear factor-kappa B zeta

IL

interleukin

iNOS

inducible nitric oxide synthase

IP-10

interferon-inducible protein–10

IRG1

immune responsive gene 1

IRI

ischemia–reperfusion injury

JNK

c-Jun N-terminal kinase

Keap1

kelch-like ECH-associated protein 1

LDH

lactate-dehydrogenase

LDL

low-density lipoprotein

LPS

lipopolysaccharide

MDA

malondialdehyde

MDMs

monocyte-derived macrophages

MFF

mitochondrial fission factor

MHC-II

MHC class II molecules

MPO

myeloperoxidase

MS

multiple sclerosis

mTOR

mammalian target of rapamycin

NADPH

nicotinamide adenine dinucleotide phosphate

NETs

neutrophil extracellular traps

NF-κB

nuclear factor kappa B

NK

natural killer

NLRP3

NOD-, LRR-, and pyrin domain-containing protein 3

NLRs

NOD-like receptors

NNK

4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone

NNN

N′-nitrosonornicotine

NO

nitric oxide

NO2

nitrogen dioxide

NOx

nitrogen oxides

Nrf2

nuclear factor erythroid 2-related factor 2

NTHI

nontypeable Haemophilus influenzae

O2

superoxide

OCR

oxygen consumption rate

OH

hydroxyl radical

OPA1

optic atrophy protein 1

ORs

odorant receptors

ox-LDL

oxidized low-density lipoprotein

PAF

platelet-activating factor

PAMPs

pathogen-associated molecular patterns

PARP

poly(ADP-ribose) polymerase

PBMCs

peripheral blood mononuclear cells

PI3-AKT

phosphoinositide-3-kinase–protein kinase B

PMA

phorbol myristate acetate

PMA

phorbol myristate acetate

PMNs

polymorphonuclear neutrophils

PRMT6

protein arginine methyltransferase 6

RA

rheumatoid arthritis

RAGE

receptor for advanced glycation end products

RLRs

retinoic acid-inducible gene-like receptors

RNS

reactive nitric species

ROS

reactive oxygen species

SDH

succinate dehydrogenase

sICAM-1

soluble intercellular adhesion molecule 1

SLE

systemic lupus erythematosus

SOD

superoxide dismutase

STAT3

signal transducer and activator of transcription 3

TCA

tricarboxylic acid

TGF-β

transforming growth factor-β

Th

T helper cell

TICAM1

toll-like receptor adaptor molecule 1

TLRs

toll-like receptors

TNF-α

tumor necrosis factor-α

Tregs

T regulatory cells

TREMs

triggering receptors expressed on myeloid cells

TS

tobacco smoke

TXNIP

thioredoxin-interacting protein

UC

ulcerative colitis

VCAM-1

vascular cell adhesion molecule 1

VEGF

vascular endothelial growth factor

VLDL

very-low-density lipoprotein

VSMCs

vascular smooth muscle cells

WT

wild type

Author Disclosure Statement

No competing financial interests exist.

Funding Information

Our research activities are supported by grants from the National Institutes of Health (NIH)/(HL131460, HL132399, HL138749, HL147565, HL130233, DK104116, DK113775, P30 DA13429, RO1 DA040619, and RO1 DA049745). The content in this article is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

