INTRODUCTION

Endometriosis is a chronic gynecological disorder characterized by the abnormal growth of endometrial-like tissue outside the uterus; it affects 6–10% of women of reproductive age1,2. This condition affects millions of women worldwide and can cause significant pain, infertility, and other debilitating symptoms3,4. Biologically, endometriosis is an estrogen-dependent, chronic, and inflammatory gynecological disease that is defined by the proliferation of functional endometrial tissue developing outside the uterine cavity5. The available evidence suggests that the development of endometriosis is characterized by a complex interplay of various factors. While the exact causes of endometriosis remain elusive, researchers have explored various factors that may contribute to its development and progression. One area of interest in understanding endometriosis is the impact of environmental factors on the disease6. Among these factors, the association between endometriosis and tobacco smoking has gained attention7-9. Tobacco use, in various forms such as cigarette smoking, cigar smoking, or smokeless tobacco products, is known to have detrimental effects on human health, contributing to numerous diseases, including cardiovascular disorders, respiratory conditions, and various types of cancers10. However, the connection between tobacco smoking and endometriosis has been a subject of debate and investigation9. Thus, understanding the relationship between endometriosis and tobacco smoking is crucial for several reasons. First, endometriosis affects a significant number of women globally, and identifying modifiable risk factors can help in preventive efforts. Second, establishing a clear connection would emphasize the importance of smoking cessation interventions and raise awareness among healthcare providers and affected individuals about the potential risks associated with tobacco use. Lastly, unravelling the underlying mechanisms could pave the way for targeted therapeutic strategies to mitigate the impact of smoking on endometriosis and improve patient outcomes.

By examining the possible molecular and cellular mechanisms through which tobacco smoking may contribute to endometriosis, we can gain valuable insights into the impact of environmental exposures on this complex and often debilitating condition. Thus, this review aims to delve into the molecular understanding of the relationship between endometriosis and tobacco smoking.

PubMed/MEDLINE and Web of Science databases were used for the search, with only articles in English language, using the following terms: ‘endometriosis’, ‘tobacco’, ‘tobacco smoking’, ‘inflammation’, ‘oxidative stress’, ‘hormonal dysregulation’, ‘DNA damage’, ‘immune dysfunction’, and ‘angiogenesis’. The search strategy is provided in the Supplementary file. Two authors performed the strategy research (AV and JMA), and three authors performed the selection of the articles (AV, AF and JMA). No restriction was made for selection of studies concerning animals and humans, and also age of women11. Only original research articles were included in this review to provide information about the association between tobacco and endometriosis. Literature was searched from inception to December 2024. Based on the 4358 articles, 44 original articles were included in the narrative review.

SMOKING AND ENDOMETRIOSIS

Overview of tobacco and endometriosis

Several environmental factors, including reproductive, lifestyle and behavioral factors, have been linked to the etiology of endometriosis; however, the association with some of these factors remains inconclusive9,12-14 . Many recent studies have reported and association between tobacco smoking and increased risk in endometriosis15-17, whereas the authors of the latest meta-analysis published in 2014 concluded that there was no association between smoking and endometriosis 9. Nevertheless, the majority of the studies included in that meta-analysis were based on self-reports and provided crude estimates of association9. In contrast, a recent study, focused on more than 2 million women, has shown that women with both a family history of smoking and smoking themselves have higher risk of endometriosis than the general population (incidence rate ratio, IRR=4.28; 95% CI: 2.43–7.55)16. Among more than 500000 women, heavy tobacco users compared with never users presented a higher risk of endometriosis (summary relative risk=1.35; 95% CI: 1.15–1.59)18. Moreover, exposure to secondhand smoke during childhood due to maternal smoking was associated with increased odds of an endometriosis diagnosis (OR=2.70; 95% CI: 1.11–6.60)17.

Tobacco use encompasses smoking cigarettes, cigars, or pipes, as well as consuming smokeless tobacco products. The harmful effects of tobacco on human health are well documented, particularly its association with cardiovascular diseases19, respiratory conditions, and various cancers20. However, the impact of tobacco on gynecological disorders like endometriosis is less widely known.

Molecular pathways involving tobacco smoking and leading to endometriosis

The association between tobacco smoking and endometriosis involves complex molecular pathways that contribute to the pathogenesis of the disease. Here, we provide detailed insights into the molecular pathways that connect tobacco smoking and endometriosis (Figure 1).

Figure 1

Molecular and cellular mechanisms involved in smoking-induced endometriosis

https://www.tobaccoinduceddiseases.org/f/fulltexts/203429/TID-23-85-g001_min.jpg

Inflammation

Tobacco smoke contains a wide array of toxic compounds that can initiate and sustain an inflammatory response in the body21-23 (Table 1). These compounds include nicotine, carbon monoxide, polycyclic aromatic hydrocarbons (PAHs), and volatile organic compounds (VOCs)24-26. When inhaled, these substances can directly activate immune cells and stimulate the release of pro-inflammatory cytokines, chemokines, and growth factors21.