References

  1. Agarwal AR, Yin F, Cadenas E. Short-term cigarette smoke exposure leads to metabolic alterations in lung alveolar cells. Am J Respir Cell Mol Biol 2014;51(2):284–293; doi: 10.1165/rcmb.2013-0523OC [DOI] [PubMed] [Google Scholar]
  2. AlQasrawi D, Qasem A, Naser SA. Divergent effect of cigarette smoke on innate immunity in inflammatory bowel disease: A nicotine-infection interaction. Int J Mol Sci 2020;21(16):E5801; doi: 10.3390/ijms21165801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alrouji M, Manouchehrinia A, Gran B, et al. . Effects of cigarette smoke on immunity, neuroinflammation and multiple sclerosis. J Neuroimmunol 2019;329:24–34; doi: 10.1016/j.jneuroim.2018.10.004 [DOI] [PubMed] [Google Scholar]
  4. Ambalavanan N, Carlo WF, Bulger A, et al. . Effect of cigarette smoke extract on neonatal porcine vascular smooth muscle cells. Toxicol Appl Pharmacol 2001;170(2):130–136; doi: 10.1006/taap.2000.9094 [DOI] [PubMed] [Google Scholar]
  5. Ambrose JA, Barua RS. The pathophysiology of cigarette smoking and cardiovascular disease: An update. J Am Coll Cardiol 2004;43(10):1731–1737; doi: 10.1016/j.jacc.2003.12.047 [DOI] [PubMed] [Google Scholar]
  6. Aoshiba K, Nagai A. Oxidative stress, cell death, and other damage to alveolar epithelial cells induced by cigarette smoke. Tob Induc Dis 2003;1(3):219–226; doi: 10.1186/1617-9625-1-3-219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Aridgides DS, Mellinger DL, Armstrong DA, et al. . Functional and metabolic impairment in cigarette smoke-exposed macrophages is tied to oxidative stress. Sci Rep 2019;9(1):9624; doi: 10.1038/s41598-019-46045-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res 2011;21(3):381–395; doi: 10.1038/cr.2011.22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Barreiro E, del Puerto-Nevado L, Puig-Vilanova E, et al. . Cigarette smoke-induced oxidative stress in skeletal muscles of mice. Respir Physiol Neurobiol 2012;182(1):9–17; doi: 10.1016/j.resp.2012.02.001 [DOI] [PubMed] [Google Scholar]
  10. Barua RS, Sharma M, Dileepan KN. Cigarette smoke amplifies inflammatory response and atherosclerosis progression through activation of the H1R-TLR2/4-COX2 axis. Front Immunol 2015;6:572; doi: 10.3389/fimmu.2015.00572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Batista-Gonzalez A, Vidal R, Criollo A, et al. . New insights on the role of lipid metabolism in the metabolic reprogramming of macrophages. Front Immunol 2019;10:2993; doi: 10.3389/fimmu.2019.02993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bauer CMT, Morissette MC, Stämpfli MR. The influence of cigarette smoking on viral infections: Translating bench science to impact COPD pathogenesis and acute exacerbations of COPD clinically. Chest 2013;143(1):196–206; doi: 10.1378/chest.12-0930 [DOI] [PubMed] [Google Scholar]
  13. Bekkering S, Domínguez-Andrés J, Joosten LAB, et al. . Trained immunity: Reprogramming innate immunity in health and disease. Annu Rev Immunol 2021;39:667–693; doi: 10.1146/annurev-immunol-102119-073855 [DOI] [PubMed] [Google Scholar]
  14. Berg RE, Forman J. The role of CD8 T cells in innate immunity and in antigen non-specific protection. Curr Opin Immunol 2006;18(3):338–343; doi: 10.1016/j.coi.2006.03.010 [DOI] [PubMed] [Google Scholar]
  15. Bermudez EA, Rifai N, Buring JE, et al. . Relation between markers of systemic vascular inflammation and smoking in women. Am J Cardiol 2002;89(9):1117–1119; doi: 10.1016/s0002-9149(02)02284-1 [DOI] [PubMed] [Google Scholar]
  16. Bowler RP, Hansel NN, Jacobson S, et al. . Electronic cigarette use in US adults at risk for or with COPD: Analysis from two observational cohorts. J Gen Intern Med 2017;32(12):1315–1322; doi: 10.1007/s11606-017-4150-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Braun M, Klingelhöfer D, Müller R, et al. . The impact of second-hand smoke on nitrogen oxides concentrations in a small interior. Sci Rep 2021;11(1):11703; doi: 10.1038/s41598-021-90994-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Breitling LP, Salzmann K, Rothenbacher D, et al. . Smoking, F2RL3 methylation, and prognosis in stable coronary heart disease. Eur Heart J 2012;33(22):2841–2848; doi: 10.1093/eurheartj/ehs091 [DOI] [PubMed] [Google Scholar]
  19. Breitling LP, Yang R, Korn B, et al. . Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am J Hum Genet 2011;88(4):450–457; doi: 10.1016/j.ajhg.2011.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Buscetta M, Di Vincenzo S, Miele M, et al. . Cigarette smoke inhibits the NLRP3 inflammasome and leads to caspase-1 activation via the TLR4-TRIF-caspase-8 axis in human macrophages. FASEB J 2020;34(1):1819–1832; doi: 10.1096/fj.201901239R [DOI] [PubMed] [Google Scholar]
  21. Carnevale R, Sciarretta S, Violi F, et al. . Acute impact of tobacco vs electronic cigarette smoking on oxidative stress and vascular function. Chest 2016;150(3):606–612; doi: 10.1016/j.chest.2016.04.012 [DOI] [PubMed] [Google Scholar]
  22. Centers for Disease Control (CDC). The Surgeon General's 1989 Report on Reducing the Health Consequences of Smoking: 25 Years of Progress. MMWR Suppl 1989;38(2):1–32. [PubMed] [Google Scholar]
  23. Centers for Disease Control and Prevention (U.S.), National Center for Chronic Disease Prevention and Health Promotion (U.S.), and Office on Smoking and Health (US). How Tobacco Smoke Causes Disease: The Biology and Behavioral Basis for Smoking-Attributable Disease: A Report of the Surgeon General. Publications and Reports of the Surgeon General. Centers for Disease Control and Prevention: Atlanta, GA; 2010. [PubMed] [Google Scholar]
  24. Cheng S-C, Quintin J, Cramer RA, et al. . mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 2014;345(6204):1250684; doi: 10.1126/science.1250684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cheng T. Chemical evaluation of electronic cigarettes. Tob Control 2014;23(Suppl 2):ii11–ii17; doi: 10.1136/tobaccocontrol-2013-051482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Choi HS, Han K-D, Oh TR, et al. . Smoking and risk of incident end-stage kidney disease in general population: A nationwide population-based cohort study from Korea. Sci Rep 2019;9(1):19511; doi: 10.1038/s41598-019-56113-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chou W-C, Rampanelli E, Li X, et al. . Impact of intracellular innate immune receptors on immunometabolism. Cell Mol Immunol 2022;19(3):337–351; doi: 10.1038/s41423-021-00780-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Christ A, Günther P, Lauterbach MAR, et al. . Western diet triggers NLRP3-dependent innate immune reprogramming. Cell 2018;172(1–2):162–175.e14; doi: 10.1016/j.cell.2017.12.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Corretti MC, Anderson TJ, Benjamin EJ, et al. . Guidelines for the ultrasound assessment of endothelial-dependent flow-mediated vasodilation of the brachial artery: A report of the international brachial artery reactivity task force. J Am Coll Cardiol 2002;39(2):257–265; doi: 10.1016/s0735-1097(01)01746-6 [DOI] [PubMed] [Google Scholar]
  30. D'Amario D, Migliaro S, Borovac JA, et al. . Electronic cigarettes and cardiovascular risk: Caution waiting for evidence. Eur Cardiol 2019;14(3):151–158; doi: 10.15420/ecr.2019.16.2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Deets KA, Vance RE. Inflammasomes and adaptive immune responses. Nat Immunol 2021;22(4):412–422; doi: 10.1038/s41590-021-00869-6 [DOI] [PubMed] [Google Scholar]
  32. Delgado-Rizo V, Martínez-Guzmán MA, Iñiguez-Gutierrez L, et al. . Neutrophil extracellular traps and its implications in inflammation: An overview. Front Immunol 2017;8:81; doi: 10.3389/fimmu.2017.00081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. DiGiacomo SI, Jazayeri M-A, Barua RS, et al. . Environmental tobacco smoke and cardiovascular disease. Int J Environ Res Public Health 2018;16(1):E96; doi: 10.3390/ijerph16010096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Dikalov S, Itani H, Richmond B, et al. . Tobacco smoking induces cardiovascular mitochondrial oxidative stress, promotes endothelial dysfunction, and enhances hypertension. Am J Physiol Heart Circ Physiol 2019;316(3):H639–H646; doi: 10.1152/ajpheart.00595.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Domínguez-Andrés J, Novakovic B, Li Y, et al. . The itaconate pathway is a central regulatory node linking innate immune tolerance and trained immunity. Cell Metab 2019;29(1):211–220.e5; doi: 10.1016/j.cmet.2018.09.003 [DOI] [PubMed] [Google Scholar]
  36. Dong Y, Ming B, Dong L. The role of HMGB1 in rheumatic diseases. Front Immunol 2022;13:815257; doi: 10.3389/fimmu.2022.815257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Drummer C, Saaoud F, Shao (Inline graphic) Y, et al. Trained immunity and reactivity of macrophages and endothelial cells. Arterioscler Thromb Vasc Biol 2021a;41(3):1032–1046; doi: 10.1161/ATVBAHA.120.315452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Drummer CIV, Saaoud F, Sun Y, et al. . Hyperlipidemia may synergize with hypomethylation in establishing trained immunity and promoting inflammation in NASH and NAFLD. J Immunol Res 2021b;2021:3928323; doi: 10.1155/2021/3928323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Duan M-C, Zhang J-Q, Liang Y, et al. . Infiltration of IL-17-Producing T cells and Treg cells in a mouse model of smoke-induced emphysema. Inflammation 2016;39(4):1334–1344; doi: 10.1007/s10753-016-0365-8 [DOI] [PubMed] [Google Scholar]
  40. Duaso M, Duncan D. Health impact of smoking and smoking cessation strategies: Current evidence. Br J Community Nurs 2012;17(8):356–363; doi: 10.12968/bjcn.2012.17.8.356 [DOI] [PubMed] [Google Scholar]
  41. El-Mahdy MA, Mahgoup EM, Ewees MG, et al. . Long-term electronic cigarette exposure induces cardiovascular dysfunction similar to tobacco cigarettes: Role of nicotine and exposure duration. Am J Physiol Heart Circ Physiol 2021;320(5):H2112–H2129; doi: 10.1152/ajpheart.00997.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Esen AM, Barutcu I, Acar M, et al. . Effect of smoking on endothelial function and wall thickness of brachial artery. Circ J 2004;68(12):1123–1126; doi: 10.1253/circj.68.1123 [DOI] [PubMed] [Google Scholar]
  43. Espinoza-Derout J, Hasan KM, Shao XM, et al. . Chronic intermittent electronic cigarette exposure induces cardiac dysfunction and atherosclerosis in apolipoprotein-E knockout mice. Am J Physiol Heart Circ Physiol 2019;317(2):H445–H459; doi: 10.1152/ajpheart.00738.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Fagenson AM, Xu K, Saaoud F, et al. . Liver ischemia reperfusion injury, enhanced by trained immunity, is attenuated in caspase 1/caspase 11 double gene knockout mice. Pathog Basel Switz 2020;9(11):E879; doi: 10.3390/pathogens9110879 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Ferreira AV, Domiguéz-Andrés J, Netea MG. The role of cell metabolism in innate immune memory. J Innate Immun 2022;14(1):42–50; doi: 10.1159/000512280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Fetterman JL, Sammy MJ, Ballinger SW. Mitochondrial toxicity of tobacco smoke and air pollution. Toxicology 2017;391:18–33; doi: 10.1016/j.tox.2017.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Flores-Gomez D, Bekkering S, Netea MG, et al. . Trained immunity in atherosclerotic cardiovascular disease. Arterioscler Thromb Vasc Biol 2021;41(1):62–69; doi: 10.1161/ATVBAHA.120.314216 [DOI] [PubMed] [Google Scholar]
  48. Foglio E, Pellegrini L, Russo MA, et al. . HMGB1-mediated activation of the inflammatory-reparative response following myocardial infarction. Cells 2022;11(2):216; doi: 10.3390/cells11020216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Galluzzi L, Vitale I, Aaronson SA, et al. . Molecular mechanisms of cell death: Recommendations of the nomenclature committee on cell death 2018. Cell Death Differ 2018;25(3):486–541; doi: 10.1038/s41418-017-0012-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gong T, Liu L, Jiang W, et al. . DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol 2020;20(2):95–112; doi: 10.1038/s41577-019-0215-7 [DOI] [PubMed] [Google Scholar]
  51. Goodson WH, Lowe L, Carpenter DO, et al. . Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: The challenge ahead. Carcinogenesis 2015;36(Suppl 1):S254–S296; doi: 10.1093/carcin/bgv039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Haw TJ, Starkey MR, Pavlidis S, et al. . Toll-like receptor 2 and 4 have opposing roles in the pathogenesis of cigarette smoke-induced chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2018;314(2):L298–L317; doi: 10.1152/ajplung.00154.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. He Z, Chen Y, Hou C, et al. . Cigarette smoke extract changes expression of endothelial nitric oxide synthase (ENOS) and P16(INK4a) and is related to endothelial progenitor cell dysfunction. Med Sci Monit Int Med J Exp Clin Res 2017;23:3224–3231; doi: 10.12659/msm.902746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Horvathova M, Jahnova E, Szabova M, et al. . The relationship between cell surface markers, cytokines, ageing, and cigarette smoking. Bratisl Lek Listy 2009;110(7):394–400. [PubMed] [Google Scholar]