Table 1

Characteristics of the original articles selected for the review

Molecular pathwayRef.YearObjectiveStudy typeTypeHuman/animalSampleExperimental techniqueFindingsMarkers
Inflammation[23]2006To investigate the molecular mechanisms of inflammatory responses caused by cigarette smoke extractIn vitroCell line studyHuman monocytic cell line (mature monocytes) MonoMac6NRWestern blotting, immuno-precipitation and posttranslational modifications; Electrophoretic Mobility Shift Assay; ELISA for IL-8 and TNF-alphaCigarette smoke-induced release of IL-8 is associated with activation of NF-B via IKK and reduction in HDAC levels/activity in macrophagesIL-8 and TNF-alpha, histone deacetylase (HDAC) activity, HDAC1, HDAC2, and HDAC3 protein levels
[24]2005To provide new data for ‘tar’ and nicotine using an updatedObservationalHuman studyVolunteer smokers52Calibrated electrochemical CO analyzer (Monoxor II, Bacharach Inc.)Increased quantities of PAH and CO among smokersPAHs and CO
[25]2020To determine and describe groups with distinct exposure profilesObservationalPopulation-basedHuman6724Concentrations of a set of urinary tobacco biomarkersHeterogeneity in urinary biomarkers of exposure to nicotine, TSNAs, VOCs, and PAHsExposure to nicotine, TSNAs, VOCs, and PAH
[32]2008To investigate the effects of tobacco smoke on apoptosis induction and NF-κB signaling modulation with the goal of understanding tobacco smoke- associated disease pathogenesisAnimalRat modelRatsNRWestern blot analyses; Electrophoretic mobility shift assay (EMSA)Tobacco smoke resulted in inhibition of NF-κB activity, noted by suppression of inhibitor of κB (IκB) kinase (IKK), accumulation of IκBα, decrease of NF-κB DNA binding activity, and downregulation of NF-κB - dependent anti-apoptotic proteins, including Bcl-2, Bcl-xl, and inhibitors of apoptosisNF-κB p65, p50, IκBα, IκBβ, HSP70, Bcl-2, Bcl-xl, c-IAP1, c-IAP2, XIAP, p53, Bax, caspase 8, caspase 9, caspase 3 and actin
[39]2019To determine that serum chemokines and MMPs will be altered in women with endometriosis compared to women without diseaseObservationalCase-control41Multiplex cytokine immunoassayChemokines (CCL1, CCL22, and CCL11) and cytokine (IL-10) are elevated in endometriosis casesChemokines and cytokines
[40]2018To model the inflammatory microenvironment in endometriotic lesionsIn vitroMonocyte cell modelTHP-1 human monocyte cell line (TIB-202)NRQuantitative real-time PCR analysisNiclosamide inhibits macrophage-dependent endometriotic epithelial cell viability and production of cytokines and chemokines in treated cells through STAT3 and/or NFKB signalingNFKB and STAT3
[43]2018To develop an in vitro screening panel to identify whether flavorings added to tobacco productsIn vitroEndothelial cellsFreshly isolated endothelial cellsNRTUNEL assay (terminal deoxynucleotidyl transferase dUTP nick-end labeling; Roche)Short-term exposure of endothelial cells to flavoring compounds used in tobacco products have adverse effects on endothelial cell phenotype
[46]2001To identify NtMEK2, a tobacco MAPKK. as an upstream kinase for both SIPK and WIPKAnimalRabbit modelRabbit cellsNRAntibody preparation and immunoblot analysisMAPK cascade controls multiple defense responses against pathogen invasionMAPK kinase, salicylic acid-induced protein kinase (SIPK) and wounding-induced protein kinase (WIPK)
[47]1998To demonstrate that ungual cell wall-derived elicitor can activate SIPK in tobacco plantsAnimalFungal cell modelFungal cell wall elicitorNRImmunoprecipitation, immunoblot analysis, and immune-complex kinase assaySIPK is involved in both disease resistance and response to woundingSIPK (SA-induced protein kinase)
Oxidative stress[51]2009To investigate oxidative and carcinogenic mechanisms of tobacco and synergistic action with other respirable particles in the respiratory system of smokersObservationalEnvironmental exposureComponents of cigarettesNRElectron Paramagnetic Resonance (EPR) and spin-trapping techniquesSynergistic effects in the generation of HO•, through the Fenton reaction, with environmental respirable particles (asbestos fibers, coal dust, etc.)Superoxide anion (O2•) and hydroxyl (HO•) radicals
[54]2021To determine the total antioxidant capacity, total oxidant status and oxidative stress index levels in the serum of active smokers, passive smokers and non-smokersObservationalBiomarker-basedHumans150Spectro-photometric method using Rel Assay Diagnostics kitOS levels in serum samples were significantly lower in non-smokers than smoker and past smoker groupsAntioxidants and total oxidant status (TOS)
[55]2016To determine whether cigarette smoking affects (anti)oxidant statusObservationalPopulation-basedHumans300NASmoking as a risk factor for CAD is closely associated with increased oxidative stress, and the number of cigarettes smoked plays an important role in increasing the level of oxidative damage and reducing antioxidant defenseConcentration of oxidants (MDA and HP)
[58]2007To determine the independent and combined impact of dietary intake and cigarette smoking on blood antioxidant capacity and oxidative stressObservationalCohort studyA sample of young smokers28ELISA procedure (Alpco Diagnostics, Salem, NH)Cigarette smoking, particularly the number of years participating in this activity, may manifest in impaired antioxidant capacity and elevated oxidative stress independent of dietary intakePlasma antioxidant reducing capacity (ARC; expressed in uric acid equivalents), serum trolox-equivalent antioxidant capacity (TEAC), whole blood total glutathione, plasma malondialdehyde (MDA), and plasma oxidized low density lipoprotein (oxLDL)
[63]2018Whether the scavenging of mitochondrial H2O2 in transgenic mice expressing mitochondria-targeted catalase (mCAT) attenuates the development of cigarette smoke/angiotensin II-induced mitochondrial oxidative stress and hypertensionAnimalMouse modelTransgenic mice expressing mitochondria-targeted catalase (mCAT) compared to wild-type miceNRWestern blot experimentTobacco smoke and angiotensin II reduce the mitochondrial deacetylase sirtuin-3 level and cause hyperacetylation of a key mitochondrial antioxidant, SOD2, which promotes mitochondrial oxidative stressSOD2