  55. Hybertson BM, Gao B, Bose SK, et al. . Oxidative stress in health and disease: The therapeutic potential of Nrf2 activation. Mol Aspects Med 2011;32(4–6):234–246; doi: 10.1016/j.mam.2011.10.006 [DOI] [PubMed] [Google Scholar]
  56. Ishii Y. Smoking and respiratory diseases [in Japanese]. Nihon Rinsho Jpn J Clin Med 2013;71(3):416–420. [PubMed] [Google Scholar]
  57. Ito K, Ito M, Elliott WM, et al. . Decreased histone deacetylase activity in chronic obstructive pulmonary disease. N Engl J Med 2005;352(19):1967–1976; doi: 10.1056/NEJMoa041892 [DOI] [PubMed] [Google Scholar]
  58. Jamal A, King BA, Neff LJ, et al. . Current cigarette smoking among adults—United States, 2005–2015. MMWR Morb Mortal Wkly Rep 2016;65(44):1205–1211; doi: 10.15585/mmwr.mm6544a2 [DOI] [PubMed] [Google Scholar]
  59. Jamaluddin MDS, Chen I, Yang F, et al. . Homocysteine inhibits endothelial cell growth via DNA hypomethylation of the cyclin A gene. Blood 2007;110(10):3648–3655; doi: 10.1182/blood-2007-06-096701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Jaspers I. Cigarette smoke effects on innate immune mechanisms in the nasal mucosa. potential effects on the microbiome. Ann Am Thorac Soc 2014;11(Suppl 1):S38–S42; doi: 10.1513/AnnalsATS.201306-154MG [DOI] [PubMed] [Google Scholar]
  61. Johnson C, Drummer Iv C, Shan H, et al. . A novel subset of CD95+ pro-inflammatory macrophages overcome miR155 deficiency and may serve as a switch from metabolically healthy obesity to metabolically unhealthy obesity. Front Immunol 2020;11:619951; doi: 10.3389/fimmu.2020.619951 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kaur R, Kaur M, Singh J. Endothelial dysfunction and platelet hyperactivity in type 2 diabetes mellitus: Molecular insights and therapeutic strategies. Cardiovasc Diabetol 2018;17(1):121; doi: 10.1186/s12933-018-0763-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Ke X, Wang J, Li L, et al. . Roles of CD4+CD25(High) FOXP3+ Tregs in lymphomas and tumors are complex. Front Biosci 2008;13:3986–4001; doi: 10.2741/2986 [DOI] [PubMed] [Google Scholar]
  64. Kearley J, Silver JS, Sanden C, et al. . Cigarette smoke silences innate lymphoid cell function and facilitates an exacerbated type I interleukin-33-dependent response to infection. Immunity 2015;42(3):566–579; doi: 10.1016/j.immuni.2015.02.011 [DOI] [PubMed] [Google Scholar]
  65. Khanna A, Guo M, Mehra M, et al. . Inflammation and oxidative stress induced by cigarette smoke in lewis rat brains. J Neuroimmunol 2013;254(1–2):69–75; doi: 10.1016/j.jneuroim.2012.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kim M, Han C-H, Lee M-Y. NADPH oxidase and the cardiovascular toxicity associated with smoking. Toxicol Res 2014;30(3):149–157; doi: 10.5487/TR.2014.30.3.149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kim MS, Huang Y, Lee J, et al. . Cellular transformation by cigarette smoke extract involves alteration of glycolysis and mitochondrial function in esophageal epithelial cells. Int J Cancer 2010;127(2):269–281; doi: 10.1002/ijc.25057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Knopik VS, Maccani MA, Francazio S, et al. . The epigenetics of maternal cigarette smoking during pregnancy and effects on child development. Dev Psychopathol 2012;24(4):1377–1390; doi: 10.1017/S0954579412000776 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kohut SJ. Interactions between nicotine and drugs of abuse: A review of preclinical findings. Am J Drug Alcohol Abuse 2017;43(2):155–170; doi: 10.1080/00952990.2016.1209513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Kowalska M, Piekut T, Prendecki M, et al. . Mitochondrial and nuclear DNA oxidative damage in physiological and pathological aging. DNA Cell Biol 2020;39(8):1410–1420; doi: 10.1089/dna.2019.5347 [DOI] [PubMed] [Google Scholar]
  71. Krause BM, Bauer B, Neudörfl J-M, et al. . ItaCORMs: Conjugation with a CO-releasing unit greatly enhances the anti-inflammatory activity of itaconates. RSC Med Chem 2021;12(12):2053–2059; doi: 10.1039/d1md00163a [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Krauss-Etschmann S, Meyer KF, Dehmel S, et al. Inter- and transgenerational epigenetic inheritance: Evidence in asthma and COPD? Clin Epigenetics 2015;7(1):53; doi: 10.1186/s13148-015-0085-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Kroemer G, Galassi C, Zitvogel L, et al. . Immunogenic cell stress and death. Nat Immunol 2022;23(4):487–500; doi: 10.1038/s41590-022-01132-2 [DOI] [PubMed] [Google Scholar]
  74. Künzi L, Holt GE. Cigarette smoke activates the parthanatos pathway of cell death in human bronchial epithelial cells. Cell Death Discov 2019;5:127; doi: 10.1038/s41420-019-0205-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Kwesi-Maliepaard EM, Jacobs H, van Leeuwen F. Signals for antigen-independent differentiation of Memory CD8+ T cells. Cell Mol Life Sci 2021;78(19–20):6395–6408; doi: 10.1007/s00018-021-03912-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kwon Y-M, Park JH, Kim H, et al. . Different susceptibility of increased DNMT1 expression by exposure to tobacco smoke according to histology in primary non-small cell lung cancer. J Cancer Res Clin Oncol 2007;133(4):219–226; doi: 10.1007/s00432-006-0160-2 [DOI] [PubMed] [Google Scholar]
  77. Lai B, Wang J, Fagenson A, et al. . Twenty novel disease group-specific and 12 new shared macrophage pathways in eight groups of 34 diseases including 24 inflammatory organ diseases and 10 types of tumors. Front Immunol 2019;10:2612; doi: 10.3389/fimmu.2019.02612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Lampropoulou V, Sergushichev A, Bambouskova M, et al. . Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab 2016;24(1):158–166; doi: 10.1016/j.cmet.2016.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Lee H-G, Cho M-Z, Choi J-M. Bystander CD4+ T cells: Crossroads between innate and adaptive immunity. Exp Mol Med 2020;52(8):1255–1263; doi: 10.1038/s12276-020-00486-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Li X, Fang P, Li Y, et al. . Mitochondrial reactive oxygen species mediate lysophosphatidylcholine-induced endothelial cell activation. Arterioscler Thromb Vasc Biol 2016a;36(6):1090–1100; doi: 10.1161/ATVBAHA.115.306964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Li X, Fang P, Mai J, et al. . Targeting mitochondrial reactive oxygen species as novel therapy for inflammatory diseases and cancers. J Hematol OncolJ Hematol Oncol 2013;6:19; doi: 10.1186/1756-8722-6-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Li X, Fang P, Sun Y, et al. . Anti-inflammatory cytokines IL-35 and IL-10 block atherogenic lysophosphatidylcholine-induced, mitochondrial ROS-mediated innate immune activation, but spare innate immune memory signature in endothelial cells. Redox Biol 2020;28:101373; doi: 10.1016/j.redox.2019.101373 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Li X, Mai J, Virtue A, et al. . IL-35 is a novel responsive anti-inflammatory cytokine—A new system of categorizing anti-inflammatory cytokines. PLoS One 2012;7(3):e33628; doi: 10.1371/journal.pone.0033628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Li X, Shao Y, Sha X, et al. . IL-35 (interleukin-35) suppresses endothelial cell activation by inhibiting mitochondrial reactive oxygen species-mediated site-specific acetylation of H3K14 (histone 3 lysine 14). Arterioscler Thromb Vasc Biol 2018;38(3):599–609; doi: 10.1161/ATVBAHA.117.310626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Li Y-F, Huang X, Li X, et al. . Caspase-1 mediates hyperlipidemia-weakened progenitor cell vessel repair. Front Biosci Landmark Ed 2016b;21(1):178–191; doi: 10.2741/4383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Li Y-F, Nanayakkara G, Sun Y, et al. . Analyses of caspase-1-regulated transcriptomes in various tissues lead to identification of novel IL-1β-, IL-18- and Sirtuin-1-independent pathways. J Hematol Oncol 2017;10(1):40; doi: 10.1186/s13045-017-0406-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Liu M, Saredy J, Zhang R, et al. . Approaching inflammation paradoxes-proinflammatory cytokine blockages induce inflammatory regulators. Front Immunol 2020;11:554301; doi: 10.3389/fimmu.2020.554301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Liu M, Wu N, Xu K, et al. . Organelle crosstalk regulators are regulated in diseases, tumors, and regulatory T cells: Novel classification of organelle crosstalk regulators. Front Cardiovasc Med 2021;8:713170; doi: 10.3389/fcvm.2021.713170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Lopez-Pastrana J, Ferrer LM, Li Y-F, et al. . Inhibition of caspase-1 activation in endothelial cells improves angiogenesis: A novel therapeutic potential for ischemia. J Biol Chem 2015;290(28):17485–17494; doi: 10.1074/jbc.M115.641191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Lu Y, Nanayakkara G, Sun Y, et al. . Procaspase-1 patrolled to the nucleus of proatherogenic lipid LPC-activated human aortic endothelial cells induces ROS promoter CYP1B1 and strong inflammation. Redox Biol 2021;47:102142; doi: 10.1016/j.redox.2021.102142 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Lu Y, Sun Y, Drummer C, et al. . Increased acetylation of H3K14 in the genomic regions that encode trained immunity enzymes in lysophosphatidylcholine-activated human aortic endothelial cells—Novel qualification markers for chronic disease risk factors and conditional DAMPs. Redox Biol 2019;24:101221; doi: 10.1016/j.redox.2019.101221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Lu Y, Sun Y, Xu K, et al. . Aorta in pathologies may function as an immune organ by upregulating secretomes for immune and vascular cell activation, differentiation and trans-differentiation-early secretomes may serve as drivers for trained immunity. Front Immunol 2022;13:858256; doi: 10.3389/fimmu.2022.858256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lugg ST, Scott A, Parekh D, et al. . Cigarette smoke exposure and alveolar macrophages: Mechanisms for lung disease. Thorax 2022;77(1):94–101; doi: 10.1136/thoraxjnl-2020-216296 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Maccani MA, Knopik VS. Cigarette smoke exposure-associated alterations to non-coding RNA. Front Genet 2012;3:53; doi: 10.3389/fgene.2012.00053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Mai J, Virtue A, Shen J, et al. . An evolving new paradigm: endothelial cells—Conditional innate immune cells. J Hematol Oncol 2013;6:61; doi: 10.1186/1756-8722-6-61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Mao C, Li D, Zhou E, et al. . Nicotine exacerbates atherosclerosis through a macrophage-mediated endothelial injury pathway. Aging 2021;13(5):7627–7643; doi: 10.18632/aging.202660 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Martin EM, Clapp PW, Rebuli ME, et al. . E-cigarette use results in suppression of immune and inflammatory-response genes in nasal epithelial cells similar to cigarette smoke. Am J Physiol Lung Cell Mol Physiol 2016;311(1):L135–L144; doi: 10.1152/ajplung.00170.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Masso-Silva JA, Moshensky A, Shin J, et al. . Chronic e-cigarette aerosol inhalation alters the immune state of the lungs and increases ACE2 expression, raising concern for altered response and susceptibility to SARS-CoV-2. Front Physiol 2021;12:649604; doi: 10.3389/fphys.2021.649604 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. McClintock TS, Khan N, Alimova Y, et al. . Encoding the odor of cigarette smoke. J Neurosci 2020;40(37):7043–7053; doi: 10.1523/JNEUROSCI.1144-20.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Michel T, Vanhoutte PM. Cellular signaling and NO production. Pflugers Arch 2010;459(6):807–816; doi: 10.1007/s00424-009-0765-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Moheimani RS, Bhetraratana M, Yin F, et al. . Increased cardiac sympathetic activity and oxidative stress in habitual electronic cigarette users: Implications for cardiovascular risk. JAMA Cardiol 2017;2(3):278–284; doi: 10.1001/jamacardio.2016.5303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Muri J, Kopf M. Redox regulation of immunometabolism. Nat Rev Immunol 2021;21(6):363–381; doi: 10.1038/s41577-020-00478-8 [DOI] [PubMed] [Google Scholar]
  103. National Center for Chronic Disease Prevention and Health Promotion (US) Office on Smoking and Health. The Health Consequences of Smoking—50 Years of Progress: A Report of the Surgeon General. Reports of the Surgeon General. Centers for Disease Control and Prevention (US): Atlanta, GA; 2014. [PubMed] [Google Scholar]
  104. Netea MG, Domínguez-Andrés J, Barreiro LB, et al. . Defining trained immunity and its role in health and disease. Nat Rev Immunol 2020;20(6):375–388; doi: 10.1038/s41577-020-0285-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Netea MG, Joosten LAB, Latz E, et al. . Trained immunity: A program of innate immune memory in health and disease. Science 2016;352(6284):aaf1098; doi: 10.1126/science.aaf1098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Netea MG, Schlitzer A, Placek K, et al. . Innate and adaptive immune memory: An evolutionary continuum in the host's response to pathogens. Cell Host Microbe 2019;25(1):13–26; doi: 10.1016/j.chom.2018.12.006 [DOI] [PubMed] [Google Scholar]