[64]2020To investigate the regulatory mechanism underlying CS-induced hypoxia-inducible factor (HIF)-1α activationIn vitroHuman endothelial cellsPrimary human endometrial stromal cells and an immortalized cell line (KC02-44D)NRWestern blot analysisCS extract (CSE) increased reactive oxygen species levels and stimulated HIF-1α protein stabilization in endometrial stromal cellsHI1-alpha expression
[72]2015To assess the extent of oxidative damage induced by long-term cigarette smoke exposureAnimalRat modelWistar ratsNRMeasurement of 8-OHdG in urine, lymphocytes, and lung tissueLong-term cigarette smoke exposure can cause obvious damages of lung tissue in ratsLevels of ROS, 8-OHdG, and total antioxidant (T-AOC), expression of DNA repair enzymes, e.g. 8-oxyguaine DNA glycosylase (OGG1), and MutThomolog 1 (Oxidized Purine Nucleoside Triphosphatase, MTH1)
[73]2019To clarify the direct effects of nicotine administration on the antioxidant defense system and lipid peroxidationIn vitroHuman endometrial cellsHuman endometrial stromal primary cellNRProcedures of Fecondo and AugusteynNicotine as a pro-oxidant affects the oxidative state of the endometrial cellsGlutathione (GSH) level, glutathione peroxidase (GPx), glutathione reductase (GR), and catalase (CAT) enzymes activity and higher levels of malondialdehyde (MDA)
[75]2021To compare DNA damage marker localization, expression of DDR genes and expression of DNA repair genes in ectopic endometrial samplesObservationalCase-controlWomen with and without endo-metriosis66RT2 Profiler PCR arraysAlterations in the expression of DDR and DNA repair genes indirectly suggest that ectopic endometrium, as compared to its healthy counterpart, encounters DNA damage-inducing stimuli, either of higher strength or for longer duration in endometriosisDNA damage response
[76]2018Examined expression levels of genes pertaining to DNA DSB repair in patients with endometriosis to assess the potential effects on ovarian reservesObservationalCase-controlWomen with endo-metriosis69ImmunohistochemistryExpression of γ-H2AX in immunoassayed endometrial and ovarian tissue preparations was greater in the endometriosis groupDNA damage event
Hormonal dysregulation[85]2011To investigate the relationship between cigarette smoking habits and endogenous sex hormone levels in postmenopausal womenObservationalHormone analysis in post-menopause womenPost-menopausal women2030Non-fasting blood samples analysesCigarette smoking is associated with higher circulating levels of androgens, estrogens, 17-hydroxprogesterone, and SHBGAndrogens, estrogens, 17-hydroxprogesterone, and SHBG
[88]2018To investigate the effect of nicotine on serum progesterone and estradiol levels as possible cause of abortion during first trimester of gestationAnimalRat modelFemale Wistar rats14Enzyme-based immunoassay systemSignificant decrease in serum progesterone and estradiol levels in the nicotine-treated group when compared to controlsSerum progesterone and estradiol levels
[92]2008To test the hypothesis that cigarette smoking is associated with hot flushes through a mechanism involving androgen levels, progesterone levels, sex hormone binding globulin levels, or the ratio of androgens to estrogensObservationalSelf-reported hormonal dataHumans628Enzyme-linked immunosorbent assays (ELISA)Cigarette smoking is associated with hot flushes through a mechanism that may not involve alterations in hormone levels or their ratiosAndrogen and andro-stenedione levels
[94]2016To investigate the effect of the non-aromatizable androgen dihydrotestosterone (DHT)In vitroEpithelial cell cultureEpithelial cellsNRReal-time PCRSignificant DHT-dependent changes in the concentrations of mRNAs encoded by genes implicated in the regulation of the cell cycleNon-aromatizable androgen dihydrotestosterone (DHT)
[96]2020The potential sex steroid signal disrupting mechanisms of nicotine and cotinineComputationalMolecular docking analysisPubChem compound databaseNRPubChem compound databaseStructural binding interactions of the tobacco alkaloid nicotine and its major metabolite cotinine with the sex-steroid nuclear receptors (nicotine and cotinine bind and interact with sex-steroid nuclear receptors and have potential to interfere in steroid hormone signaling resulting in reproductive dysfunction)Estrogen receptor-α (ERα), ERβ, androgen receptor (AR), and progesterone receptor (PR)
[99]2002To examine the circulating concentrations of IGF-1, IGFBP-3, and soluble ICAM-1ObservationalHormonal studyHumans20 smokers and 20 nonsmokersELISA (sICAM-1 Parameter Immunoassay, R&D Systems, Minneapolis, MN)Soluble ICAM-1 concentrations were significantly increased in smokers, compared to non-smokersICAM-1; IGF-1; IGFBP-3
[101]2004To examine whether nicotine inhibits the pulsatile gonadotropin-releasing hormone (GnRH) release, and whether this inhibition of GnRH release by nicotine is mediated by the GABA receptor systemAnimalRat modelWistar strain ratsNRRoller tube cultureNicotine stimulates GABA release, which then inhibits GnRH release through GABAA receptor systemGnRH, GABA
[102]1975To evaluate the diagnostic and prognostic usefulness of the GnRH test, gonadotropin responses to iv GnRHObservationalHumanPatients82Radio-immunoassayGnRH reflect the readily releasable amount of LH which seems to correlate with previous exposure to endogenous GnRHLH and FSH
DNA damage[108]2003To evaluate whether mutagen sensitivity can predict the risk of endometriosis developmentObservationalGenetic analysisPatients65 subjects and 46 control groupCytogenetic analysisSensitivity to bleomycin-induced chromatid breaks in lymphocytes is associated with the risk of endometriosis developmentMutagen sensitivity of peripheral lymphocyte
[110]2008To examine the levels and types of ROS that are produced in response to DNA damageAnimalRat modelIsogenic S. cerevisiae strainsNRDNA damage-induced increase in intracellular ROS levels is a generalized stress response that is likely to function in various signaling pathwaysROS induced by DNA damage
[116]2020To evaluate the direct effect of nicotine on the epigenome profilingIn vitroEpigenetic studyHuman endometrial stromal cells (HESC)NRImmunocytochemistry stainingNicotine treatments reduced the average level of DNMTs gene expressionGenomic DNA methylation status and DNA methyl-transferases (DNMTs) gene expression
[117]2019To investigate the impact of smoking on lung cells collected from bronchoalveolar lavage (BAL)ObservationalGenetic analysis in lung cellsBroncho-alveolar lavage samples from healthy volunteer49RNA sequencingTobacco smoke exposure epigenetically modifies BAL cells, possibly involving a continuous active demethylation and subsequent increased activity of inflammatory processes in the lungsDNA methylation
[118]2014To investigate if tobacco exposure can cause site-specific posttranslational histone modifications (PTMs)In vitroHistone modification studyMouse and human bronchial epithelial cells (H292)NRBottom-up mass spectrometry approachHistone marks may play an important role in epigenetic state during the pathogenesis of smoking-induced chronic lung diseasesHistone H3 and histone H4
Immune dysfunction[122]2014To test cigarette smoke extract on ovulation, oocyte morphology and ovarian gene expression associated with inhibition of oxidative stressAnimalMouse ovarian studyC57BL/6 miceNRMice in the experimental group were administered a cigarette smoke extract (CSE) solution (2 mg/mL) orally daily, while the blank control group was given dimethylsulfoxide (DMSO). RNA extraction from ovariesCSE group manifested a reduced diameter of zona pellucidafree oocyte (ZP-free OD) and a morphologically misshapen first polar body (PB)Oocyte morphology and ovarian gene expression associated with inhibition of oxidative stress
[123]2003To test the immunoregulatory effects of nicotineIn vitroImmune cell studyDendritic cells (DCs)NRELISA kitsNicotine can exert its immunosuppressive effects on immune surveillance through functional impairment of the DC systemCytokines
[124]2020To test the effects of smoking on inflammatory markers, innate and adaptive immune responses to bacterial and viral challenges and blood cell compositionObservationalImmune biomarker analysisPlasma samples from heavy smokers30Luminex analysis and immunophenotypingSmokers had lower NK cells and higher Tregs than controls, suggesting that smoking may reduce the ability to kill nascent tumor cellsCRP, fibrinogen, IL-6 and CEA levels
[127]2020To evaluate the relationship between NK cell activity and urinary cotinine levelObservationalNK cell function analysisPlasma by NK cells12249ELISANK cell activity was lower in current smokersNK cell activity (IFN-gamma)
[128]2020To detect the involvement of immune cells in the pathogenesis of endometriosis in patients with stable status or pelvic painObservationalImmune gene expression studyBlood was collected from patients with endo-metriosisNRFlow cytometrySAMD9 and RGL2 expression levels were significantly upregulated in patients with pelvic painSAMD9 and RGL2 expression levels
[133]2017Do cell adhesion molecules play a role in endometriosis, and can they be used as a biomarker for diagnosing endometriosis?ObservationalCell adhesion molecules in serumSerum of women138Quantitative real-time PCRThe mRNA levels of both VCAM-1 and ICAM-1 were higher in ectopic endometriotic lesions than in ectopic endometriumVascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1)
[135]2012To mimic, in vitro, the long-term exposure of human lung epithelium to smokeIn vitroLung epithelium modelHuman lung adenocarcinoma cells (A549)NRImmunohistochemistryExpression of Smad3 is lower in lung tumors of current smokers compared to that observed in never-smokersSmad 3
[145]2006To elucidate the role of angiogenic factors, we investigated in vivo whether blockade of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), and platelet-derived growth factor (PDGF) affects angiogenesis of ectopic endometriumAnimalHamster angiogenesis studySyrian golden hamstersNRHistology and immunohistochemistryVascularization of endometriotic lesions is not solely driven by VEGF, but depends on the crosstalk between VEGF, FGF and PDGFVEGF, FGF and PDGF inhibitor SU6668
[149]2016To elucidate pathophysiological processes in vitro and in vivo effects of tobacco extract on the transcription factor, hypoxia-inducible factor 1 (HIF-1)In vitroHypoxia transcription factor studyA549 and BEAS-2B cellsNRImmunoblot assaysCSE and CS induced HIF-1 activation in vitro and in vivoHIF1-alpha expression
[151]2015To investigate the expression of HIF-1a, HIF-2a, VEGF-A, PAR-1, and PAR-4 mRNA in lesions from patients with ovarian endometrioma (OMA) and deep infiltrating endometriosis (DIE)ObservationalHIF1-alpha and VEGF analysisOvarian endometrioma (OMA; n 1⁄4 16) or deep infiltrating endo-metriosis (DIE; n 1⁄4 11)NRImmunoblot assaysOvarian endometrioma expresses high levels of HIF-1/2a, PAR-1/4, and VEGF-A. A positive correlation between the expression of HIF-1/2a and VEGF-A mRNA was observed in OMAHIF-1a, HIF-2a, VEGF-A, PAR-1, and PAR-4 mRNA
[152]2017To investigate whether autophagy was regulated by HIF-1α, as well as whether the effect of HIF-1α on cell migration and invasion is mediated through autophagy upregulationObservationalHumans autophagy and invasion studyHuman endometrial stromal cells (HESCs)NRImmunohistochemistryHIF-1α promotes HESCs invasion and metastasis by upregulating autophagyHIF-1α
[154]2014To evaluate effects on remodeling and hyperreactivity face to tobacco exposeIn vitroAirway smooth muscle cell studyCanalicular-stage (18–20 wk gestational age) human fetal airway smooth muscle (fASM) cellsNRWestern blot analysisThese results demonstrate that cigarette smoke may enhance remodeling in developing human ASM through hyperplasia and ECM productionSignal-related kinase (ERK) and p38

[i] Ref.: reference. NR: not reported.

Tobacco smoke components can activate immune cells in the pelvic cavity, including macrophages, neutrophils, and lymphocytes27. Activation of these immune cells triggers the secretion of proinflammatory mediators, such as interleukin 1beta (IL-1β), IL-6, IL-8, and tumor necrosis factor alpha (TNF-α)28,29. These cytokines play crucial roles in promoting inflammation, recruiting immune cells to the site of inflammation, and stimulating tissue remodeling processes30.

The nuclear factor-kappa B (NF-κB) pathway is a central regulator of inflammation31. The components in tobacco can activate the NF-κB pathway32, leading to the transcriptional upregulation of various pro-inflammatory genes33. NF-κB promotes the expression of cytokines, chemokines, adhesion molecules, and enzymes involved in the inflammatory response34,35. This sustained activation of NF-κB perpetuates the inflammatory environment in endometriosis36,37.

Tobacco smoke can stimulate the production of chemokines, such as IL-8 and monocyte chemoattractant protein-1 (MCP-1)21,38. These chemokines attract leucocytes, including neutrophils and macrophages, to endometriotic lesions39-41. The recruited immune cells contribute to the local inflammatory response and produce additional pro-inflammatory mediators, amplifying the inflammatory cascade42.

Tobacco can induce vascular permeability, leading to the leakage of plasma proteins and immune cells into the surrounding tissues43. This increased vascular permeability facilitates the infiltration of inflammatory cells into endometriotic lesions, exacerbating the inflammatory response44. Moreover, leakage of plasma proteins can further contribute to tissue inflammation and promote angiogenesis45.

Various signaling pathways involved in inflammation can be induced by tobacco, including the mitogen-activated protein kinase (MAPK) pathway46,47 and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway48. These pathways regulate the expression of pro-inflammatory genes and modulate immune cell function49. Activation of these pathways by tobacco smoke could contribute to the sustained inflammatory state in endometriosis50.

Thus, the inflammatory response could be triggered by tobacco smoke can lead to a dysregulated cytokine network in endometriosis. Cytokines, such as IL-1β, TNF-α, and IL-6, can induce the production of other inflammatory mediators and promote the activation of immune cells29. This cytokine crosstalk further amplifies the inflammatory cascade, perpetuating the chronic inflammatory environment in endometriotic lesions.

Oxidative stress

Tobacco contains a variety of toxic chemicals and free radicals that can generate reactive oxygen species (ROS) when inhaled51,52 (Table 1). ROS, such as the superoxide anion (O2•-), hydrogen peroxide (H2O2), and the hydroxyl radical (OH•), are highly reactive molecules that can cause oxidative damage to cellular components, including lipids, proteins, and DNA53.

Oxidative stress induced by tobacco smoke overwhelms the body’s antioxidant defense mechanisms52,54,55. Antioxidants, such as glutathione, superoxide dismutase (SOD), and catalase, neutralize ROS and protect cells from oxidative damage56. However, tobacco smoke can deplete these antioxidants and impair their ability to counteract the excessive ROS production, leading to an imbalance between oxidative stress and the antioxidant capacity57-59.

ROS generated by tobacco can initiate lipid peroxidation, a process that damages cell membranes and disrupts their integrity52. Lipid peroxidation products, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), can induce inflammation, impair cellular functions, and contribute to tissue damage60. In endometriosis, lipid peroxidation can affect the viability and function of endometrial cells, exacerbating the disease61,62.

Tobacco smoke-induced oxidative stress can impair mitochondrial function, including endometrial cells63,64. Mitochondria are a major source of ROS production, and their dysfunction can lead to increased ROS generation65,66. The compounds in tobacco can directly target mitochondria, disrupting their electron transport chain and impairing adenosine triphosphate (ATP) production67,68. Mitochondrial dysfunction further exacerbates oxidative stress69,70, perpetuating the cycle of oxidative damage and inflammation in endometriosis71.

ROS generated by tobacco can directly damage DNA in endometrial cells72,73. This DNA damage includes DNA strand breaks, base modifications, and DNA adduct formation74. Accumulated DNA damage can lead to genetic instability, mutations, and chromosomal aberrations in endometriotic lesions75,76. The compromised DNA repair mechanisms in endometriosis may exacerbate the impact of tobacco smoke-induced DNA damage on disease progression77.

Oxidative stress can activate various inflammatory signaling pathways in endometriosis61,78,79. ROS can stimulate the NF-κB pathway, leading to the production of pro-inflammatory cytokines and chemokines80. This activation of inflammatory pathways further amplifies the inflammatory response and contributes to the pathogenesis of endometriosis81,82.

Tobacco smoke-induced oxidative stress can result in the oxidation and modification of proteins and enzymes involved in cellular functions52,57. Oxidative modifications can disrupt protein structure and impair enzyme activity, changes that affect essential cellular processes83. In endometriosis, oxidative stress can target proteins and enzymes involved in inflammation, hormone signaling, and tissue remodeling, further contributing to disease progression84.

Hormonal dysregulation

Tobacco has been associated with alterations in estrogen levels, which play a crucial role in the development and maintenance of endometriosis7,15,85 (Table 1). Smoking can decrease circulating estrogen levels by accelerating the metabolism and clearance of estrogen from the body86. This estrogen imbalance can disrupt the normal endocrine environment, potentially promoting the growth and survival of endometrial tissue outside the uterus87.

A decrease in progesterone levels has been linked by tobacco88,89. Progesterone is an important hormone that helps regulate the menstrual cycle and maintain the endometrium87. A decrease in progesterone levels may disrupt the balance between estrogen and progesterone, promoting the growth and proliferation of endometriotic lesions90.

Androgen hormone levels can be modulated by tobacco91. Smoking has been associated with increased androgen production and alterations in androgen metabolism92. These changes in androgen levels can affect the growth and survival of endometrial tissue outside the uterus93,94. Androgens, such as testosterone, can stimulate the growth of endometriotic lesions and contribute to the pathogenesis of endometriosis95.

Tobacco contains numerous chemicals that can interact with hormone receptors, including estrogen receptors (ERs) and progesterone receptors (PRs)96. These interactions can disrupt the normal signaling pathways regulated by these receptors. Altered receptor activation and signaling can affect gene expression patterns, leading to dysregulation of key genes involved in inflammation, cell proliferation, and tissue remodeling in endometriosis97.

The compounds in tobacco can interfere with hormone-related signaling pathways involved in endometriosis. For example, smoking has been shown to modulate the insulin-like growth factor (IGF) signaling pathway, which plays a role in cell growth and survival98,99. Dysregulation of hormone-related signaling pathways can contribute to the aberrant growth and survival of endometrial tissue in endometriosis100.

Tobacco can affect the hypothalamic-pituitary-gonadal axis by influencing the secretion and function of gonadotropin-releasing hormone (GnRH)98,101. GnRH is a key hormone that regulates the production of luteinizing hormone (LH) and follicle-stimulating hormone (FSH)102. Disruption of GnRH signaling by smoking can lead to imbalances in LH and FSH levels, which can impact ovarian function and the menstrual cycle, potentially contributing to endometriosis development and progression98.

Epigenetic modifications were associated with tobacco, including DNA methylation and histone modifications103,104. Epigenetic changes can alter gene expression patterns without altering the DNA sequence itself105. Smoking-induced epigenetic modifications can affect the expression of genes involved in hormonal regulation and contribute to hormonal dysregulation in endometriosis104.

DNA damage and epigenetic modifications

Tobacco contains numerous harmful chemicals that can directly damage DNA106 (Table 1). These chemicals, such as PAHs, aromatic amines, and nitrosamines, can form DNA adducts and induce DNA strand breaks107. The DNA damage caused by tobacco smoke can lead to genetic alterations and chromosomal abnormalities in endometrial cells, potentially promoting the development and progression of endometriosis8,108.

The ROS generated by tobacco smoking can also cause oxidative damage to DNA51. ROS can react with DNA bases, leading to the formation of DNA adducts and base modifications109. Additionally, ROS can induce DNA strand breaks and impair DNA repair mechanisms110. The accumulation of oxidative DNA damage in endometrial cells can contribute to genomic instability and the pathogenesis of endometriosis111.

Tobacco smoking can interfere with DNA repair mechanisms in endometrial cells. The chemicals present in tobacco smoke can inhibit DNA repair enzymes, such as DNA polymerases and DNA repair proteins112. This impaired DNA repair capacity can lead to the persistence of DNA damage and genomic instability in endometriotic lesions, promoting disease progression113.

Epigenetic modifications refer to heritable changes in gene expression patterns without altering the DNA sequence itself114. Tobacco smoking has been associated with epigenetic modifications, including DNA methylation and histone modifications103,104. Smoking-induced epigenetic changes can alter the expression of genes involved in cellular processes such as inflammation, cell proliferation, and hormone signaling115. Thus, these modifications can contribute to the dysregulation of gene expression in endometrial cells and the pathogenesis of endometriosis.

Aberrant DNA methylation patterns in endometrial cells have been induced by tobacco smoking 116. DNA methylation is a common epigenetic modification that involves the addition of a methyl group to DNA molecules, typically leading to gene silencing116. Smoking-induced DNA methylation changes can affect the expression of genes involved in hormone metabolism, inflammation, and tissue remodeling, potentially promoting the development and progression of endometriosis104,117.

Tobacco smoking can also influence histone modifications, which regulate the accessibility of DNA to transcription factors and other proteins involved in gene expression118. Smoking-induced histone modifications can alter the structure of chromatin and affect the expression of genes implicated in endometriosis104,119. These modifications can lead to dysregulated gene expression patterns and contribute to the molecular and cellular changes associated with the disease.

Tobacco smoking-induced DNA damage and epigenetic modifications can potentially have transgenerational effects on offspring120. Smoking-related alterations in sperm and egg cells can lead to inherited epigenetic changes that may influence the susceptibility to endometriosis in future generations121,122. These transgenerational effects highlight the long-lasting impact of tobacco smoking on the molecular pathways involved in endometriosis.

Immune dysfunction

Tobacco can modulate the immune system, leading to dysregulation of immune cells and molecules involved in the pathogenesis of endometriosis27 (Table 1). Smoking can suppress the activity of immune cells, such as natural killer (NK) cells, macrophages, and T cells, reducing their ability to eliminate endometrial cells outside the uterus8,123. This impaired immune response allows the survival and proliferation of ectopic endometrial tissue, contributing to the development of endometriosis.

Tobacco may disrupt the production and balance of cytokines, which are important immune signaling molecules21. Smoking has been associated with increased production of pro-inflammatory cytokines, such as IL-6 and TNF-α, and decreased production of anti-inflammatory cytokines, such as IL-10124. This imbalance in cytokine production can contribute to chronic inflammation and tissue damage in endometriosis125.

A chronic inflammatory state in the body is induced by tobacco smoking, characterized by elevated levels of inflammatory markers and immune cells124. Smoking-related inflammation can promote the recruitment of immune cells to endometriotic lesions and exacerbate tissue inflammation22. This persistent inflammatory response can contribute to the growth, invasion, and persistence of endometriotic lesions126.

Tobacco smoking can affect the function of immune cells involved in endometriosis. For example, smoking can impair the cytotoxic activity of NK cells, which play a crucial role in eliminating abnormal cells, including endometrial cells127,128. Smoking-related alterations in immune cell function can compromise the surveillance and clearance of endometrial cells outside the uterus, contributing to the establishment and progression of endometriosis129.

Immune tolerance refers to the ability of the immune system to recognize and tolerate selft-issues130. In endometriosis, there is a breakdown in immune tolerance, allowing ectopic endometrial tissue to survive and evade immune surveillance126,131. Tobacco smoking can further disrupt immune tolerance mechanisms, leading to an aberrant immune response against endometrial cells and perpetuating the immune dysregulation observed in endometriosis21,27,104.

The compounds found in tobacco smoke can modulate the expression and function of cellular adhesion molecules involved in immune cell trafficking and tissue inflammation. Smoking-induced alterations in adhesion molecules, such as intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), can recruit immune cells to endometriotic lesions and contribute to the inflammatory process132,133.

Tobacco smoking can impair wound healing and tissue repair processes, which are essential for the resolution of inflammation and the restoration of tissue integrity134. Smoking-related factors can interfere with the production and activity of growth factors, such as transforming growth factor beta (TGF-β), which play a critical role in tissue repair135. Impaired wound healing can perpetuate the inflammatory response and contribute to the persistence and progression of endometriotic lesions136.

Angiogenesis

Angiogenesis refers to the formation of new blood vessels from pre-existing ones137 (Table 1). In endometriosis, angiogenesis plays a crucial role in the establishment and growth of ectopic endometrial tissue138. Tobacco smoking has been linked to increased angiogenesis139-141, which can contribute to the progression and persistence of endometriotic lesions.

Tobacco smoke contains various chemicals that can promote angiogenesis. For example, nicotine, a key component of tobacco, has been shown to stimulate the release of pro-angiogenic factors, such as vascular endothelial growth factor (VEGF)142 and basic fibroblast growth factor (bFGF)143. These factors can enhance the formation of new blood vessels, providing a blood supply to endometriotic lesions and supporting their growth144,145.

In addition to promoting angiogenesis, tobacco smoking can disrupt the balance of angiogenesis inhibitors 141. Endostatin, thrombospondin-1 (TSP-1), and angiostatin are examples of naturally occurring substances that inhibit blood vessel formation146,147. Smoking-related factors could interfere with the production and function of these angiogenesis inhibitors, thus allowing angiogenesis to proceed unchecked in endometriosis.

Inflammatory cells and cytokines present in the endometriotic micro-environment can promote the production of pro-angiogenic factors, which contribute to neovascularization137,148. These newly formed blood vessels provide nutrients and oxygen to endometriotic lesions, facilitating their survival and growth.

Tobacco can induce hypoxic conditions in tissues due to decreased oxygen availability149,150. Hypoxia is a potent stimulator of angiogenesis, as it triggers the release of hypoxia-inducible factors (HIFs)137. These proteins promote the expression of VEGF and other pro-angiogenic factors, facilitating the formation of new blood vessels in the hypoxic environment of endometriotic lesions151,152.

Tobacco smoking can disrupt the remodeling of the extracellular matrix (ECM), which is essential for angiogenesis153. The ECM provides structural support for blood vessels and influences their formation and stability154. Smoking-related factors can affect the production and degradation of ECM components155, leading to an imbalance in ECM remodeling and promoting angiogenesis in endometriosis.

Angiogenesis supports the growth of endometriotic lesions and facilitates their invasion into surrounding tissues. The newly formed blood vessels provide a pathway for the migration of endometrial cells, enabling them to establish new lesions and to expand the disease. Therefore, smoking-induced angiogenesis can contribute to the invasive and metastatic behavior of endometriosis.

Limitations

While this narrative review provides a synthesis of existing literature on the relationship between tobacco smoking and endometriosis, several limitations must be acknowledged. First, most of the included studies rely on observational data, which inherently limits causal inference (Table 2). Although emerging evidence suggests a potential link between smoking and endometriosis, confounding factors such as genetic predisposition, environmental exposures, and lifestyle factors may influence the observed associations. Second, self-reported smoking status, a common data collection method in epidemiological studies, may introduce recall bias and misclassification. Individuals may underreport or overestimate their smoking behavior, leading to potential misinterpretation of results. Additionally, differences in study designs, population characteristics, and exposure definitions contribute to heterogeneity across studies, making direct comparisons challenging.

Table 2. Risk of bias assessment of studies included, based on Newcastle-Ottawa Scale (Nos) for observational studies, Risk of Bias 2 (RoB2) for randomized trials, and SYRCLE’s Risk of Bias tool for animal studies

Ref.Risk of bias
[23]Moderate (in vitro, lacks systemic physiological context)
[24]High (self-reported exposure, potential confounding)
[25]Moderate (biomarker-based, but cross-sectional design)
[32]Moderate (controlled setting, but animal model limitations)
[39]High (small sample size, selection bias)
[40]Moderate (no clinical correlation)
[43]Moderate (limited real-world application)
[46]Moderate (animal model not directly transferable)
[47]Moderate (animal model not directly transferable)
[51]Moderate (experimental setting, lacks direct application to humans)
[54]Moderate (used objective biomarkers, but cross-sectional)
[55]High (no control for confounders, limited sample size)
[58]Moderate (controlled for diet but self-reported smoking)
[63]Moderate (animal model, potential extrapolation issues)
[64]Moderate (cell-based, lacks human validation)
[72]Moderate (measured oxidative stress markers, no human validation)
[73]Moderate (in vitro, lacks physiological relevance)
[75]High (small sample, lacks confounder control)
[76]High (observational, small sample, no randomization)
[85]Moderate (association study, no causal inference)
[88]Moderate (animal model, indirect human relevance)
[92]High (self-reported symptoms, potential recall bias)
[94]Moderate (lacks systemic validation)
[96]Low (computational, needs experimental validation)
[99]Moderate (small cohort, observational limitations)
[101]Moderate (animal model, indirect human relevance)
[102]High (small sample, endocrine condition variability)
[108]High (genetic study, lacks environmental control)
[110]Moderate (oxidative stress analysis, extrapolation issues)
[116]Moderate (epigenetic changes, lacks longitudinal data)
[117]Moderate (DNA methylation, lacks direct causality)
[118]Moderate (histone modifications, lacks human correlation)
[122]Moderate (ovarian morphology, unclear generalizability)
[123]Moderate (immune study, lacks functional validation)
[124]High (observational, lacks control group)
[127]High (NK activity, lacks exposure quantification)
[128]High (immune markers, lacks comprehensive control)
[133]Moderate (cell adhesion markers, lacks validation)
[135]Moderate (lung epithelium, not reproductive model)
[145]Moderate (vascular markers, lacks systemic relevance)
[149]Moderate (oxidative markers, lacks intervention)
[151]Moderate (HIF-1α activation, lacks real-world correlation)
[152]Moderate (autophagy markers, lacks systemic insight)
[154]Moderate (cell model, lacks in vivo validation)

[i] Ref.: reference.

The biological mechanisms linking tobacco smoking to endometriosis remain complex and incompletely understood. While this narrative review highlights several molecular pathways, such as inflammation, oxidative stress, hormonal dysregulation, and epigenetic modifications, causality cannot be definitively established. Further experimental and longitudinal studies are needed to clarify these mechanisms. Finally, while this narrative review provides an overview of the evidence, a systematic review with a comprehensive search strategy, critical appraisal of included studies, and synthesis of findings, would have provided a more conclusive evidence base and hence would be warranted.

CONCLUSION

This review highlights the growing body of evidence linking tobacco smoking to the pathogenesis of endometriosis through multiple biological mechanisms, including chronic inflammation, oxidative stress, hormonal dysregulation, immune dysfunction, and epigenetic modifications. While early studies provided conflicting results, recent large-scale epidemiological data and mechanistic insights suggest that smoking is not only a risk factor for endometriosis but may also exacerbate its severity and progression. The detrimental effects of tobacco on endometrial tissue underscore the broader impact of smoking on women’s reproductive health. This result highlights once again the specific impact of tobacco consumption on women’s health, and adds endometriosis to an already long list (hormone-dependent156, infertility157, cardiovascular pathologies158, for example). Despite these findings, several critical gaps remain. The causality between smoking and endometriosis has yet to be definitively established, necessitating prospective cohort studies with robust control for confounding factors. Future research should also integrate omics approaches, such as transcriptomics, proteomics, and metabolomics, to unravel the molecular pathways underlying the link between tobacco exposure and endometriosis. Additionally, identifying biomarkers of tobacco-induced endometriotic changes could facilitate early diagnosis and risk stratification. From a clinical and public health perspective, these findings reinforce the need for targeted smoking cessation interventions, particularly for women at risk of or diagnosed with endometriosis. Healthcare professionals should incorporate smoking history assessments into routine gynecological care and emphasize the role of smoking in disease progression. Public health policies should also focus on prevention strategies to reduce smoking rates among young women, thereby mitigating a modifiable risk factor for endometriosis and improving reproductive health outcomes.