  107. Ni D, Tang T, Lu Y, et al. . Canonical secretomes, innate immune caspase-1-, 4/11-gasdermin D non-canonical secretomes and exosomes may contribute to maintain Treg-Ness for Treg immunosuppression, tissue repair and modulate anti-tumor immunity via ROS pathways. Front Immunol 2021;12:678201; doi: 10.3389/fimmu.2021.678201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. O'Neill LAJ, Artyomov MN. Itaconate: The poster child of metabolic reprogramming in macrophage function. Nat Rev Immunol 2019;19(5):273–281; doi: 10.1038/s41577-019-0128-5 [DOI] [PubMed] [Google Scholar]
  109. Owen AM, Fults JB, Patil NK, et al. . TLR agonists as mediators of trained immunity: mechanistic insight and immunotherapeutic potential to combat infection. Front Immunol 2020;11:622614; doi: 10.3389/fimmu.2020.622614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Park J-M, Jeong H, Seo Y-S, et al. . Cigarette smoke extract produces superoxide in aqueous media by reacting with bicarbonate. Toxics 2021;9(11):316; doi: 10.3390/toxics9110316 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Pauwels NS, Bracke KR, Dupont LL, et al. . Role of IL-1α and the Nlrp3/Caspase-1/IL-1β axis in cigarette smoke-induced pulmonary inflammation and COPD. Eur Respir J 2011;38(5):1019–1028; doi: 10.1183/09031936.00158110 [DOI] [PubMed] [Google Scholar]
  112. Pryor WA. Cigarette smoke radicals and the role of free radicals in chemical carcinogenicity. Environ Health Perspect 1997;105(Suppl 4):875–882; doi: 10.1289/ehp.97105s4875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Ras RT, Streppel MT, Draijer R, et al. . Flow-mediated dilation and cardiovascular risk prediction: A systematic review with meta-analysis. Int J Cardiol 2013;168(1):344–351; doi: 10.1016/j.ijcard.2012.09.047 [DOI] [PubMed] [Google Scholar]
  114. Rogers TJ. Bidirectional regulation of opioid and chemokine function. Front Immunol 2020;11:94; doi: 10.3389/fimmu.2020.00094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Salama SA, Arab HH, Omar HA, et al. . Nicotine mediates hypochlorous acid-induced nuclear protein damage in mammalian cells. Inflammation 2014;37(3):785–792; doi: 10.1007/s10753-013-9797-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Samir P, Malireddi RKS, Kanneganti T-D. The PANoptosome: A deadly protein complex driving pyroptosis, apoptosis, and necroptosis (PANoptosis). Front Cell Infect Microbiol 2020;10:238; doi: 10.3389/fcimb.2020.00238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Sampilvanjil A, Karasawa T, Yamada N, et al. . Cigarette smoke extract induces ferroptosis in vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2020;318(3):H508–H518; doi: 10.1152/ajpheart.00559.2019 [DOI] [PubMed] [Google Scholar]
  118. Schnack L, Sohrabi Y, Lagache SMM, et al. . Mechanisms of trained innate immunity in OxLDL primed human coronary smooth muscle cells. Front Immunol 2019;10:13; doi: 10.3389/fimmu.2019.00013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Scott A, Lugg ST, Aldridge K, et al. . Pro-inflammatory effects of e-cigarette vapour condensate on human alveolar macrophages. Thorax 2018;73(12):1161–1169; doi: 10.1136/thoraxjnl-2018-211663 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Serra-Bardenys G, Peiró S. Enzymatic lysine oxidation as a posttranslational modification. FEBS J 2021. [Epub ahead of print]; doi: 10.1111/febs.16205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Seyda M, Elkhal A, Quante M, et al. . T cells going innate. Trends Immunol 2016;37(8):546–556; doi: 10.1016/j.it.2016.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Shao Y, Cheng Z, Li X, et al. . Immunosuppressive/anti-inflammatory cytokines directly and indirectly inhibit endothelial dysfunction—A novel mechanism for maintaining vascular function. J Hematol Oncol 2014;7:80; doi: 10.1186/s13045-014-0080-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Shao Y, Chernaya V, Johnson C, et al. . Metabolic diseases downregulate the majority of histone modification enzymes, making a few upregulated enzymes novel therapeutic targets—“Sand Out and Gold Stays”. J Cardiovasc Transl Res 2016;9(1):49–66; doi: 10.1007/s12265-015-9664-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Shao Y, Cornwell W, Xu K, et al. . Chronic exposure to the combination of cigarette smoke and morphine decreases CD4+ regulatory T cell numbers by reprogramming the Treg cell transcriptome. Front Immunol 2022;13:887681; doi: 10.3389/fimmu.2022.887681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Shao Y, Saredy J, Xu K, et al. . Endothelial immunity trained by coronavirus infections, DAMP stimulations and regulated by anti-oxidant NRF2 may contribute to inflammations, myelopoiesis, COVID-19 cytokine storms and thromboembolism. Front Immunol 2021a;12:653110; doi: 10.3389/fimmu.2021.653110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Shao Y, Saredy J, Yang WY, et al. . Vascular endothelial cells and innate immunity. Arterioscler Thromb Vasc Biol 2020;40(6):e138–e152; doi: 10.1161/ATVBAHA.120.314330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Shao Y, Yang WY, Saaoud F, et al. . IL-35 promotes CD4+Foxp3+ Tregs and inhibits atherosclerosis via maintaining CCR5-amplified Treg-suppressive mechanisms. JCI Insight 2021b;6(19):e152511; doi: 10.1172/jci.insight.152511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Shen H, Wu N, Nanayakkara G, et al. . Co-signaling receptors regulate T-cell plasticity and immune tolerance. Front Biosci Landmark Ed 2019;24(1):96–132; doi: 10.2741/4710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Shen Y, Rattan V, Sultana C, et al. . Cigarette smoke condensate-induced adhesion molecule expression and transendothelial migration of monocytes. Am J Physiol 1996;270(5 Pt 2):H1624–H1633; doi: 10.1152/ajpheart.1996.270.5.H1624 [DOI] [PubMed] [Google Scholar]
  130. Shenker NS, Ueland PM, Polidoro S, et al. . DNA methylation as a long-term biomarker of exposure to tobacco smoke. Epidemiol Camb Mass 2013;24(5):712–716; doi: 10.1097/EDE.0b013e31829d5cb3 [DOI] [PubMed] [Google Scholar]
  131. Shinton R, Beevers G. Meta-analysis of relation between cigarette smoking and stroke. BMJ 1989;298(6676):789–794; doi: 10.1136/bmj.298.6676.789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Siedlinski M, Klanderman B, Sandhaus RA, et al. . Association of cigarette smoking and CRP levels with DNA methylation in α-1 antitrypsin deficiency. Epigenetics 2012;7(7):720–728; doi: 10.4161/epi.20319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Stäger S, Kaye PM. CD8+ T-cell priming regulated by cytokines of the innate immune system. Trends Mol Med 2004;10(8):366–371; doi: 10.1016/j.molmed.2004.06.003 [DOI] [PubMed] [Google Scholar]
  134. Stämpfli MR, Anderson GP. How cigarette smoke skews immune responses to promote infection, lung disease and cancer. Nat Rev Immunol 2009;9(5):377–384; doi: 10.1038/nri2530 [DOI] [PubMed] [Google Scholar]
  135. Su Y, Han W, Giraldo C, et al. . Effect of cigarette smoke extract on nitric oxide synthase in pulmonary artery endothelial cells. Am J Respir Cell Mol Biol 1998;19(5):819–825; doi: 10.1165/ajrcmb.19.5.3091 [DOI] [PubMed] [Google Scholar]
  136. Sun Y, Lu Y, Saredy J, et al. . ROS systems are a new integrated network for sensing homeostasis and alarming stresses in organelle metabolic processes. Redox Biol 2020;37:101696; doi: 10.1016/j.redox.2020.101696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Sundar IK, Nevid MZ, Friedman AE, et al. . Cigarette smoke induces distinct histone modifications in lung cells: Implications for the pathogenesis of COPD and lung cancer. J Proteome Res 2014;13(2):982–996; doi: 10.1021/pr400998n [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Suter M, Ma J, Harris A, et al. . Maternal tobacco use modestly alters correlated epigenome-wide placental DNA methylation and gene expression. Epigenetics 2011;6(11):1284–1294; doi: 10.4161/epi.6.11.17819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Talhout R, Schulz T, Florek E, et al. . Hazardous compounds in tobacco smoke. Int J Environ Res Public Health 2011;8(2):613–628; doi: 10.3390/ijerph8020613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Tang D, Kang R, Berghe TV, et al. . The molecular machinery of regulated cell death. Cell Res 2019;29(5):347–364; doi: 10.1038/s41422-019-0164-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Teasdale JE, Hazell G, Newby AC, et al. . Paradoxical effects of cigarette smoke extract and high laminar flow on tumour necrosis factor-alpha induced VCAM-1 up-regulation—Implications for endothelial dysfunction. Atherosclerosis 2014;237(2):e13–e14; doi: 10.1016/j.atherosclerosis.2014.10.074 [DOI] [Google Scholar]
  142. Timilshina M, You Z, Lacher SM, et al. . Activation of mevalonate pathway via LKB1 is essential for stability of Treg cells. Cell Rep 2019;27(10):2948–2961.e7; doi: 10.1016/j.celrep.2019.05.020 [DOI] [PubMed] [Google Scholar]
  143. Titz B, Szostak J, Sewer A, et al. . Multi-omics systems toxicology study of mouse lung assessing the effects of aerosols from two heat-not-burn tobacco products and cigarette smoke. Comput Struct Biotechnol J 2020;18:1056–1073; doi: 10.1016/j.csbj.2020.04.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Valiathan R, Miguez MJ, Patel B, et al. . Tobacco smoking increases immune activation and impairs T-cell function in HIV infected patients on antiretrovirals: A cross-sectional pilot study. PLoS One 2014;9(5):e97698; doi: 10.1371/journal.pone.0097698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. van der Heijden CDCC, Noz MP, Joosten LAB, et al. . Epigenetics and trained immunity. Antioxid Redox Signal 2018;29(11):1023–1040; doi: 10.1089/ars.2017.7310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Vane JR, Anggård EE, Botting RM. Regulatory functions of the vascular endothelium. N Engl J Med 1990;323(1):27–36; doi: 10.1056/NEJM199007053230106 [DOI] [PubMed] [Google Scholar]
  147. Virtue A, Johnson C, Lopez-Pastraña J, et al. . MicroRNA-155 deficiency leads to decreased atherosclerosis, increased white adipose tissue obesity, and non-alcoholic fatty liver disease: A novel mouse model of obesity paradox. J Biol Chem 2017;292(4):1267–1287; doi: 10.1074/jbc.M116.739839 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Voss K, Hong HS, Bader JE, et al. . A guide to interrogating immunometabolism. Nat Rev Immunol 2021;21(10):637–652; doi: 10.1038/s41577-021-00529-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Wan ES, Qiu W, Baccarelli A, et al. . Cigarette smoking behaviors and time since quitting are associated with differential DNA methylation across the human genome. Hum Mol Genet 2012;21(13):3073–3082; doi: 10.1093/hmg/dds135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Wang J, Lai B, Nanayakkara G, et al. . Experimental data-mining analyses reveal new roles of low-intensity ultrasound in differentiating cell death regulatome in cancer and non-cancer cells via potential modulation of chromatin long-range interactions. Front Oncol 2019;9:600; doi: 10.3389/fonc.2019.00600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Wang L, Fu H, Nanayakkara G, et al. . Novel extracellular and nuclear caspase-1 and inflammasomes propagate inflammation and regulate gene expression: A comprehensive database mining study. J Hematol Oncol 2016a;9(1):122; doi: 10.1186/s13045-016-0351-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Wang X, Li Y-F, Nanayakkara G, et al. . Lysophospholipid receptors, as novel conditional danger receptors and homeostatic receptors modulate inflammation-novel paradigm and therapeutic potential. J Cardiovasc Transl Res 2016b;9(4):343–359; doi: 10.1007/s12265-016-9700-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Warren GW, Alberg AJ, Kraft AS, et al. . The 2014 Surgeon General's Report: “The Health Consequences of Smoking—50 Years of Progress”: A paradigm shift in cancer care. Cancer 2014;120(13):1914–1916; doi: 10.1002/cncr.28695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Wauters E, Janssens W, Vansteenkiste J, et al. . DNA methylation profiling of non-small cell lung cancer reveals a COPD-driven immune-related signature. Thorax 2015;70(12):1113–1122; doi: 10.1136/thoraxjnl-2015-207288 [DOI] [PubMed] [Google Scholar]
  155. Witschi H. A/J mouse as a model for lung tumorigenesis caused by tobacco smoke: Strengths and weaknesses. Exp Lung Res 2005;31(1):3–18; doi: 10.1080/01902140490494959 [DOI] [PubMed] [Google Scholar]
  156. Wood AM, Stockley RA. Alpha one antitrypsin deficiency: From gene to treatment. Respiration 2007;74(5):481–492; doi: 10.1159/000105536 [DOI] [PubMed] [Google Scholar]
  157. Wu X, Zhang H, Qi W, et al. . Nicotine promotes atherosclerosis via ROS-NLRP3-mediated endothelial cell pyroptosis. Cell Death Dis 2018;9(2):171; doi: 10.1038/s41419-017-0257-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Xiong Z, Yan Y, Song J, et al. . Expression of TCTP antisense in CD25(High) regulatory T cells aggravates cuff-injured vascular inflammation. Atherosclerosis 2009;203(2):401–408; doi: 10.1016/j.atherosclerosis.2008.07.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Xu K, Saaoud F, Yu S, et al. . Monocyte adhesion assays for detecting endothelial cell activation in vascular inflammation and atherosclerosis. Methods Mol Biol 2022;2419:169–182; doi: 10.1007/978-1-0716-1924-7_10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Xu K, Shao Y, Saaoud F, et al. . Novel knowledge-based transcriptomic profiling of lipid lysophosphatidylinositol-induced endothelial cell activation. Front Cardiovasc Med 2021;8:773473; doi: 10.3389/fcvm.2021.773473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Xu K, Yang WY, Nanayakkara GK, et al. . GATA3, HDAC6, and BCL6 regulate FOXP3+ Treg plasticity and determine Treg conversion into either novel antigen-presenting cell-like Treg or Th1-Treg. Front Immunol 2018;9:45; doi: 10.3389/fimmu.2018.00045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Yan Y, Xiong Z, Zhang S, et al. . CD25high T cells with a prolonged survival inhibit development of diabetes. Int J Immunopathol Pharmacol 2008;21(4):767–780; doi: 10.1177/039463200802100401 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Yang D, Han Z, Oppenheim JJ. Alarmins and immunity. Immunol Rev 2017a;280(1):41–56; doi: 10.1111/imr.12577 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Yang F, Chen IH, Xiong Z, et al. . Model of stimulation-responsive splicing and strategies in identification of immunogenic isoforms of tumor antigens and autoantigens. Clin Immunol 2006;121(2):121–133; doi: 10.1016/j.clim.2006.06.007 [DOI] [PubMed] [Google Scholar]
  165. Yang F, Yang X-F. New concepts in tumor antigens: Their significance in future immunotherapies for tumors. Cell Mol Immunol 2005;2(5):331–341 [PubMed] [Google Scholar]
  166. Yang Q, Nanayakkara GK, Drummer C, et al. . Low-intensity ultrasound-induced anti-inflammatory effects are mediated by several new mechanisms including gene induction, immunosuppressor cell promotion, and enhancement of exosome biogenesis and docking. Front Physiol 2017b;8:818; doi: 10.3389/fphys.2017.00818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Yang Q, Zhang R, Tang P, et al. . Ultrasound may suppress tumor growth, inhibit inflammation, and establish tolerogenesis by remodeling innatome via pathways of ROS, immune checkpoints, cytokines, and trained immunity/tolerance. J Immunol Res 2021;2021:6664453; doi: 10.1155/2021/6664453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Yang WY, Shao Y, Lopez-Pastrana J, et al. . Pathological conditions re-shape physiological Tregs into pathological Tregs. Burns Trauma 2015;3(1):1; doi: 10.1186/s41038-015-0001-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Yang XF, Weber GF, Cantor H. A novel Bcl-x isoform connected to the T cell receptor regulates apoptosis in T cells. Immunity 1997;7(5):629–639; doi: 10.1016/s1074-7613(00)80384-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Yang X-F, Ye Q, Press B, et al. . Analysis of the complex genomic structure of Bcl-x and its relationship to Bcl-x(Gamma) expression after CD28-dependent costimulation. Mol Immunol 2002;39(1–2):45–55; doi: 10.1016/s0161-5890(02)00049-4 [DOI] [PubMed] [Google Scholar]
  171. Yang X-F, Yin Y, Wang H. Vascular inflammation and atherogenesis are activated via receptors for PAMPs and suppressed by regulatory T cells. Drug Discov Today 2008;5(2):125–142; doi: 10.1016/j.ddstr.2008.11.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Yang Y, Peng N, Chen G, et al. . Interaction between smoking and diabetes in relation to subsequent risk of cardiovascular events. Cardiovasc Diabetol 2022;21(1):14; doi: 10.1186/s12933-022-01447-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Yang Y, Xiong Z, Zhang S, et al. . Bcl-XL inhibits T-cell apoptosis induced by expression of SARS coronavirus E protein in the absence of growth factors. Biochem J 2005;392(Pt 1):135–143; doi: 10.1042/BJ20050698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Yin M, O'Neill LAJ. The role of the electron transport chain in immunity. FASEB J 2021;35(12):e21974; doi: 10.1096/fj.202101161R [DOI] [PubMed] [Google Scholar]
  175. Yin Y, Li X, Sha X, et al. . Early hyperlipidemia promotes endothelial activation via a caspase-1-sirtuin 1 pathway. Arterioscler Thromb Vasc Biol 2015;35(4):804–816; doi: 10.1161/ATVBAHA.115.305282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Yin Y, Pastrana JL, Li X, et al. . Inflammasomes: Sensors of metabolic stresses for vascular inflammation. Front Biosci Landmark Ed 2013;18(2):638–649; doi: 10.2741/4127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Yoshiyama S, Chen Z, Okagaki T, et al. . Nicotine exposure alters human vascular smooth muscle cell phenotype from a contractile to a synthetic type. Atherosclerosis 2014;237(2):464–470; doi: 10.1016/j.atherosclerosis.2014.10.019 [DOI] [PubMed] [Google Scholar]
  178. Yu J, Qiu Y, Yang J, et al. . DNMT1-PPARγ pathway in macrophages regulates chronic inflammation and atherosclerosis development in mice. Sci Rep 2016;6:30053; doi: 10.1038/srep30053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Zeilinger S, Kühnel B, Klopp N, et al. . Tobacco smoking leads to extensive genome-wide changes in DNA methylation. PLoS One 2013;8(5):e63812; doi: 10.1371/journal.pone.0063812 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Zeng H, Nanayakkara GK, Shao Y, et al. . DNA checkpoint and repair factors are nuclear sensors for intracellular organelle stresses-inflammations and cancers can have high genomic risks. Front Physiol 2018;9:516; doi: 10.3389/fphys.2018.00516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Zhang R, Saredy J, Shao Y, et al. . End-stage renal disease is different from chronic kidney disease in upregulating ROS-modulated proinflammatory secretome in PBMCs—A novel multiple-hit model for disease progression. Redox Biol 2020a;34:101460; doi: 10.1016/j.redox.2020.101460 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Zhang R, Xu K, Shao Y, et al. . Tissue Treg secretomes and transcription factors shared with stem cells contribute to a treg niche to maintain Treg-Ness with 80% innate immune pathways, and functions of immunosuppression and tissue repair. Front Immunol 2020b;11:632239; doi: 10.3389/fimmu.2020.632239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Zhang S, Liang Y, Li L, et al. . Succinate: A novel mediator to promote atherosclerotic lesion progression. DNA Cell Biol 2022;41(3):285–291; doi: 10.1089/dna.2021.0345 [DOI] [PubMed] [Google Scholar]
  184. Zhong C, Yang X, Feng Y, et al. . Trained immunity: An underlying driver of inflammatory atherosclerosis. Front Immunol 2020;11:284; doi: 10.3389/fimmu.2020.00284 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Antioxidants & Redox Signaling are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES