Next Article in Journal
Carbonate Pore Shape Evaluation Using Digital Image Analysis, Tomography, and Effective Medium Theory
Next Article in Special Issue
Allergic Rhinitis and Asthma: Relationship with Transverse Maxillary Contraction and Transverse Expansion Stability in Children
Previous Article in Journal
A Forecasting Approach to Cryptocurrency Price Index Using Reinforcement Learning
Previous Article in Special Issue
A Preliminary Investigation of Mobile Respiratory Function Testing in Western Australian Communities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sex-Based Differences in Bronchial Asthma: What Are the Mechanisms behind Them?

1
Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
2
Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
*
Author to whom correspondence should be addressed.
Appl. Sci. 2023, 13(4), 2694; https://doi.org/10.3390/app13042694
Submission received: 16 January 2023 / Revised: 10 February 2023 / Accepted: 18 February 2023 / Published: 19 February 2023
(This article belongs to the Special Issue Asthma and Respiratory Disease: Prediction, Diagnosis and Treatment)

Abstract

:
Sex-based differences in bronchial asthma can already be observed in childhood, at which time allergic atopic asthma is more frequently found in boys than in girls. In adulthood, higher prevalence of asthma is reported in women, especially for the more severe neutrophilic subtype associated with obesity, which responds poorly to corticosteroids. Sex-based differences seem to be attributable to changing levels of estrogens, progesterone, and testosterone, which may exert mainly pro-inflammatory (estrogens, progesterone) or anti-inflammatory effects (testosterone). Sex steroids differentially influence lung immune responses, airway reactivity, and pulmonary circulation and may thereby contribute to the higher susceptibility of females to more serious complications resulting from inflammatory lung diseases compared to males. However, other factors, such as anatomical and physiological differences in the lungs, differences in genetically conditioned factors, obesity and lifestyle, smoking, exposure to environmental and occupational factors, chronic stress, etc., may also contribute to the sex-based differences in asthma. Elucidation of the mechanisms behind these differences may contribute to more appropriate personalized therapy for asthma. For the review, articles in the English language from the PubMed database were used.

1. Introduction

Bronchial asthma (or asthma) is one of the most frequently occurring chronic respiratory diseases. Worldwide, asthma affects more than 330 million people of all age groups, and its prevalence is generally higher in high-income countries [1]. Asthma has been recognized as an extremely heterogenic respiratory disorder that presents in the form of several endotypes with distinct underlying pathophysiological mechanisms and various phenotypes with observable clinical characteristics potentially responsible for varying responses to treatment [2,3,4]. The main features of asthma include chronic airway inflammation leading to remodeling of the airways and epithelial fibrosis, hypersecretion of mucus, metaplasia and hyperplasia of goblet cells, and hypertrophy and hyperplasia of airway smooth muscle. Clinical symptoms of asthma include episodes of airway narrowing associated with wheezing, shortness of breath, chest tightness, and as a non-specific symptom, cough, while the intensity of the respiratory signs may be variable. Careful investigation of personal history pointing to relations between the occurrence of respiratory symptoms and exposure to environmental factors, viral infection, allergies, obesity, etc. is very important for making diagnoses [1,3]. In addition to the heterogenic pathophysiological and clinical characteristics of the various subtypes of asthma, sex-based differences have been also described.
Thus, the purpose of this article was to summarize current knowledge on the sex-based differences found in male versus female patients with bronchial asthma, as well as differences demonstrated in various animal models of airway inflammation and hyperreactivity resembling human asthma. For the review, articles in the English language from the PubMed database were used. The collected material highlighted the potential mechanisms responsible for the differences between males and females and gave rise to many questions about how these differences may influence the therapy given, how different the treatments given to women and men with asthma should be, and which preventive measures can be employed to improve the status of asthma patients.

2. Sex- or Gender-Based Differences in Asthma?

For decades, women and female laboratory animals were under-estimated in studies because of greater variability in the physiological parameters. Male subjects dominated in clinical studies, as well in experimental measurements, which could have resulted in misinterpretations and detrimental consequences for females [5,6]. On the other hand, the opposite bias exists for some models of diseases with female predominance (e.g., osteoporosis) where the data collected from females does not completely fit men. Currently, modern trends in biomedical research recommend the recruitment of patients and laboratory animals of both sexes to reliably reproduce the standard situation in populations [5,6].
The terms “sex” and “gender” are often used interchangeably; however, they bear different meanings. While “sex” refers to genetically determined anatomical, physiological, and biological differences between males and females, including the production of hormones, “gender” is more complex and refers to social and cultural differences, including the self-identification of the individual [7].

3. Endotypes and Phenotypes of Bronchial Asthma

In accordance with the dominant type of granulocytic inflammation and the contributions of specific subtypes of T-lymphocytes, three main endotypes of asthma may be distinguished: eosinophilic, non-eosinophilic, and mixed granulocytic asthma [1]. However, the different asthma endotypes and phenotypes are not fully understood, and additional research is needed to adequately define the different asthma endotypes and phenotypes and describe their characteristics.
The eosinophilic (or T2-high) endotype of asthma, which is present in about 50% of adult asthma patients, is divided into allergic and non-allergic subendotypes. The allergic subendotype is characterized by sensitization to allergens (pollen, house-dust mites, etc.). Contact between allergens and alarmins—epithelial barrier-derived mediators such as thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33—activate dendritic cells and group 2 innate lymphoid cells (ILC2s) and induce the type 2 signaling pathway through the mediation of T-helper (Th)2 lymphocytes. Activated dendritic cells and ILC2s are potent producers of IL-4, IL-5, and IL-13, which recruit eosinophils into the lungs, promote goblet cell overexpression, stimulate the secretion of mucus, increase airway hyperresponsiveness, and increase expression of extracellular matrix proteins (e.g., periostin), contributing to airway fibrotic remodeling [3]. IL-4 drives the switching of the B-cell isotype and synthesis of immunoglobulin (Ig)E, which binds to mast cell high-affinity IgE receptors. Subsequent activation of mast cells leads to allergen-mediated IgE cross-linking [1]. In the non-allergic subendotype of eosinophilic asthma, ILC2s produce IL-5 and IL-13 in response to prostaglandin (PG)D2 and the alarmins IL-33, IL-25, and TSLP released from the epithelium due to deterioration resulting from pollutants and microbes [1,2,3].
T2-high asthma clinically presents in three phenotypes that have been identified up to now. Atopic asthma, one subtype of early-onset asthma, is corticosteroid-sensitive and typically appears during childhood because of allergic sensitization. Late-onset asthma is a steroid-resistant phenotype of eosinophilic asthma that develops in older patients as more severe asthma with fixed airflow obstruction. Aspirin-exacerbated respiratory disease (AERD) is a subset of late-onset asthma that originates due to dysregulated arachidonic acid metabolism [3].
The non-eosinophilic (or non-T2 or T2-low) endotype of asthma is characterized by the absence of eosinophilia and the presence of neutrophilic inflammation with neutrophil sputum levels greater than 40% (i.e., neutrophilic asthma) or may manifest as the paucigranulocytic type of asthma with normal sputum levels of eosinophils and neutrophils [3]. Although the mechanisms of neutrophilic asthma are not yet fully understood, chronic infection with atypical bacteria, obesity, and smoking likely contribute to the activation of Th1 and/or Th17 lymphocytes or type 3 ILCs [1,2,3]. Paucigranulocytic asthma is a special endotype of asthma in which both eosinophils and neutrophils are below the thresholds for the diagnosis of the previously mentioned types of asthma. Paucigranulocytic asthma is poorly understood and, instead of inflammation, airway smooth muscle dysfunction and bronchial hyperresponsiveness appear as the main pathophysiological features [2,8].
Several T2-low phenotypes have also been identified. Obesity-associated asthma is a non-atopic and corticosteroid-refractory form of asthma occurring in middle-aged women with obesity. Smoking-associated asthma originates due to smoking exposure-induced oxidative stress. Very late-onset asthma develops in older patients because of decreased lung function due to reduced elastic recoil and immunosenescence [3]. However, non-eosinophilic asthma may also present in children and adolescents as one form of early-onset asthma [9].
In the mixed granulocytic endotype of asthma, both allergic-dependent and allergic-independent mechanisms may occur hand in hand, leading to mixed granulocytic inflammation [1]. The pathophysiological characteristics of the main endotypes of asthma are summarized in Table 1.
Differences in the pathophysiological backgrounds of the major asthma endotypes not only result in different diagnostic criteria for individual types of asthma but may also be responsible for variations in the appropriate treatment [2,10,11,12,13]. For instance, eosinophilic subtypes of asthma usually respond well to corticosteroids [12,14], while both non-eosinophilic subtypes of asthma are insensitive to corticosteroids and, therefore, novel treatments have been intensively studied [13].
A review of clinical and biochemical biomarkers for eosinophilic, neutrophilic, and paucigranulocytic asthma is provided in Table 2, and currently available and promising treatments are listed in Table 3.

4. Sex-Based Differences in Different Life Periods

Sex-based differences in the incidence and severity of asthma occur in all periods of life, as recently demonstrated by Shah and Newcomb [49] and Chowdhury et al. [50] in their excellent reviews. Therefore, only fundamental information necessary for understanding the other parts of this article is provided in the following section.

4.1. Childhood

Sex-based differences in the development of asthma or wheeze in childhood are probably linked to several contributing factors, including prenatal stress, preterm birth, low birth weight, maternal smoking, maternal obesity during pregnancy, no or shorter period of breastfeeding, repetitive respiratory tract infections, use of antibiotics or corticosteroids, and/or genetic susceptibility [51].
Prematurity, low birth weight, and/or prenatal maternal stress have been confirmed as factors increasing the risk of asthma and impaired lung function in early childhood, which may persist up to adulthood [52,53], while reduced expiratory flow and higher vulnerability are more strongly associated with male sex [54]. Greater occurrences of preterm births with worse response to prenatal corticosteroids and worse prognosis in boys than girls [55,56,57] are likely caused by sex hormones. Female neonates exhibit decreased susceptibility to infection and accelerated lung maturation under the stimulatory action of estrogen on the transition from the terminal saccular stage to the alveolar stage and the production of pulmonary surfactant, while testosterone has the opposite effects [58]. Dysanaptic lung development, characterized by retarded growth in the large airways in comparison to the lung parenchyma in boys, leading to narrower airways, is an additional factor resulting in sexual disparity in children [59]. A relation between low birth weight and decreased lung function or risk of asthma later in childhood and/or in adulthood has been demonstrated in several studies [60,61], with some of them finding a higher risk of asthma in males [62,63].
Higher risk of asthma or wheeze during childhood is also associated with other factors [64], such as maternal smoking during pregnancy [65,66,67], maternal obesity in pregnancy [68], maternal use of antibiotics during pregnancy [69], exposure to allergens and endotoxins [70], and low vitamin D status during pregnancy [71]. However, while the lung functions of male fetuses exposed to smoking in utero may be more adversely affected than those of females [72], no convincing sex-specific associations between the other mentioned risk factors and the development of wheeze in children have been found [65,73].
The higher prevalence of asthma—mainly allergic asthma—in boys of prepubertal age is linked to two times more frequent admissions to hospital due to asthma exacerbation [74,75,76,77]. In addition, boys have increased serum immunoglobulin (Ig)E levels and interferon (IFN)-γ levels, and positive skin prick tests for allergens are more frequent [78,79]. In both sexes, higher incidences of asthma were found in overweight or obese children; however, respiratory symptoms of asthma (wheezing, shortness of breath) were more frequently reported in obese boys than in obese girls [80].

4.2. Adolescence

During adolescence, the occurrence of asthma gradually decreases in boys and simultaneously increases in girls. In addition, there is a positive correlation between body mass index (BMI) and the risk of asthma in adolescent girls [81]. The likelihood of the development of asthma symptoms particularly increases in girls who were already overweight or obese between 6 and 11 years of age, but this association was not observed in boys [82].
The BAMSE (Swedish abbreviation for Children, Allergy, Milieu, Stockholm, Epidemiology) study evaluating body mass index (BMI) from birth to adolescence among children with and without asthma found the highest BMIs in girls with persistent asthma, while there were no clear associations between asthma and BMI in boys [83]. The abovementioned studies indicate a more obvious association between pediatric obesity and asthma in boys. In the puberty period, the association between obesity and asthma in girls increases.

4.3. Adulthood

In adulthood, the higher prevalence of asthma in women in comparison to men continues, and the course of the disease in women is more severe and associated with three times more frequent admissions to hospital with asthma complications [84,85]. This sex-related difference remains until menopause [76,77,86,87].
Worsening scores in the pulmonary function tests, more frequent asthmatic symptoms, and higher requirements for medication are observed in about one third of women before and during menstruation [88,89,90]. Pauli et al. [91] found that, in women with asthma, the morning values for peak flows and asthma symptoms deteriorated from the follicular to the luteal phase of the menstruation cycle. These findings were confirmed by other authors, who found the best airflow values at the end of the luteal phase through to the beginning of menstruation, followed by a decline over the subsequent two weeks [92]. However, in healthy controls, no changes in airway functions were observed in relation to fluctuations in estrogen and progesterone during the menstruation cycle [91,92], suggesting more complex mechanisms may be responsible for perimenstrual worsening of asthma status in women [76]. In this context, the recent finding by Eid et al. [93] that 24% of women with AERD also show perimenstrual worsening of asthma could be interesting.
Several studies have investigated the effects of contraceptive pill use in premenopausal women on the changes in lung functions and the occurrence of asthma symptoms. However, the results of these studies were often contradictory. While Macsali et al. [94] demonstrated that normal and overweight women taking hormonal oral contraceptives reported higher occurrences of wheezing and other asthma symptoms in comparison to lean women, other authors found no differences in lung functions or asthma medication use in women taking contraceptives compared to those with a natural menstrual cycle [95] or even found decreased airway responsiveness in women taking contraceptives [96]. This discrepancy indicates that further studies should be performed to evaluate the effects of contraceptives containing gestagen only or combined hormones (gestagen + estrogen) with different doses or ratios of gestagen and estrogen in relation to current asthma status, as well as new onsets of asthma.
Asthma control may change during pregnancy. In an older study by Schatz et al. [97], asthma symptoms increased in about one third of pregnant women while the status reverted within the first three months post-partum in about 73% of them. However, other studies found no change or even improvements in asthma symptoms [98,99]. A more recent analysis by Schatz et al. showed that asthma exacerbations during pregnancy occurred in 12.6% of patients with asthma initially classified as mild, 25.7% of patients with asthma initially classified as moderate, and 51.9% of patients with asthma initially classified as severe, while in 30% of mild asthma patients, the course of the disease worsened to moderate and, vice versa, 23% of patients with moderate–severe asthma showed improvements in their status to mild asthma [100].
The contradictory results of the studies performed with women undergoing menopause may be attributable to fluctuating levels of hormones, the use of hormone replacement therapy (HRT), and the influence of other factors, such as smoking or concomitant diseases [101,102,103,104,105,106,107].
Nevertheless, similarly to the puberty period, a clear relation between asthma prevalence and obesity can be observed in women but not in men, and a higher risk of asthma was reported in obese women than in women with normal body weight [108,109]. Late-onset asthma in obese women, which represents a special phenotype of asthma, is characterized by more severe symptoms, increased counts of neutrophils in sputum, and higher requirements for treatment [110,111].

5. Factors Contributing to Gender-Based Differences in Asthma

5.1. Sex Hormones

Sex hormones seem to be primarily responsible for the differences between females and males in the prevalence and severity of asthma. Nevertheless, the mechanisms responsible for the sex-based differences in asthma may be more complex.
Sex steroids, such as estrogens, progesterone, and testosterone, have been mainly defined through their association with the normal functioning of the reproductive system. However, via their signaling mechanisms, they influence many processes in the body [112] and contribute to varying incidences of several diseases in females and males [113]; e.g., cardiovascular [114,115,116], neurological [117,118,119], metabolic [120,121,122], and respiratory diseases, including asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, lung cancer, and respiratory infections [113]. While androgens mainly exert anti-inflammatory effects, estrogens promote inflammation and may even induce tumorigenesis [123]. Interestingly, women complain more frequently of cough, including dry cough, which is known as an adverse effect associated with the use of angiotensin-converting enzyme (ACE) inhibitors due to the increased excitability of peripheral and central cough pathways in women [124].
Sex steroids are produced in hormone-secreting organs (ovaria, testes, placenta), as well as in other tissues; e.g., adrenal glands and adipose tissue. All sex steroids are synthesized from common precursor dietary cholesterol in the process of steroidogenesis [125] (Figure 1).
Production of sex steroids from other tissues—e.g., adipose tissue, skin fibroblasts, bones, or brain—can contribute significantly to the circulating pool of estrogens [130,131].
However, in addition to signaling intermediates, the action of steroid receptors can be modified by various coregulators, which act as coactivators or corepressors of steroid-responsive gene expression [132]. The coregulators not only regulate hormonal events in pregnancy, sex differentiation, development, reproduction, and sexual behavior but also influence the functions of non-reproductive tissues, such as the heart, kidneys, pancreas, bone, and brain. Mutation and/or aberrant expression of the coregulators affecting the normal function of steroid hormones may result in the development of obesity, diabetes mellitus, atherosclerosis, osteoporosis, cancer, neurodegenerative disorders, or breast cancer [132].

5.1.1. Estrogens

A group of endogenous estrogens containing 18 carbons are known as C18 steroids. While estradiol (or 17-β estradiol) is produced in ovarian follicles and corpora lutea and represents the predominant circulating estrogen during the reproductive period, estriol and estretol are synthesized during pregnancy in the placenta or fetal liver, while estrone is generated by extraglandular tissues during menopause [133].
In cells, estrogens act by binding to specific nuclear estrogen receptors (ERs), ER-α and ER-β. They activate transcriptional processes and/or signaling events that result in the control of gene expression. The actions of estrogens can be mediated through direct binding of ER complexes to specific sequences in gene promoters (genomic effects) or through mechanisms that do not comprise direct binding to DNA (non-genomic effects) [134].
In genomic signaling, after binding of estrogen to ER-α or ER-β, the complex translocates to the nucleus, inducing transcriptional changes in estrogen-responsive genes with or without DNA estrogen-response elements [135]. In indirect genomic signaling, estrogen receptor complexes exert their actions via protein–protein interactions with other transcription factors and response elements [136]. Non-genomic effects are mediated via membrane-associated estrogen receptors and contribute to modulation of intracellular Ca2+ and activation of signal-transduction mechanisms, with subsequent production of intracellular second messengers, regulation of cyclic adenosine monophosphate (cAMP), and protein-kinase activation of signaling cascades, leading to rapid biological effects [137]. In addition, as an ER-independent action, estrogen may exert antioxidant effects, and as estrogen-independent actions, ligand-independent genomic events may occur [134]. Nevertheless, the actions of estrogens in the various cells are likely mediated by a combination of processes comprising genomic and non-genomic pathways, as well as additional convergent pathways enhancing transcriptional activity in specific tissues and physiological processes [138,139].
Estrogens exert a broad spectrum of effects, including induction of primary and secondary sexual characteristics, regulation of the menstrual cycle and reproduction, effects on bone density, effects on brain function (including cognitive processes and memory), and cholesterol mobilization [140]. Moreover, estrogens have many direct biological effects in the lungs, where significant distributions of ER-α and ER-β—with more abundant ER-β—have been demonstrated [141]. ER-β acts as an inhibitor of ER-α-mediated activity and may reduce the cell response to estrogens [142].
In the lungs, twice as much ER-β as ER-α is expressed in human bronchial epithelial cells, regulating epithelial Ca2+ signaling [143,144]. Estradiol stimulates the production of nitric oxide (NO) from bronchial epithelial cells and increases the activation of endothelial NO synthase (eNOS), contributing to bronchodilation, largely via ER-α [145]. In addition, estradiol may modulate epithelial-to-mesenchymal transition and barrier function in bronchial epithelial cells [144,146]. Estrogens also increase the number of goblet cells and the production of mucus, mainly via ER-β [147]. Although data from animal studies are not consistent, they suggest that estrogens may have a protective effect on airways, modulating airway responsiveness via activation of the NO-cyclic guanosine monophosphate (cGMP) pathway or by decreasing Ca2+ influx [148,149,150,151]. However, estrogens stimulate airway inflammation, as confirmed in animal studies [152,153,154]. Estrogens enhance the differentiation of dendritic cells [155] and elevate counts of ILC2s [156], eosinophils [152,154,157], and IFN-γ-producing Th1 cells [158] more obviously in females.

5.1.2. Progesterone

Progesterone action is mediated via two isoforms of the progesterone receptor (PR), PR-A and PR-B [159], leading to interaction with progesterone-response elements and ensuing regulation of target genes [160]. However, PRs can also interact with regulatory proteins and exert actions via non-genomic mechanisms [161].
PRs are distributed in many tissues, including the lungs. In addition to reproductive functions, progesterone participates in the regulation of neurosteroid activity in the central nervous system, inhibition of smooth muscle contractile activity in the gastrointestinal tract, vasodilation, and regulation of the development and maturation of the lungs [162].
Although there are fewer studies evaluating the effects of progesterone in asthma or its models, the available results indicate a mild pro-inflammatory effect as a consequence of increased IL-4 and IL-5 levels, eosinophil counts, and airway hyperreactivity [163,164]. In human bronchial epithelial cells, progesterone was found to decrease ciliary beat frequency, leading to inhibition of the mucociliary apparatus; however, this effect was prevented by co-administration of estradiol and progesterone [165].

5.1.3. Testosterone

The effects of testosterone and its highly active metabolite 5α-dihydrotestosterone (DHT) are mediated via androgen receptors (ARs), leading to binding to androgen-responsive elements and downstream upregulation or downregulation of the target genes [166]. Similarly to ERs, ARs are distributed in many tissues, including the lungs, in which high concentrations of ARs have been detected in the alveolar type II cells and bronchial epithelium [167]. Androgens show potent anti-inflammatory action. They reduced populations of Th2 and ILC2 cells, which subsequently led to reduced eosinophilic inflammation, but also decreased Th17-mediated production of IL-17A and reduced neutrophilic inflammation [156,168]. Via suppression of IL-17A, testosterone may reduce Ca2+ sensitization and airway smooth muscle contractions [169]. However, in a murine model, it was demonstrated that male sex hormones may promote airway responsiveness to cholinergic stimulation via vagal nerve-mediated reflex mechanisms [170].
Lower levels of testosterone may be associated with higher risk of asthma not only in women but also in men older than 50 years [86]. Bulkhi et al. found that serum testosterone was inversely associated with current asthma prevalence regardless of sex, and this value correlated with better lung function [171]. In accordance with these results, higher expression of bronchial ARs and higher androgen levels were associated with better lung function, fewer symptoms, and a lower FeNO in human asthma [172], while lower expression of ARs in the airways was linked to more frequent occurrence of asthma in both sexes [173]. Decreased values for the testosterone precursors dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEAS) were also found in postmenopausal asthmatic women compared to non-asthmatic subjects [174].
The therapeutic effect of testosterone on neutrophilic inflammation may be partially related to testosterone-induced decreases in IL-17A. IL-17A induces expression of glucocorticoid receptor (GR)-β, which inhibits GR-α and is primarily responsible for anti-inflammatory action via the suppression of the expression of genes relating to inflammatory cytokines. Thus, testosterone-induced declines in IL-17A may decrease expression of GR-β, resulting in enhanced corticoid action [175]. Potent therapeutic action was also demonstrated for precursors of testosterone produced by adrenal glands. For instance, DHEA inhibited the epithelial–mesenchymal transition, an important mechanism involved in airway remodeling, via the inhibition of the PI3K/Akt-dependent signaling pathway stimulated by transforming growth factor (TGF)-β1 [176]. The anti-inflammatory action of DHEAS was confirmed in an in vitro study where DHEAS dose-dependently inhibited chemotaxis of human peripheral blood neutrophils and airway smooth muscle cells [177]. Supplementation with DHEAS improved lung function in asthma patients; in particular, those women who had low DHEAS levels [178]. These and other studies suggest a potential future role for testosterone supplementation in the treatment of asthma [175,179,180,181]. However, further research is needed to identify an appropriate androgen derivative and establish appropriate dosing, delivery methods, etc.
An outline of the actions of the abovementioned sex hormones is provided in Figure 2.

5.2. Anatomical and Physiological Differences in the Lungs

Differences in lung architecture may be considered additional factors contributing to sex-related differences in asthma [6]. Due to the action of sex hormones, differences between females and males may be found in the development of the lung, the production of pulmonary surfactant, and the local production and functions of various bioactive substances [179]. Estrogens enhance the development of the lung and the maturation of surfactant-producing type II cells, while androgens have the opposite effect [182], resulting in “male disadvantage” involving a higher risk of respiratory distress syndrome (RDS) and a more severe course in male preterm neonates. Although female neonates have smaller lungs with fewer respiratory bronchioles at birth, they are more mature than the lungs of male neonates [183].
In the prepubertal stage, the higher prevalence of asthma in boys is partially attributable to dysanaptic lung growth expressed as retarded airway growth compared to parenchymal growth [6]. In adulthood, a disproportional relationship between lung and airways sizes has been found in women, who had smaller airways in relation to the lung size, and women’s smaller lung volumes and lower maximum expiratory flow compared to men may also contribute to sex differences in asthma [6]. In addition, the lower capacity of the airways in women may be partially responsible for the lower efficacy of inhaled corticosteroids because of the presumably lower airway deposition of the delivered drug [184].

5.3. Obesity and Lifestyle

As mentioned above, obesity is an important risk factor for asthma. Chen et al. demonstrated that obesity is associated with higher risk of asthma and increased asthma severity in women but not in men [108]. This was later confirmed by other studies, including a cluster analysis, which demonstrated female predominance in the less atopic—but corticosteroid-unresponsive—asthma associated with obesity [185]. More recently, several authors have suggested that obesity-associated phenotyping should differentiate between two phenotypes: (1) obese asthmatics with an age of onset < 12 years, more atopic features, poor control, significant airway hyperreactivity (AHR), and increased type 2 biomarkers, who do not demonstrate sex predominance; and (2) obese asthmatics with an age of onset > 12 years, fewer atopic features, lower AHR, better control, and normal values for type 2 biomarkers, who do show female predominance [186,187,188].
The mechanisms proposed for how obesity can interact with asthma have still not been fully elucidated [188,189]. Obesity is associated with increased macrophages and ILC2s producing IL-5 and IL-13. Obesity-associated inflammation is also linked with elevated levels of pro-inflammatory cytokines, such as IL-1β, IL-6, TNFα, and IFN-γ, and increased oxidative stress. In addition, adipose tissue produces estrogens, leptin, and adiponectin. Adiponectin predominantly exerts anti-inflammatory effects via the inhibition of pro-inflammatory NF-κB, IL-6, and TNFα [190,191]. On the other hand, potentially increased levels of leptin due to estrogens and progesterone treatment affect both immune and adaptive immune responses [192]. They enhance production of Th1 cytokines (IL-2, IFN-γ, and TNFα) and decrease production of Th2 cytokines (IL-4, IL-5, and IL-10) [191]. In analyses of estrogens and leptin concentrations in serum, no differences between obese and non-obese women were found for estrogens [111], but increased leptin levels were detected in women compared to men at any given BMI [193]. High obesity-induced concentrations of pro-inflammatory cytokines may also be responsible for poor response to corticosteroids, as the cytokines reduce the induction of mitogen-activated kinase phosphatase 1 by glucocorticoids [194,195].
Furthermore, increases in chest wall loading in severe obesity may reduce lung volume and thereby worsen dyspnea [196]. Weight loss may improve the situation, as demonstrated in the study by Dixon et al., where reducing BMI improved asthma symptoms and pulmonary function tests [197]. However, the response to the weight loss may depend on the asthma subtype. Chapman et al. found that weight loss in obese patients could enhance lung function only in T2-high but not in T2-low asthmatics, suggesting a complex interplay between asthma and obesity [198].
In relation to obesity, adults with asthma usually show lower physical activity [199], and women are generally less engaged in regular physical activity than men [200], which may contribute to the association between obesity and asthma in women [181].

5.4. Environmental Factors and Smoking

The occurrence of occupational exposure-associated asthma depends on the individual occupations predominantly performed by men and women. Among women, inorganic dust, hair products, and ozone cause asthma more frequently than among men, while among men, organic dust, diesel, flour/bakery products, and wood or wood-component dust deteriorate lung function more frequently than among women [50,201].
Although men are generally more likely to smoke, smoking among women has dramatically increased during the last 50 years. In addition, women who smoke have an increased risk of asthma [202]. Greater lung vulnerability in women to tobacco smoke may be explained by, for example, estrogen-induced upregulation of the expression of cytochrome P450 (CYP) enzymes involved in the metabolism of the constituents of cigarette smoke, which may lead to production of intermediates potentially toxic for the lungs [203]. Moreover, cigarette smoke exposure may alter estrogen signaling in the airway smooth muscle, which may increase airway contractility in women [204].
Smoking also affects responses to treatment. In the study by Lazarus et al., inhaled corticosteroid beclomethasone decreased sputum eosinophils and ECP in both smokers and non-smokers but only increased forced expiratory volume in 1 s (FEV1) in non-smokers [205]. Similar declines in therapeutic responses were found for oral corticosteroids in smokers with asthma [206]. Reduced effectiveness of corticosteroids in smokers may be attributable to smoking-induced oxidative and nitrosative stress and the related reduction in histone deacetylase activity or to neutrophilic inflammation and overproduction of pro-inflammatory cytokines. However, other factors, such as reduced numbers of glucocorticoid receptors (GRs) or altered affinity of the ligand for GRs, should be also taken into account [207,208,209], suggesting that smoking may be considered a predictor of weak response to corticosteroids [205]. On the other hand, oral administration of the leukotriene receptor antagonist montelukast enhanced peak flow in smokers but not in non-smokers, which may be explained by enhanced leukotriene synthesis or sensitivity in smokers [205].
However, little is known about differing sensitivity to treatment among smoking men and women. The 23 year follow-up study by Dijkstra et al. showed that both male and female adult patients with moderate to severe asthma demonstrated comparable declines in lung function before therapy with inhaled corticosteroids [208]. The beneficial effects of inhaled corticosteroids were dose-dependent and present in men with <5 pack years but not in men with >5 pack years or in women [210]. This may have been related to the fact that smoking contributes predominantly to neutrophilic inflammation, which is more frequent in women and against which inhaled corticosteroids are less effective.

5.5. Chronic Stress

In chronic respiratory disorders, the influence of other hormonal systems—primarily, activation of the hypothalamic–pituitary–adrenal (HPA) axis—should be considered. Chronic stress-induced elevation in the production of glucocorticoids may limit certain actions of synthetic corticosteroids, contributing to corticosteroid resistance [211]. There is a very close relationship between stress, activation of immune responses, and disorders of the respiratory system, including asthma [212,213,214]. As reviewed in the article by Miyasaka et al., stress response facilitates events in the central nervous and endocrine systems. Secretion of stress hormones, including glucocorticoids, epinephrine, and norepinephrine, into the blood enhances Th2 and Th17 immune responses and attenuates regulatory T (Treg) cell responses in humans and rodents, thus exacerbating asthma [215]. For instance, psychosocial stress in mice increased airway reactivity to allergens, elevated counts of inflammatory cells infiltrating the lungs, and increased concentrations of cytokines in serum and bronchoalveolar lavage fluid in comparison to an ovalbumin-sensitized group without exposure to psychosocial stress [216]. Similar results were also observed in humans, among whom chronic psychosocial stress was associated with more frequent asthma exacerbations [217]. However, it is not clear whether any sex-related differences exist in the relationship between stress and asthma. While no such differences were found in the study by Eng et al. [218], in the study by Runeson-Broberg and Norbäck, work-related psychosocial stress was reported as a risk factor for asthma only in men [219]. In contrast, associations between a higher degree of work-related stress and the onset of asthma [220] and between both work stress and family stress and asthma [221] have been clearly demonstrated in women. Moreover, women perceive asthma more sensitively and report more respiratory discomfort than men, even if the symptoms and objective clinical measures are similar [222].

5.6. Genetically Conditioned Factors

5.6.1. Sex-Specific Genes, Epigenetic Changes, and miRNA

The influences of the genetic framework, epigenetic changes, and miRNA on gender differences in asthma have been thoroughly described in the excellent review by Ekpruke and Silveyra [179]. Recently, researchers have identified male- and female-specific genes, as well as male-and-female specific genes, that are mainly expressed in the airway epithelium, among which several are involved in the pathogenesis of asthma. Gautam et al. found that the male-specific genes FBXL7, ITPR3, and ALOX15 are increased and RAD51B decreased in asthma, while a female-specific gene HLA_DQA1 is decreased. Additional differences were observed for more male-specific hypoxia-inducible factor 1 signaling pathways and for more female-specific IL-17 and chemokine signaling pathways [223]. In a genome analysis of Latin American and African American populations, the 17q12-21 asthma locus was identified as a significant contributor to asthma susceptibility in women [224]. In addition, several asthma-risk sex-specific alleles have been identified for women (2q23.3, 2q34, 6q27, 17213.3) and men (5q31.1, 10q26.1) [225], as well as sex-specific single nucleotide polymorphisms (SNPs) in β2-adrenergic receptors [226] and TSLP [227,228]. Determination of gene alleles or SNPs in asthma patients may indicate which polymorphisms are more or less sensitive for a given therapy, as was demonstrated in the differential response to β2-agonists [229,230].
Gene expression is regulated by small non-coding microRNAs (miRNA). Recent studies have demonstrated that there are sex-specific miRNAs associated with lung function, regulation of immune responses, and asthma [231,232,233]. Circulating miRNA correlate well with clinical parameters and the response to therapy [234,235], suggesting their potential for precise diagnostics and personalized medicine in the future.

5.6.2. Susceptibility to Several Diseases in Relation to Bronchial Asthma

Sex-related differences in asthma prevalence and severity can also be considered from a more global perspective. As recently shown, susceptibilities toward many diseases differ between males and females. In general, the female immune system responds more efficiently to common viral and bacterial infections [236,237], which may result in over-reactive immune responses; e.g., as observed in the higher prevalence of autoimmune diseases [238] and the greater adverse effects of vaccination [239]. Sex-related differences have been also demonstrated for COVID-19, with males exhibiting greater disease severity and mortality than females, likely due to differences in the innate immune system and renin–angiotensin–aldosterone (R-A-A) system [240]. Estrogens were found to interact with the R-A-A system, which is considered one of the most critical pathways affecting COVID-19 infectivity, and modulate vasomotor homeostasis. In contrast, testosterone was found to enhance the expressions of ACE2 receptor and transmembrane protease serine-type 2 (TMPRSS2), thereby increasing viral load and delaying viral clearance in men compared to women [241]. In the study by Wark et al., elevated ACE2 gene expression was associated with older age and male sex but not with smoking pack years. However, ACE2 expression was lower in asthma patients and reduced ACE2 protein expression was also found in endobronchial biopsies of asthma patients [242]. Additional studies indicated that having a T2-high asthma phenotype might be associated with reduced COVID-19 morbidity and mortality because of the anti-viral action of eosinophils [243,244], while non-T2 asthma accompanied by high IFN-γ levels, eosinopenia, lymphopenia, a higher neutrophil-to-lymphocyte ratio in peripheral blood, and increased lymphocytes in the bronchoalveolar lavage fluid could be associated with a higher risk of more severe COVID-19 [245].

6. Sex-Related Differences in Responses to Therapies Given for Asthma

Generally, more currently available treatment options target the T2-high type of asthma, which is less common in women. This may partially explain the worse control of asthma and more severe clinical manifestations among women [50,246]. The sex-related differences in responses to therapy may be caused by several factors, including the actions of male/female sex hormones, airway geometry, the deposition and particle size of inhaled corticosteroids, and differences in lung compliance [50,210].
In both male and female mice with a T2-high model of asthma, intranasally delivered corticosteroid budesonide dose-dependently diminished pulmonary inflammation and airway hyperreactivity; however, female mice were slightly less sensitive to the inhibitory effects of budesonide on IL-5 synthesis and the generation of airway hyperreactivity [153]. Similarly, in human adults, inhaled steroids are more effective in men compared to women [210].
In contrast, anti-leukotrienes are more effective in women compared to men, presumably due to androgen-induced attenuation of 5-lipoxygenase protein (FLAP), a protein necessary for activation of 5-lipoxygenase and production of leukotrienes [247]. In women, a stronger bronchodilator response [248] and a tendency for a more sensitive response to bronchodilator [249] have also been found, as well as a better response to anti-IL-5 mepozulimab [250]. Nevertheless, no gender- or sex-related differences were observed for the combination of ICS/LABA/LAMA [251]. The treatment effect of omalizumab, an anti-IgE antibody, was greatest in patients with more severe asthma, while no gender- or sex-related differences were found [252]. In a more recent study, males reported fewer asthma symptoms after omalizumab treatment than women [222].
Major sex-based differences in lung-related features and asthma are presented in Table 4.

7. Future Directions

The differences between the sexes may be primarily caused by the different actions of female and male sex hormones in the lungs. The influence of changing levels of sex hormones on the production of pro- and anti-inflammatory cytokines, airway reactivity, and the production of mucus in the airways represents an important factor in the pathogenesis of asthma. This should be considered in the diagnostics, especially in complex therapy for asthma. However, there are additional factors that may contribute to the sex-related differences in asthma, such as anatomical and physiological differences in the lungs of males and females; differences in genetically conditioned factors; obesity and lifestyle; and exposure to environmental factors, including smoking, chronic stress, etc. Although experimental and clinical research in recent decades has gathered the principal information necessary for differentiation of asthma into several endotypes and phenotypes, resulting in the introduction of more appropriate treatment, there are many questions that should be answered in the future. In particular, large clinical studies should be carried out on the evaluation of sex-related differences in the response to the asthma therapy given. In female patients with asthma, the effects of hormonal contraceptive pills in pre-menopausal women, as well as the effects of HRT in menopause/post-menopause, should also be thoroughly investigated in relation to asthma endotypes/phenotypes. Searches for novel biomarkers should continue in order to find more sensitive and specific indicators for precise diagnoses of asthma endotypes/phenotypes. Further analysis of genetic, epigenetic, and other factors may reveal higher susceptibilities for asthma in selected groups of patients, which may be of importance for early screening of asthma. In addition, with increasing numbers of transgender individuals, clinical studies showing the impact of hormonal substitution are warranted to estimate the effects of exogenously administered hormones on asthma incidence and changes in the severity of asthma in these individuals.

8. Conclusions

Sex-related differences seem to be primarily attributable to the complex actions of sex hormones. However, differences in lung architecture and airway geometry, genetically conditioned factors, obesity and lifestyle, smoking, exposure to environmental and occupational factors, chronic stress, etc. may also contribute to the differences between females and males suffering from asthma. Our article demonstrates that many of these mechanisms and interactions have already been elucidated. Nevertheless, further research is needed to reveal the effects and clinical importance of the mentioned contributing factors. In addition, many additional questions need to be answered; e.g., how sex-based differences may influence the asthma therapy given and what preventive measures could be undertaken to improve the status of asthma patients.

Author Contributions

Writing—original draft preparation, D.M.; writing—review and editing, D.M., R.B. and J.M.; funding acquisition, D.M. and J.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants APVV-15-0075 and VEGA 1/0131/22 to D.M. and grants APVV-18-0084 and VEGA 1/0093/22 to J.M. provided by the Ministry of Education, Science, Research and Sport of the Slovak Republic.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Papi, A.; Brightling, C.; Pedersen, S.E.; Reddel, H.K. Asthma. Lancet 2018, 391, 783–800. [Google Scholar] [CrossRef] [PubMed]
  2. Schleich, F.; Demarche, S.; Louis, R. Biomarkers in the Management of Difficult Asthma. Curr. Top. Med. Chem. 2016, 16, 1561–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kuruvilla, M.E.; Lee, F.E.; Lee, G.B. Understanding Asthma Phenotypes, Endotypes, and Mechanisms of Disease. Clin. Rev. Allergy Immunol. 2019, 56, 219–233. [Google Scholar] [CrossRef] [PubMed]
  4. Palova, R.; Adamcakova, J.; Mokra, D.; Mokry, J. Bronchial asthma: Current trends in treatment. Acta Med. Martiniana 2020, 20, 9–17. [Google Scholar] [CrossRef]
  5. Plevkova, J.; Brozmanova, M.; Harsanyiova, J.; Sterusky, M.; Honetschlager, J.; Buday, T. Various aspects of sex and gender bias in biomedical research. Physiol. Res. 2020, 69, S367–S378. [Google Scholar] [CrossRef] [PubMed]
  6. LoMauro, A.; Aliverti, A. Sex differences in respiratory function. Breathe 2018, 14, 131–140. [Google Scholar] [CrossRef] [Green Version]
  7. Clayton, J.A.; Tannenbaum, C. Reporting Sex, Gender, or Both in Clinical Research? JAMA 2016, 316, 1863–1864. [Google Scholar] [CrossRef] [Green Version]
  8. Tliba, O.; Panettieri, R.A., Jr. Paucigranulocytic asthma: Uncoupling of airway obstruction from inflammation. J. Allergy Clin. Immunol. 2019, 143, 1287–1294. [Google Scholar] [CrossRef] [Green Version]
  9. Pembrey, L.; Brooks, C.; Mpairwe, H.; Figueiredo, C.A.; Oviedo, A.Y.; Chico, M.; Ali, H.; Nambuya, I.; Tumwesige, P.; Robertson, S.; et al. WASP Study Group. Asthma inflammatory phenotypes on four continents: Most asthma is non-eosinophilic. Int. J. Epidemiol. 2022, dyac173. [Google Scholar] [CrossRef]
  10. Svenningsen, S.; Nair, P. Asthma Endotypes and an Overview of Targeted Therapy for Asthma. Front. Med. 2017, 4, 158. [Google Scholar] [CrossRef] [Green Version]
  11. Esteban-Gorgojo, I.; Antolín-Amérigo, D.; Domínguez-Ortega, J.; Quirce, S. Non-eosinophilic asthma: Current perspectives. J. Asthma Allergy. 2018, 11, 267–281. [Google Scholar] [CrossRef] [Green Version]
  12. Muneswarao, J.; Hassali, M.A.; Ibrahim, B.; Saini, B.; Ali, I.A.H.; Verma, A.K. It is time to change the way we manage mild asthma: An update in GINA 2019. Respir. Res. 2019, 20, 183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hinks, T.S.C.; Levine, S.J.; Brusselle, G.G. Treatment options in type-2 low asthma. Eur. Respir. J. 2021, 57, 2000528. [Google Scholar] [CrossRef]
  14. Ramadan, A.A.; Gaffin, J.M.; Israel, E.; Phipatanakul, W. Asthma and Corticosteroid Responses in Childhood and Adult Asthma. Clin. Chest Med. 2019, 40, 163–177. [Google Scholar] [CrossRef] [PubMed]
  15. Papi, A.; Chipps, B.E.; Beasley, R.; Panettieri, R.A., Jr.; Israel, E.; Cooper, M.; Dunsire, L.; Jeynes-Ellis, A.; Johnsson, E.; Rees, R.; et al. Albuterol-Budesonide Fixed-Dose Combination Rescue Inhaler for Asthma. N. Engl. J. Med. 2022, 386, 2071–2083. [Google Scholar] [CrossRef] [PubMed]
  16. Mahemuti, G.; Zhang, H.; Li, J.; Tieliwaerdi, N.; Ren, L. Efficacy and side effects of intravenous theophylline in acute asthma: A systematic review and meta-analysis. Drug Des. Dev. Ther. 2018, 12, 99–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Nakamura, Y.; Tamaoki, J.; Nagase, H.; Yamaguchi, M.; Horiguchi, T.; Hozawa, S.; Ichinose, M.; Iwanaga, T.; Kondo, R.; Nagata, M.; et al. Japanese guidelines for adult asthma 2020. Allergol. Int. 2020, 69, 519–548. [Google Scholar] [CrossRef]
  18. Navinés-Ferrer, A.; Serrano-Candelas, E.; Molina-Molina, G.J.; Martín, M. IgE-Related Chronic Diseases and Anti-IgE-Based Treatments. J. Immunol. Res. 2016, 2016, 8163803. [Google Scholar] [CrossRef] [Green Version]
  19. McGregor, M.C.; Krings, J.G.; Nair, P.; Castro, M. Role of Biologics in Asthma. Am. J. Respir. Crit. Care Med. 2019, 199, 433–445. [Google Scholar] [CrossRef]
  20. Bakakos, A.; Rovina, N.; Bakakos, P. Treatment Challenges in Severe Eosinophilic Asthma: Differential Response to Anti-IL-5 and Anti-IL-5R Therapy. Int. J. Mol. Sci. 2021, 22, 3969. [Google Scholar] [CrossRef]
  21. Farne, H.A.; Wilson, A.; Milan, S.; Banchoff, E.; Yang, F.; Powell, C.V. Anti-IL-5 therapies for asthma. Cochrane Database Syst. Rev. 2022, 7, CD010834. [Google Scholar] [CrossRef] [PubMed]
  22. Delgado, J.; Dávila, I.J.; Domínguez-Ortega, J.; Severe Asthma Group (SEAIC). Clinical Recommendations for the Management of Biological Treatments in Severe Asthma Patients: A Consensus Statement. J. Investig. Allergol. Clin. Immunol. 2021, 31, 36–43. [Google Scholar] [CrossRef] [PubMed]
  23. Ragnoli, B.; Morjaria, J.; Pignatti, P.; Montuschi, P.; Barbieri, M.; Mondini, L.; Ruggero, L.; Trotta, L.; Malerba, M. Dupilumab and tezepelumab in severe refractory asthma: New opportunities. Ther. Adv. Chronic Dis. 2022, 13, 20406223221097327. [Google Scholar] [CrossRef] [PubMed]
  24. Marone, G.; Spadaro, G.; Braile, M.; Poto, R.; Criscuolo, G.; Pahima, H.; Loffredo, S.; Levi-Schaffer, F.; Varricchi, G. Tezepelumab: A novel biological therapy for the treatment of severe uncontrolled asthma. Expert Opin. Investig. Drugs. 2019, 28, 931–940. [Google Scholar] [CrossRef]
  25. Chauhan, B.F.; Jeyaraman, M.M.; Singh Mann, A.; Lys, J.; Abou-Setta, A.M.; Zarychanski, R.; Ducharme, F.M. Addition of anti-leukotriene agents to inhaled corticosteroids for adults and adolescents with persistent asthma. Cochrane Database Syst. Rev. 2017, 3, CD010347. [Google Scholar] [CrossRef]
  26. Bruno, F.; Spaziano, G.; Liparulo, A.; Roviezzo, F.; Nabavi, S.M.; Sureda, A.; Filosa, R.; D’Agostino, B. Recent advances in the search for novel 5-lipoxygenase inhibitors for the treatment of asthma. Eur. J. Med. Chem. 2018, 153, 65–72. [Google Scholar] [CrossRef]
  27. Moran, A.; Pavord, I.D. Anti-IL-4/IL-13 for the treatment of asthma: The story so far. Expert Opin. Biol. Ther. 2020, 20, 283–294. [Google Scholar] [CrossRef]
  28. Braithwaite, I.E.; Cai, F.; Tom, J.A.; Galanter, J.M.; Owen, R.P.; Zhu, R.; Williams, M.; McGregor, A.G.; Eliahu, A.; Durk, M.R.; et al. Inhaled JAK inhibitor GDC-0214 reduces exhaled nitric oxide in patients with mild asthma: A randomized, controlled, proof-of-activity trial. J. Allergy Clin. Immunol. 2021, 148, 783–789. [Google Scholar] [CrossRef]
  29. Georas, S.N.; Donohue, P.; Connolly, M.; Wechsler, M.E. JAK inhibitors for asthma. J. Allergy Clin. Immunol. 2021, 148, 953–963. [Google Scholar] [CrossRef]
  30. Sunata, K.; Kabata, H.; Kuno, T.; Takagi, H.; So, M.; Masaki, K.; Fukunaga, K. The effect of statins for asthma. A systematic review and meta-analysis. J. Asthma 2022, 59, 801–810. [Google Scholar] [CrossRef]
  31. Alabed, M.; Elemam, N.M.; Ramakrishnan, R.K.; Sharif-Askari, N.S.; Kashour, T.; Hamid, Q.; Halwani, R. Therapeutic effect of statins on airway remodeling during asthma. Expert Rev. Respir. Med. 2022, 16, 17–24. [Google Scholar] [CrossRef] [PubMed]
  32. Hiles, S.A.; McDonald, V.M.; Guilhermino, M.; Brusselle, G.G.; Gibson, P.G. Does maintenance azithromycin reduce asthma exacerbations? An individual participant data meta-analysis. Eur. Respir. J. 2019, 54, 1901381. [Google Scholar] [CrossRef] [PubMed]
  33. Gonem, S.; Berair, R.; Singapuri, A.; Hartley, R.; Laurencin, M.F.M.; Bacher, G.; Holzhauer, B.; Bourne, M.; Mistry, V.; Pavord, I.D.; et al. Fevipiprant, a prostaglandin D2 receptor 2 antagonist, in patients with persistent eosinophilic asthma: A single-centre, randomised, double-blind, parallel-group, placebo-controlled trial. Lancet Respir. Med. 2016, 4, 699–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gibson, P.G.; Yang, I.A.; Upham, J.W.; Reynolds, P.N.; Hodge, S.; James, A.L.; Jenkins, C.; Peters, M.J.; Marks, G.B.; Baraket, M.; et al. Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): A randomised, double-blind, placebo-controlled trial. Lancet 2017, 390, 659–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kew, K.M.; Dahri, K. Long-acting muscarinic antagonists (LAMA) added to combination long-acting beta2-agonists and inhaled corticosteroids (LABA/ICS) versus LABA/ICS for adults with asthma. Cochrane Database Syst. Rev. 2016, 2016, CD011721. [Google Scholar] [CrossRef] [Green Version]
  36. Meng, J.F.; Li, H.; Luo, M.J.; Li, H.B. Efficacy of tiotropium in treating patients with moderate-to-severe asthma: A meta-analysis and systematic review based on 14 randomized controlled trials. Medicine 2019, 98, e16637. [Google Scholar] [CrossRef]
  37. Meltzer, E.O.; Chervinsky, P.; Busse, W.; Ohta, K.; Bardin, P.; Bredenbröker, D.; Bateman, E.D. Roflumilast for asthma: Efficacy findings in placebo-controlled studies. Pulm. Pharmacol. Ther. 2015, 35, S20–S27. [Google Scholar] [CrossRef]
  38. Al-Sajee, D.; Yin, X.; Gauvreau, G.M. An evaluation of roflumilast and PDE4 inhibitors with a focus on the treatment of asthma. Expert Opin. Pharmacother. 2019, 20, 609–620. [Google Scholar] [CrossRef]
  39. O’Byrne, P.M.; Metev, H.; Puu, M.; Richter, K.; Keen, C.; Uddin, M.; Larsson, B.; Cullberg, M.; Nair, P. Efficacy and safety of a CXCR2 antagonist, AZD5069, in patients with uncontrolled persistent asthma: A randomised, double-blind, placebo-controlled trial. Lancet Respir. Med. 2016, 4, 797–806. [Google Scholar] [CrossRef]
  40. Watz, H.; Uddin, M.; Pedersen, F.; Kirsten, A.; Goldmann, T.; Stellmacher, F.; Groth, E.; Larsson, B.; Böttcher, G.; Malmgren, A.; et al. Effects of the CXCR2 antagonist AZD5069 on lung neutrophil recruitment in asthma. Pulm. Pharmacol. Ther. 2017, 45, 121–123. [Google Scholar] [CrossRef]
  41. Berry, M.A.; Hargadon, B.; Shelley, M.; Parker, D.; Shaw, D.E.; Green, R.H.; Bradding, P.; Brightling, C.E.; Wardlaw, A.J.; Pavord, I.D. Evidence of a role of tumor necrosis factor alpha in refractory asthma. N. Engl. J. Med. 2006, 354, 697–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Morjaria, J.B.; Chauhan, A.J.; Babu, K.S.; Polosa, R.; Davies, D.E.; Holgate, S.T. The role of a soluble TNFalpha receptor fusion protein (etanercept) in corticosteroid refractory asthma: A double blind, randomised, placebo controlled trial. Thorax 2008, 63, 584–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Busse, W.W.; Holgate, S.; Kerwin, E.; Chon, Y.; Feng, J.; Lin, J.; Lin, S.L. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am. J. Respir. Crit. Care Med. 2013, 188, 1294–1302. [Google Scholar] [CrossRef] [PubMed]
  44. Hynes, G.M.; Hinks, T.S.C. The role of interleukin-17 in asthma: A protective response? ERJ Open Res. 2020, 6, 00364-2019. [Google Scholar] [CrossRef]
  45. Trivedi, A.; Pavord, I.D.; Castro, M. Bronchial thermoplasty and biological therapy as targeted treatments for severe uncontrolled asthma. Lancet Respir. Med. 2016, 4, 585–592. [Google Scholar] [CrossRef]
  46. Goorsenberg, A.W.M.; d’Hooghe, J.N.S.; Srikanthan, K.; Ten Hacken, N.H.T.; Weersink, E.J.M.; Roelofs, J.J.T.H.; Kemp, S.V.; Bel, E.H.; Shah, P.L.; Annema, J.T.; et al. Bronchial Thermoplasty Induced Airway Smooth Muscle Reduction and Clinical Response in Severe Asthma. The TASMA Randomized Trial. Am. J. Respir. Crit. Care Med. 2021, 203, 175–184. [Google Scholar] [CrossRef]
  47. Paivandy, A.; Pejler, G. Novel Strategies to Target Mast Cells in Disease. J. Innate Immun. 2021, 13, 131–147. [Google Scholar] [CrossRef]
  48. Elieh Ali Komi, D.; Bjermer, L. Mast Cell-Mediated Orchestration of the Immune Responses in Human Allergic Asthma: Current Insights. Clin. Rev. Allergy Immunol. 2019, 56, 234–247. [Google Scholar] [CrossRef] [Green Version]
  49. Shah, R.; Newcomb, D.C. Sex Bias in Asthma Prevalence and Pathogenesis. Front. Immunol. 2018, 9, 2997. [Google Scholar] [CrossRef] [Green Version]
  50. Chowdhury, N.U.; Guntur, V.P.; Newcomb, D.C.; Wechsler, M.E. Sex and gender in asthma. Eur. Respir. Rev. 2021, 30, 210067. [Google Scholar] [CrossRef]
  51. Duijts, L. Fetal and infant origins of asthma. Eur. J. Epidemiol. 2012, 27, 5–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Canoy, D.; Pekkanen, J.; Elliott, P.; Pouta, A.; Laitinen, J.; Hartikainen, A.L.; Zitting, P.; Patel, S.; Little, M.P.; Järvelin, M.R. Early growth and adult respiratory function in men and women followed from the fetal period to adulthood. Thorax 2007, 62, 396–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hancox, R.J.; Poulton, R.; Greene, J.M.; McLachlan, C.R.; Pearce, M.S.; Sears, M.R. Associations between birth weight, early childhood weight gain and adult lung function. Thorax 2009, 64, 228–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Jones, M. Effect of preterm birth on airway function and lung growth. Paediatr. Respir. Rev. 2009, 10, 9–11. [Google Scholar] [CrossRef] [PubMed]
  55. Fanaroff, A.A.; Wright, L.L.; Stevenson, D.K.; Shankaran, S.; Donovan, E.F.; Ehrenkranz, R.A.; Younes, N.; Korones, S.B.; Stoll, B.J.; Tyson, J.E.; et al. Very-low-birth-weight outcomes of the National Institute of Child Health and Human Development Neonatal Research Network, May 1991 through December 1992. Am. J. Obstet. Gynecol. 1995, 173, 1423–1431. [Google Scholar] [CrossRef] [PubMed]
  56. Ingemarsson, I. Gender aspects of preterm birth. BJOG 2003, 110, 34–38. [Google Scholar] [CrossRef] [PubMed]
  57. Manktelow, B.N.; Lal, M.K.; Field, D.J.; Sinha, S.K. Antenatal corticosteroids and neonatal outcomes according to gestational age: A cohort study. Arch. Dis. Child. Fetal Neonatal Ed. 2010, 95, F95–F98. [Google Scholar] [CrossRef] [PubMed]
  58. Gortner, L.; Shen, J.; Tutdibi, E. Sexual dimorphism of neonatal lung development. Klin. Padiatr. 2013, 225, 64–69. [Google Scholar] [CrossRef] [PubMed]
  59. Carey, M.A.; Card, J.W.; Voltz, J.W.; Arbes, S.J., Jr.; Germolec, D.R.; Korach, K.S.; Zeldin, D.C. It’s all about sex: Gender, lung development and lung disease. Trends Endocrinol. Metab. 2007, 18, 308–313. [Google Scholar] [CrossRef] [Green Version]
  60. Pei, L.; Chen, G.; Mi, J.; Zhang, T.; Song, X.; Chen, J.; Ji, Y.; Li, C.; Zheng, X. Low birth weight and lung function in adulthood: Retrospective cohort study in China, 1948-1996. Pediatrics 2010, 125, e899–e905. [Google Scholar] [CrossRef]
  61. Sonnenschein-van der Voort, A.M.; Arends, L.R.; de Jongste, J.C.; Annesi-Maesano, I.; Arshad, S.H.; Barros, H.; Basterrechea, M.; Bisgaard, H.; Chatzi, L.; Corpeleijn, E.; et al. Preterm birth, infant weight gain, and childhood asthma risk: A meta-analysis of 147,000 European children. J. Allergy Clin. Immunol. 2014, 133, 1317–1329. [Google Scholar] [CrossRef] [Green Version]
  62. Matheson, M.C.; D Olhaberriague, A.L.; Burgess, J.A.; Giles, G.G.; Hopper, J.L.; Johns, D.P.; Abramson, M.J.; Walters, E.H.; Dharmage, S.C. Preterm birth and low birth weight continue to increase the risk of asthma from age 7 to 43. J. Asthma 2017, 54, 616–623. [Google Scholar] [CrossRef]
  63. Takata, N.; Tanaka, K.; Nagata, C.; Arakawa, M.; Miyake, Y. Preterm birth is associated with higher prevalence of wheeze and asthma in a selected population of Japanese children aged three years. Allergol. Immunopathol. 2019, 47, 425–430. [Google Scholar] [CrossRef]
  64. Castro-Rodriguez, J.A.; Forno, E.; Rodriguez-Martinez, C.E.; Celedón, J.C. Risk and Protective Factors for Childhood Asthma: What Is the Evidence? J. Allergy Clin. Immunol. Pract. 2016, 4, 1111–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lux, A.L.; Henderson, A.J.; Pocock, S.J. Wheeze associated with prenatal tobacco smoke exposure: A prospective, longitudinal study. ALSPAC study team. Arch. Dis. Child. 2000, 83, 307–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lannerö, E.; Wickman, M.; Pershagen, G.; Nordvall, L. Maternal smoking during pregnancy increases the risk of recurrent wheezing during the first years of life (BAMSE). Respir. Res. 2006, 7, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Håberg, S.E.; Stigum, H.; Nystad, W.; Nafstad, P. Effects of pre- and postnatal exposure to parental smoking on early childhood respiratory health. Am. J. Epidemiol. 2007, 166, 679–686. [Google Scholar] [CrossRef] [Green Version]
  68. Forno, E.; Young, O.M.; Kumar, R.; Simhan, H.; Celedón, J.C. Maternal obesity in pregnancy, gestational weight gain, and risk of childhood asthma. Pediatrics 2014, 134, e535–e546. [Google Scholar] [CrossRef] [Green Version]
  69. Zhao, D.; Su, H.; Cheng, J.; Wang, X.; Xie, M.; Li, K.; Wen, L.; Yang, H. Prenatal antibiotic use and risk of childhood wheeze/asthma: A meta-analysis. Pediatr. Allergy Immunol. 2015, 26, 756–764. [Google Scholar] [CrossRef]
  70. Hasunuma, H.; Yoda, Y.; Tokuda, N.; Taniguchi, N.; Takeshima, Y.; Shima, M.; Japan Environment and Children’s Study (JECS) Group. Effects of early-life exposure to dust mite allergen and endotoxin on the development of asthma and wheezing: The Japan Environment and Children’s Study. Clin. Transl. Allergy 2021, 11, e12071. [Google Scholar] [CrossRef]
  71. Jensen, M.E.; Murphy, V.E.; Gibson, P.G.; Mattes, J.; Camargo, C.A., Jr. Vitamin D status in pregnant women with asthma and its association with adverse respiratory outcomes during infancy. J. Matern. Fetal Neonatal Med. 2019, 32, 1820–1825. [Google Scholar] [CrossRef]
  72. Hayatbakhsh, M.R.; Sadasivam, S.; Mamun, A.A.; Najman, J.M.; Williams, G.M.; O’Callaghan, M.J. Maternal smoking during and after pregnancy and lung function in early adulthood: A prospective study. Thorax 2009, 64, 810–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Duijts, L.; Jaddoe, V.W.V.; van der Valk, R.J.P.; Henderson, J.A.; Hofman, A.; Raat, H.; Steegers, E.A.P.; Moll, H.A.; de Jongste, J.C. Fetal exposure to maternal and paternal smoking and the risks of wheezing in preschool children. Chest 2012, 141, 876–885. [Google Scholar] [CrossRef] [PubMed]
  74. Wijga, A.; Tabak, C.; Postma, D.S.; Kerkhof, M.; Wieringa, M.H.; Hoekstra, M.O.; Brunekreef, B.; de Jongste, J.C.; Smit, H.A. Sex differences in asthma during the first 8 years of life: The Prevention and Incidence of Asthma and Mite Allergy (PIAMA) birth cohort study. J. Allergy Clin. Immunol. 2011, 127, 275–277. [Google Scholar] [CrossRef] [PubMed]
  75. Fu, L.; Freishtat, R.J.; Gordish-Dressman, H.; Teach, S.J.; Resca, L.; Hoffman, E.P.; Wang, Z. Natural progression of childhood asthma symptoms and strong influence of sex and puberty. Ann. Am. Thorac. Soc. 2014, 11, 939–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Fuseini, H.; Newcomb, D.C. Mechanisms Driving Gender Differences in Asthma. Curr. Allergy Asthma Rep. 2017, 17, 19. [Google Scholar] [CrossRef] [Green Version]
  77. Naeem, A.; Silveyra, P. Sex Differences in Paediatric and Adult Asthma. Eur. Med. J. (Chelmsf). 2019, 4, 27–35. [Google Scholar] [CrossRef]
  78. Uekert, S.J.; Akan, G.; Evans, M.D.; Li, Z.; Roberg, K.; Tisler, C.; Dasilva, D.; Anderson, E.; Gangnon, R.; Allen, D.B.; et al. Sex-related differences in immune development and the expression of atopy in early childhood. J. Allergy Clin. Immunol. 2006, 118, 1375–1381. [Google Scholar] [CrossRef]
  79. Mohammad, H.R.; Belgrave, D.; Kopec Harding, K.; Murray, C.S.; Simpson, A.; Custovic, A. Age, sex and the association between skin test responses and IgE titres with asthma. Pediatr. Allergy Immunol. 2016, 27, 313–319. [Google Scholar] [CrossRef] [Green Version]
  80. Bibi, H.; Shoseyov, D.; Feigenbaum, D.; Genis, M.; Friger, M.; Peled, R.; Sharff, S. The relationship between asthma and obesity in children: Is it real or a case of over diagnosis? J. Asthma 2004, 41, 403–410. [Google Scholar] [CrossRef]
  81. Ulrik, C.S.; Lophaven, S.N.; Andersen, Z.J.; Sørensen, T.I.; Baker, J.L. BMI at school age and incident asthma admissions in early adulthood: A prospective study of 310,211 children. Clin. Epidemiol. 2018, 10, 605–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Castro-Rodriguez, J.A.; Holberg, C.J.; Morgan, W.J.; Wright, A.L.; Martinez, F.D. Increased incidence of asthmalike symptoms in girls who become overweight or obese during the school years. Am. J. Respir. Crit. Care Med. 2001, 163, 1344–1349. [Google Scholar] [CrossRef] [PubMed]
  83. Ekström, S.; Magnusson, J.; Kull, I.; Andersson, N.; Bottai, M.; Besharat Pour, M.; Melén, E.; Bergström, A. Body mass index development and asthma throughout childhood. Am. J. Epidemiol. 2017, 186, 255–263. [Google Scholar] [CrossRef] [Green Version]
  84. Chen, Y.; Stewart, P.; Johansen, H.; McRae, L.; Taylor, G. Sex difference in hospitalization due to asthma in relation to age. J. Clin. Epidemiol. 2003, 56, 180–187. [Google Scholar] [CrossRef] [PubMed]
  85. Schatz, M.; Camargo, C.A., Jr. The relationship of sex to asthma prevalence, health care utilization, and medications in a large managed care organization. Ann. Allergy Asthma Immunol. 2003, 91, 553–558. [Google Scholar] [CrossRef] [PubMed]
  86. de Marco, R.; Locatelli, F.; Sunyer, J.; Burney, P. Differences in incidence of reported asthma related to age in men and women. A retrospective analysis of the data of the European Respiratory Health Survey. Am. J. Respir. Crit. Care Med. 2000, 162, 68–74. [Google Scholar] [CrossRef] [PubMed]
  87. Fagan, J.K.; Scheff, P.A.; Hryhorczuk, D.; Ramakrishnan, V.; Ross, M.; Persky, V. Prevalence of asthma and other allergic diseases in an adolescent population: Association with gender and race. Ann. Allergy Asthma Immunol. 2001, 86, 177–184. [Google Scholar] [CrossRef] [PubMed]
  88. Eliasson, O.; Scherzer, H.H.; DeGraff, A.C., Jr. Morbidity in asthma in relation to the menstrual cycle. J. Allergy Clin. Immunol. 1986, 77, 87–94. [Google Scholar] [CrossRef]
  89. Shames, R.S.; Heilbron, D.C.; Janson, S.L.; Kishiyama, J.L.; Au, D.S.; Adelman, D.C. Clinical differences among women with and without self-reported perimenstrual asthma. Ann. Allergy Asthma Immunol. 1998, 81, 65–72. [Google Scholar] [CrossRef]
  90. Rao, C.K.; Moore, C.G.; Bleecker, E.; Busse, W.W.; Calhoun, W.; Castro, M.; Chung, K.F.; Erzurum, S.C.; Israel, E.; Curran-Everett, D.; et al. Characteristics of perimenstrual asthma and its relation to asthma severity and control: Data from the Severe Asthma Research Program. Chest 2013, 143, 984–992. [Google Scholar] [CrossRef] [Green Version]
  91. Pauli, B.D.; Reid, R.L.; Munt, P.W.; Wigle, R.D.; Forkert, L. Influence of the menstrual cycle on airway function in asthmatic and normal subjects. Am. Rev. Respir. Dis. 1989, 140, 358–362. [Google Scholar] [CrossRef] [PubMed]
  92. Farha, S.; Asosingh, K.; Laskowski, D.; Hammel, J.; Dweik, R.A.; Wiedemann, H.P.; Erzurum, S.C. Effects of the menstrual cycle on lung function variables in women with asthma. Am. J. Respir. Crit. Care Med. 2009, 180, 304–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Eid, R.C.; Palumbo, M.L.; Cahill, K.N. Perimenstrual Asthma in Aspirin-Exacerbated Respiratory Disease. J. Allergy Clin. Immunol. Pract. 2020, 8, 573–578.e4. [Google Scholar] [CrossRef] [PubMed]
  94. Macsali, F.; Real, F.G.; Omenaas, E.R.; Bjorge, L.; Janson, C.; Franklin, K.; Svanes, C. Oral contraception, body mass index, and asthma: A cross-sectional Nordic-Baltic population survey. J. Allergy Clin. Immunol. 2009, 123, 391–397. [Google Scholar] [CrossRef] [PubMed]
  95. Juniper, E.F.; Kline, P.A.; Roberts, R.S.; Hargreave, F.E.; Daniel, E.E. Airway responsiveness to methacholine during the natural menstrual cycle and the effect of oral contraceptives. Am. Rev. Respir. Dis. 1987, 135, 1039–1042. [Google Scholar] [CrossRef] [PubMed]
  96. Nwaru, B.I.; Sheikh, A. Hormonal contraceptives and asthma in women of reproductive age: Analysis of data from serial national Scottish Health Surveys. J. R. Soc. Med. 2015, 108, 358–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Schatz, M.; Harden, K.; Forsythe, A.; Chilingar, L.; Hoffman, C.; Sperling, W.; Zeiger, R.S. The course of asthma during pregnancy, post partum, and with successive pregnancies: A prospective analysis. J. Allergy Clin. Immunol. 1988, 81, 509–517. [Google Scholar] [CrossRef]
  98. Belanger, K.; Hellenbrand, M.E.; Holford, T.R.; Bracken, M. Effect of pregnancy on maternal asthma symptoms and medication use. Obs. Gynecol. 2010, 115, 559–567. [Google Scholar] [CrossRef] [Green Version]
  99. Juniper, E.F.; Daniel, E.E.; Roberts, R.S.; Kline, P.A.; Hargreave, F.E.; Newhouse, M.T. Improvement in airway responsiveness and asthma severity during pregnancy. A prospective study. Am. Rev. Respir. Dis. 1989, 140, 924–931. [Google Scholar] [CrossRef]
  100. Schatz, M.; Dombrowski, M.P.; Wise, R.; Thom, E.A.; Landon, M.; Mabie, W.; Newman, R.B.; Hauth, J.C.; Lindheimer, M.; Caritis, S.N.; et al. Asthma morbidity during pregnancy can be predicted by severity classification. J. Allergy Clin. Immunol. 2003, 112, 283–288. [Google Scholar] [CrossRef]
  101. Troisi, R.J.; Speizer, F.E.; Willett, W.C.; Trichopoulos, D.; Rosner, B. Menopause, postmenopausal estrogen preparations, and the risk of adultonset asthma: A prospective cohort study. Am. J. Respir Crit. Care Med. 1995, 152, 1183–1188. [Google Scholar] [CrossRef] [PubMed]
  102. Gómez Real, F.; Svanes, C.; Bjornsson, E.H.; Franklin, K.A.; Gislason, D.; Gislason, T.; Gulsvik, A.; Janson, C.; Jögi, R.; Kiserud, T.; et al. Hormone replacement therapy, body mass index and asthma in perimenopausal women: A cross sectional survey. Thorax 2006, 61, 34–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Real, F.G.; Svanes, C.; Omenaas, E.R.; Antò, J.M.; Plana, E.; Jarvis, D.; Janson, C.; Neukirch, F.; Zemp, E.; Dratva, J.; et al. Lung function, respiratory symptoms, and the menopausal transition. J. Allergy Clin. Immunol. 2008, 121, 72–80.e3. [Google Scholar] [CrossRef] [PubMed]
  104. Triebner, K.; Johannessen, A.; Puggini, L.; Benediktsdóttir, B.; Bertelsen, R.J.; Bifulco, E.; Dharmage, S.C.; Dratva, J.; Franklin, K.A.; Gíslason, T.; et al. Menopause as a predictor of new-onset asthma: A longitudinal Northern European population study. J. Allergy Clin. Immunol. 2016, 137, 50–57. [Google Scholar] [CrossRef]
  105. Scioscia, G.; Carpagnano, G.E.; Lacedonia, D.; Soccio, P.; Quarato, C.M.I.; Trabace, L.; Fuso, P.; Barbaro, M.P.F. The Role of Airways 17β-Estradiol as a Biomarker of Severity in Postmenopausal Asthma: A Pilot Study. J. Clin. Med. 2020, 9, 2037. [Google Scholar] [CrossRef]
  106. Nwaru, B.I.; Shah, S.A.; Tibble, H.; Pillinger, R.; McLean, S.; Ryan, D.; Critchley, H.; Hawrylowicz, C.M.; Simpson, C.R.; Soyiri, I.N.; et al. Hormone Replacement Therapy and Risk of Severe Asthma Exacerbation in Perimenopausal and Postmenopausal Women: 17-Year National Cohort Study. J. Allergy Clin. Immunol. Pract. 2021, 9, 2751–2760.e1. [Google Scholar] [CrossRef]
  107. Shah, S.A.; Tibble, H.; Pillinger, R.; McLean, S.; Ryan, D.; Critchley, H.; Price, D.; Hawrylowicz, C.M.; Simpson, C.R.; Soyiri, I.N.; et al. Hormone replacement therapy and asthma onset in menopausal women: National cohort study. J. Allergy Clin. Immunol. 2021, 147, 1662–1670. [Google Scholar] [CrossRef]
  108. Chen, Y.; Dales, R.; Tang, M.; Krewski, D. Obesity may increase the incidence of asthma in women but not in men: Longitudinal observations from the Canadian national population health surveys. Am. J. Epidemiol. 2002, 155, 191–197. [Google Scholar] [CrossRef]
  109. Loerbroks, A.; Apfelbacher, C.J.; Amelang, M.; Stürmer, T. Obesity and adult asthma: Potential effect modification by gender, but not by hay fever. Ann. Epidemiol. 2008, 18, 283–289. [Google Scholar] [CrossRef]
  110. Sutherland, E.R.; Goleva, E.; King, T.S.; Lehman, E.; Stevens, A.D.; Jackson, L.P.; Stream, A.R.; Fahy, J.V.; Leung, D.Y.; Asthma Clinical Research Network. Cluster analysis of obesity and asthma phenotypes. PLoS ONE 2012, 7, e36631. [Google Scholar] [CrossRef]
  111. Scott, H.A.; Gibson, P.G.; Garg, M.L.; Upham, J.W.; Wood, L.G. Sex hormones and systemic inflammation are modulators of the obese-asthma phenotype. Allergy 2016, 71, 1037–1047. [Google Scholar] [CrossRef] [PubMed]
  112. Chakraborty, S.; Pramanik, J.; Mahata, B. Revisiting steroidogenesis and its role in immune regulation with the advanced tools and technologies. Genes Immun. 2021, 22, 125–140. [Google Scholar] [CrossRef] [PubMed]
  113. Ambhore, N.S.; Kalidhindi, R.S.R.; Sathish, V. Sex-Steroid Signaling in Lung Diseases and Inflammation. Adv. Exp. Med. Biol. 2021, 1303, 243–273. [Google Scholar] [CrossRef] [PubMed]
  114. Pérez-Lopez, F.R.; Larrad-Mur, L.; Kallen, A.; Chedraui, P.; Taylor, H.S. Gender differences in cardiovascular disease: Hormonal and biochemical influences. Reprod. Sci. 2010, 17, 511–531. [Google Scholar] [CrossRef] [Green Version]
  115. Shufelt, C.L.; Pacheco, C.; Tweet, M.S.; Miller, V.M. Sex-Specific Physiology and Cardiovascular Disease. Adv. Exp. Med. Biol. 2018, 1065, 433–454. [Google Scholar] [CrossRef]
  116. Stanhewicz, A.E.; Wenner, M.M.; Stachenfeld, N.S. Sex differences in endothelial function important to vascular health and overall cardiovascular disease risk across the lifespan. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H1569–H1588. [Google Scholar] [CrossRef] [Green Version]
  117. Gaignard, P.; Fréchou, M.; Liere, P.; Thérond, P.; Schumacher, M.; Slama, A.; Guennoun, R. Sex differences in brain mitochondrial metabolism: Influence of endogenous steroids and stroke. J. Neuroendocrinol. 2018, 30, e12497. [Google Scholar] [CrossRef] [Green Version]
  118. Rainville, J.R.; Tsyglakova, M.; Hodes, G.E. Deciphering sex differences in the immune system and depression. Front. Neuroendocrinol. 2018, 50, 67–90. [Google Scholar] [CrossRef]
  119. Rubinow, D.R.; Schmidt, P.J. Sex differences and the neurobiology of affective disorders. Neuropsychopharmacology 2019, 44, 111–128. [Google Scholar] [CrossRef] [Green Version]
  120. Palmisano, B.T.; Zhu, L.; Stafford, J.M. Role of Estrogens in the Regulation of Liver Lipid Metabolism. Adv. Exp. Med. Biol. 2017, 1043, 227–256. [Google Scholar] [CrossRef] [Green Version]
  121. Palmisano, B.T.; Zhu, L.; Eckel, R.H.; Stafford, J.M. Sex differences in lipid and lipoprotein metabolism. Mol. Metab. 2018, 15, 45–55. [Google Scholar] [CrossRef] [PubMed]
  122. Tramunt, B.; Smati, S.; Grandgeorge, N.; Lenfant, F.; Arnal, J.F.; Montagner, A.; Gourdy, P. Sex differences in metabolic regulation and diabetes susceptibility. Diabetologia 2020, 63, 453–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Reyes-García, J.; Montaño, L.M.; Carbajal-García, A.; Wang, Y.X. Sex Hormones and Lung Inflammation. Adv. Exp. Med. Biol. 2021, 1304, 259–321. [Google Scholar] [CrossRef] [PubMed]
  124. Plevkova, J.; Buday, T.; Kavalcikova-Bogdanova, N.; Ioan, I.; Demoulin-Alexikova, S. Sex differences in cough reflex. Respir. Physiol. Neurobiol. 2017, 245, 122–129. [Google Scholar] [CrossRef]
  125. Miller, W.L. Steroidogenesis: Unanswered Questions. Trends Endocrinol. Metab. 2017, 28, 771–793. [Google Scholar] [CrossRef]
  126. Cholesterol. Available online: https://en.wikipedia.org/wiki/Cholesterol (accessed on 5 January 2023).
  127. Progesterone. Available online: https://en.wikipedia.org/wiki/Progesterone (accessed on 5 January 2023).
  128. Testosterone. Available online: https://en.wikipedia.org/wiki/Testosterone (accessed on 5 January 2023).
  129. Estradiol. Available online: https://en.wikipedia.org/wiki/Estradiol (accessed on 5 January 2023).
  130. Nelson, L.R.; Bulun, S.E. Estrogen production and action. J. Am. Acad. Dermatol. 2001, 45, S116–S124. [Google Scholar] [CrossRef]
  131. Katzenellenbogen, B.S. Estrogen receptors: Bioactivities and interactions with cell signaling pathways. Biol. Reprod. 1996, 54, 287–293. [Google Scholar] [CrossRef] [Green Version]
  132. Thakur, M.K.; Paramanik, V. Role of steroid hormone coregulators in health and disease. Horm. Res. 2009, 71, 194–200. [Google Scholar] [CrossRef]
  133. Samavat, H.; Kurzer, M.S. Estrogen metabolism and breast cancer. Cancer Lett. 2015, 356, 231–243. [Google Scholar] [CrossRef] [Green Version]
  134. Fuentes, N.; Silveyra, P. Estrogen receptor signaling mechanisms. Adv. Protein Chem. Struct. Biol. 2019, 116, 135–170. [Google Scholar] [CrossRef]
  135. Klinge, C.M. Estrogen receptor interaction with estrogen response elements. Nucleic Acids Res. 2001, 29, 2905–2919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Göttlicher, M.; Heck, S.; Herrlich, P. Transcriptional cross-talk, the second mode of steroid hormone receptor action. J. Mol. Med. 1998, 76, 480–489. [Google Scholar] [CrossRef] [PubMed]
  137. Lösel, R.; Wehling, M. Nongenomic actions of steroid hormones. Nat. Rev. Mol. Cell. Biol. 2003, 4, 46–56. [Google Scholar] [CrossRef] [PubMed]
  138. Björnström, L.; Sjöberg, M. Mechanisms of estrogen receptor signaling: Convergence of genomic and nongenomic actions on target genes. Mol. Endocrinol. 2005, 19, 833–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Vrtačnik, P.; Ostanek, B.; Mencej-Bedrač, S.; Marc, J. The many faces of estrogen signaling. Biochem. Med. 2014, 24, 329–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Liang, J.; Shang, Y. Estrogen and cancer. Annu. Rev. Physiol. 2013, 75, 225–240. [Google Scholar] [CrossRef] [Green Version]
  141. Kuiper, G.G.; Shughrue, P.J.; Merchenthaler, I.; Gustafsson, J.A. The estrogen receptor beta subtype: A novel mediator of estrogen action in neuroendocrine systems. Front. Neuroendocrinol. 1998, 19, 253–286. [Google Scholar] [CrossRef]
  142. Hall, J.M.; McDonnell, D.P. The estrogen receptor beta-isoform (ERbeta) of the human estrogen receptor modulates ERalpha transcriptional activity and is a key regulator of the cellular response to estrogens and antiestrogens. Endocrinology 1999, 140, 5566–5578. [Google Scholar] [CrossRef]
  143. Ivanova, M.M.; Mazhawidza, W.; Dougherty, S.M.; Minna, J.D.; Klinge, C.M. Activity and intracellular location of estrogen receptors alpha and beta in human bronchial epithelial cells. Mol. Cell. Endocrinol. 2009, 305, 12–21. [Google Scholar] [CrossRef] [Green Version]
  144. Chiarella, S.E.; Cardet, J.C.; Prakash, Y.S. Sex, Cells, and Asthma. Mayo Clin. Proc. 2021, 96, 1955–1969. [Google Scholar] [CrossRef]
  145. Townsend, E.A.; Meuchel, L.W.; Thompson, M.A.; Pabelick, C.M.; Prakash, Y.S. Estrogen increases nitric-oxide production in human bronchial epithelium. J. Pharmacol. Exp. Ther. 2011, 339, 815–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Smith, L.C.; Moreno, S.; Robertson, L.; Robinson, S.; Gant, K.; Bryant, A.J.; Sabo-Attwood, T. Transforming growth factor beta1 targets estrogen receptor signaling in bronchial epithelial cells. Respir. Res. 2018, 19, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Tam, A.; Wadsworth, S.; Dorscheid, D.; Man, S.F.; Sin, D.D. Estradiol increases mucus synthesis in bronchial epithelial cells. PLoS ONE 2014, 9, e100633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Degano, B.; Prévost, M.C.; Berger, P.; Molimard, M.; Pontier, S.; Rami, J.; Escamilla, R. Estradiol decreases the acetylcholine-elicited airway reactivity in ovariectomized rats through an increase in epithelial acetylcholinesterase activity. Am. J. Respir. Crit. Care Med. 2001, 164, 1849–1854. [Google Scholar] [CrossRef] [PubMed]
  149. Card, J.W.; Carey, M.A.; Bradbury, J.A.; DeGraff, L.M.; Morgan, D.L.; Moorman, M.P.; Flake, G.P.; Zeldin, D.C. Gender differences in murine airway responsiveness and lipopolysaccharide-induced inflammation. J. Immunol. 2006, 177, 621–630. [Google Scholar] [CrossRef] [Green Version]
  150. Dimitropoulou, C.; White, R.E.; Ownby, D.R.; Catravas, J.D. Estrogen reduces carbachol-induced constriction of asthmatic airways by stimulating large-conductance voltage and calcium-dependent potassium channels. Am. J. Respir. Cell. Mol. Biol. 2005, 32, 239–247. [Google Scholar] [CrossRef]
  151. Matsubara, S.; Swasey, C.H.; Loader, J.E.; Dakhama, A.; Joetham, A.; Ohnishi, H.; Balhorn, A.; Miyahara, N.; Takeda, K.; Gelfand, E.W. Estrogen determines sex differences in airway responsiveness after allergen exposure. Am. J. Respir. Cell. Mol. Biol. 2008, 38, 501–508. [Google Scholar] [CrossRef] [Green Version]
  152. Hayashi, T.; Adachi, Y.; Hasegawa, K.; Morimoto, M. Less sensitivity for late airway inflammation in males than females in BALB/c mice. Scand. J. Immunol. 2003, 57, 562–567. [Google Scholar] [CrossRef]
  153. Corteling, R.; Trifilieff, A. Gender comparison in a murine model of allergen-driven airway inflammation and the response to budesonide treatment. BMC. Pharmacol. 2004, 4, 4. [Google Scholar] [CrossRef] [Green Version]
  154. Melgert, B.N.; Postma, D.S.; Kuipers, I.; Geerlings, M.; Luinge, M.A.; van der Strate, B.W.; Kerstjens, H.A.; Timens, W.; Hylkema, M.N. Female mice are more susceptible to the development of allergic airway inflammation than male mice. Clin. Exp. Allergy 2005, 35, 1496–1503. [Google Scholar] [CrossRef]
  155. Paharkova-Vatchkova, V.; Maldonado, R.; Kovats, S. Estrogen preferentially promotes the differentiation of CD11c+ CD11b(intermediate) dendritic cells from bone marrow precursors. J. Immunol. 2004, 172, 1426–1436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Cephus, J.Y.; Stier, M.T.; Fuseini, H.; Yung, J.A.; Toki, S.; Bloodworth, M.H.; Zhou, W.; Goleniewska, K.; Zhang, J.; Garon, S.L.; et al. Testosterone Attenuates Group 2 Innate Lymphoid Cell-Mediated Airway Inflammation. Cell. Rep. 2017, 21, 2487–2499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Takeda, M.; Tanabe, M.; Ito, W.; Ueki, S.; Konnno, Y.; Chihara, M.; Itoga, M.; Kobayashi, Y.; Moritoki, Y.; Kayaba, H.; et al. Gender difference in allergic airway remodelling and immunoglobulin production in mouse model of asthma. Respirology 2013, 18, 797–806. [Google Scholar] [CrossRef] [PubMed]
  158. Maret, A.; Coudert, J.D.; Garidou, L.; Foucras, G.; Gourdy, P.; Krust, A.; Dupont, S.; Chambon, P.; Druet, P.; Bayard, F.; et al. Estradiol enhances primary antigen-specific CD4 T cell responses and Th1 development in vivo. Essential role of estrogen receptor alpha expression in hematopoietic cells. Eur. J. Immunol. 2003, 33, 512–521. [Google Scholar] [CrossRef]
  159. Conneely, O.M.; Mulac-Jericevic, B.; Lydon, J.P. Progesterone-dependent regulation of female reproductive activity by two distinct progesterone receptor isoforms. Steroids 2003, 68, 771–778. [Google Scholar] [CrossRef]
  160. Giangrande, P.H.; McDonnell, D.P. The A and B isoforms of the human progesterone receptor: Two functionally different transcription factors encoded by a single gene. Recent Prog. Horm. Res. 1999, 54, 291–313. [Google Scholar]
  161. Piette, P.C.M. The pharmacodynamics and safety of progesterone. Best Pract. Res. Clin. Obstet. Gynaecol. 2020, 69, 13–29. [Google Scholar] [CrossRef]
  162. Asavasupreechar, T.; Saito, R.; Miki, Y.; Edwards, D.P.; Boonyaratanakornkit, V.; Sasano, H. Systemic distribution of progesterone receptor subtypes in human tissues. J. Steroid. Biochem. Mol. Biol. 2020, 199, 105599. [Google Scholar] [CrossRef]
  163. Hellings, P.W.; Vandekerckhove, P.; Claeys, R.; Billen, J.; Kasran, A.; Ceuppens, J.L. Progesterone increases airway eosinophilia and hyper-responsiveness in a murine model of allergic asthma. Clin. Exp. Allergy 2003, 33, 1457–1463. [Google Scholar] [CrossRef]
  164. Mitchell, V.L.; Gershwin, L.J. Progesterone and environmental tobacco smoke act synergistically to exacerbate the development of allergic asthma in a mouse model. Clin. Exp. Allergy 2007, 37, 276–286. [Google Scholar] [CrossRef]
  165. Jain, R.; Ray, J.M.; Pan, J.H.; Brody, S.L. Sex hormone-dependent regulation of cilia beat frequency in airway epithelium. Am. J. Respir. Cell. Mol. Biol. 2012, 46, 446–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Chang, C.; Saltzman, A.; Yeh, S.; Young, W.; Keller, E.; Lee, H.J.; Wang, C.; Mizokami, A. Androgen receptor: An overview. Crit. Rev. Eukaryot. Gene Expr. 1995, 5, 97–125. [Google Scholar] [CrossRef] [PubMed]
  167. Mikkonen, L.; Pihlajamaa, P.; Sahu, B.; Zhang, F.P.; Jänne, O.A. Androgen receptor and androgen-dependent gene expression in lung. Mol. Cell. Endocrinol. 2010, 317, 14–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Fuseini, H.; Yung, J.A.; Cephus, J.Y.; Zhang, J.; Goleniewska, K.; Polosukhin, V.V.; Peebles, R.S., Jr.; Newcomb, D.C. Testosterone Decreases House Dust Mite-Induced Type 2 and IL-17A-Mediated Airway Inflammation. J. Immunol. 2018, 201, 1843–1854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Sakai, H.; Suto, W.; Kai, Y.; Chiba, Y. Mechanisms underlying the pathogenesis of hyper-contractility of bronchial smooth muscle in allergic asthma. J. Smooth Muscle Res. 2017, 53, 37–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Card, J.W.; Voltz, J.W.; Ferguson, C.D.; Carey, M.A.; DeGraff, L.M.; Peddada, S.D.; Morgan, D.L.; Zeldin, D.C. Male sex hormones promote vagally mediated reflex airway responsiveness to cholinergic stimulation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 292, L908–L914. [Google Scholar] [CrossRef] [Green Version]
  171. Bulkhi, A.A.; Shepard, K.V., 2nd; Casale, T.B.; Cardet, J.C. Elevated Testosterone Is Associated with Decreased Likelihood of Current Asthma Regardless of Sex. J. Allergy Clin. Immunol. Pract. 2020, 8, 3029–3035.e4. [Google Scholar] [CrossRef]
  172. Zein, J.G.; McManus, J.M.; Sharifi, N.; Erzurum, S.C.; Marozkina, N.; Lahm, T.; Giddings, O.; Davis, M.D.; DeBoer, M.D.; Comhair, S.A.; et al. Benefits of Airway Androgen Receptor Expression in Human Asthma. Am. J. Respir. Crit. Care Med. 2021, 204, 285–293. [Google Scholar] [CrossRef]
  173. McManus, J.M.; Gaston, B.; Zein, J.; Sharifi, N. Association Between Asthma and Reduced Androgen Receptor Expression in Airways. J. Endocr. Soc. 2022, 6, bvac047. [Google Scholar] [CrossRef]
  174. Weinstein, R.E.; Lobocki, C.A.; Gravett, S.; Hum, H.; Negrich, R.; Herbst, J.; Greenberg, D.; Pieper, D.R. Decreased adrenal sex steroid levels in the absence of glucocorticoid suppression in postmenopausal asthmatic women. J. Allergy Clin. Immunol. 1996, 97, 1–8. [Google Scholar] [CrossRef]
  175. Montaño, L.M.; Flores-Soto, E.; Sommer, B.; Solís-Chagoyán, H.; Perusquía, M. Androgens are effective bronchodilators with anti-inflammatory properties: A potential alternative for asthma therapy. Steroids 2020, 153, 108509. [Google Scholar] [CrossRef] [PubMed]
  176. Xu, L.; Xiang, X.; Ji, X.; Wang, W.; Luo, M.; Luo, S.; Li, K.; Gong, S.; Liu, S.; Ma, L.; et al. Effects and mechanism of dehydroepiandrosterone on epithelial-mesenchymal transition in bronchial epithelial cells. Exp. Lung Res. 2014, 40, 211–221. [Google Scholar] [CrossRef] [PubMed]
  177. Koziol-White, C.J.; Goncharova, E.A.; Cao, G.; Johnson, M.; Krymskaya, V.P.; Panettieri, R.A., Jr. DHEA-S inhibits human neutrophil and human airway smooth muscle migration. Biochim. Biophys. Acta 2012, 1822, 1638–1642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Marozkina, N.; Zein, J.; DeBoer, M.D.; Logan, L.; Veri, L.; Ross, K.; Gaston, B. Dehydroepiandrosterone Supplementation May Benefit Women with Asthma Who Have Low Androgen Levels: A Pilot Study. Pulm. Ther. 2019, 5, 213–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Ekpruke, C.D.; Silveyra, P. Sex Differences in Airway Remodeling and Inflammation: Clinical and Biological Factors. Front. Allergy. 2022, 3, 875295. [Google Scholar] [CrossRef]
  180. Zhang, G.Q.; Özuygur Ermis, S.S.; Rådinger, M.; Bossios, A.; Kankaanranta, H.; Nwaru, B. Sex Disparities in Asthma Development and Clinical Outcomes: Implications for Treatment Strategies. J. Asthma Allergy 2022, 15, 231–247. [Google Scholar] [CrossRef]
  181. Jenkins, C.R.; Boulet, L.P.; Lavoie, K.L.; Raherison-Semjen, C.; Singh, D. Personalized Treatment of Asthma: The Importance of Sex and Gender Differences. J. Allergy Clin. Immunol. Pract. 2022, 10, 963–971. [Google Scholar] [CrossRef]
  182. Torday, J.S.; Nielsen, H.C. The sex difference in fetal lung surfactant production. Exp. Lung. Res. 1987, 12, 1–19. [Google Scholar] [CrossRef]
  183. Laube, M.; Thome, U.H. Y It Matters-Sex Differences in Fetal Lung Development. Biomolecules 2022, 12, 437. [Google Scholar] [CrossRef]
  184. Esmailpour, N.; Högger, P.; Rabe, K.F.; Heitmann, U.; Nakashima, M.; Rohdewald, P. Distribution of inhaled fluticasone propionate between human lung tissue and serum in vivo. Eur. Respir. J. 1997, 10, 1496–1499. [Google Scholar] [CrossRef] [Green Version]
  185. Wu, W.; Bleecker, E.; Moore, W.; Busse, W.W.; Castro, M.; Chung, K.F.; Calhoun, W.J.; Erzurum, S.; Gaston, B.; Israel, E.; et al. Unsupervised phenotyping of Severe Asthma Research Program participants using expanded lung data. J. Allergy Clin. Immunol. 2014, 133, 1280–1288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Holguin, F.; Bleecker, E.R.; Busse, W.W.; Calhoun, W.J.; Castro, M.; Erzurum, S.C.; Fitzpatrick, A.M.; Gaston, B.; Israel, E.; Jarjour, N.N.; et al. Obesity and asthma: An association modified by age of asthma onset. J. Allergy Clin. Immunol. 2011, 127, 1486–1493.e2. [Google Scholar] [CrossRef] [PubMed]
  187. Diaz, J.; Farzan, S. Clinical implications of the obese-asthma phenotypes. Immunol. Allergy Clin. North Am. 2014, 34, 739–751. [Google Scholar] [CrossRef] [PubMed]
  188. Mohan, A.; Grace, J.; Wang, B.R.; Lugogo, N. The Effects of Obesity in Asthma. Curr. Allergy Asthma Rep. 2019, 19, 49. [Google Scholar] [CrossRef] [PubMed]
  189. Sharma, V.; Cowan, D.C. Obesity, Inflammation, and Severe Asthma: An Update. Curr. Allergy Asthma Rep. 2021, 21, 46. [Google Scholar] [CrossRef] [PubMed]
  190. Wulster-Radcliffe, M.C.; Ajuwon, K.M.; Wang, J.; Christian, J.A.; Spurlock, M.E. Adiponectin differentially regulates cytokines in porcine macrophages. Biochem. Biophys. Res. Commun. 2004, 316, 924–929. [Google Scholar] [CrossRef] [PubMed]
  191. Ali Assad, N.; Sood, A. Leptin, adiponectin and pulmonary diseases. Biochimie 2012, 94, 2180–2189. [Google Scholar] [CrossRef] [Green Version]
  192. Messinis, I.E.; Papageorgiou, I.; Milingos, S.; Asprodini, E.; Kollios, G.; Seferiadis, K. Oestradiol plus progesterone treatment increases serum leptin concentrations in normal women. Hum. Reprod. 2001, 16, 1827–1832. [Google Scholar] [CrossRef] [Green Version]
  193. Kennedy, A.; Gettys, T.W.; Watson, P.; Wallace, P.; Ganaway, E.; Pan, Q.; Garvey, W.T. The metabolic significance of leptin in humans: Gender-based differences in relationship to adiposity, insulin sensitivity, and energy expenditure. J. Clin. Endocrinol. Metab. 1997, 82, 1293–1300. [Google Scholar] [CrossRef]
  194. Peters-Golden, M.; Swern, A.; Bird, S.S.; Hustad, C.M.; Grant, E.; Edelman, J.M. Influence of body mass index on the response to asthma controller agents. Eur. Respir. J. 2006, 27, 495–503. [Google Scholar] [CrossRef] [Green Version]
  195. Sutherland, E.R.; Goleva, E.; Strand, M.; Beuther, D.A.; Leung, D.Y. Body mass and glucocorticoid response in asthma. Am. J. Respir. Crit. Care Med. 2008, 178, 682–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Jones, R.L.; Nzekwu, M.M. The effects of body mass index on lung volumes. Chest 2006, 130, 827–833. [Google Scholar] [CrossRef] [PubMed]
  197. Dixon, A.E.; Pratley, R.E.; Forgione, P.M.; Kaminsky, D.A.; Whittaker-Leclair, L.A.; Griffes, L.A.; Garudathri, J.; Raymond, D.; Poynter, M.E.; Bunn, J.Y.; et al. Effects of obesity and bariatric surgery on airway hyperresponsiveness, asthma control, and inflammation. J. Allergy Clin. Immunol. 2011, 128, 508–515.e2. [Google Scholar] [CrossRef] [Green Version]
  198. Chapman, D.G.; Irvin, C.G.; Kaminsky, D.A.; Forgione, P.M.; Bates, J.H.; Dixon, A.E. Influence of distinct asthma phenotypes on lung function following weight loss in the obese. Respirology 2014, 19, 1170–1177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Xu, M.; Lodge, C.J.; Lowe, A.J.; Dharmage, S.C.; Cassim, R.; Tan, D.; Russell, M.A. Are adults with asthma less physically active? A systematic review and meta-analysis. J. Asthma 2021, 58, 1426–1443. [Google Scholar] [CrossRef] [PubMed]
  200. Rosenfeld, C.S. Sex-dependent differences in voluntary physical activity. J. Neurosci. Res. 2017, 95, 279–290. [Google Scholar] [CrossRef] [Green Version]
  201. Moscato, G.; Apfelbacher, C.; Brockow, K.; Eberle, C.; Genuneit, J.; Mortz, C.G.; Quecchia, C.; Quirce, S.; Siracusa, A.; Tarlo, S.M.; et al. Gender and occupational allergy: Report from the task force of the EAACI Environmental and Occupational Allergy Interest Group. Allergy 2020, 75, 2753–2763. [Google Scholar] [CrossRef]
  202. Langhammer, A.; Johnsen, R.; Holmen, J.; Gulsvik, A.; Bjermer, L. Cigarette smoking gives more respiratory symptoms among women than among men. The Nord-Trondelag Health Study (HUNT). J. Epidemiol. Community Health 2000, 54, 917–922. [Google Scholar] [CrossRef] [Green Version]
  203. Ben-Zaken Cohen, S.; Paré, P.D.; Man, S.F.; Sin, D.D. The growing burden of chronic obstructive pulmonary disease and lung cancer in women: Examining sex differences in cigarette smoke metabolism. Am. J. Respir. Crit. Care Med. 2007, 176, 113–120. [Google Scholar] [CrossRef]
  204. Sathish, V.; Freeman, M.R.; Long, E.; Thompson, M.A.; Pabelick, C.M.; Prakash, Y.S. Cigarette Smoke and Estrogen Signaling in Human Airway Smooth Muscle. Cell. Physiol. Biochem. 2015, 36, 1101–1115. [Google Scholar] [CrossRef]
  205. Lazarus, S.C.; Chinchilli, V.M.; Rollings, N.J.; Boushey, H.A.; Cherniack, R.; Craig, T.J.; Deykin, A.; DiMango, E.; Fish, J.E.; Ford, J.G.; et al. National Heart Lung and Blood Institute’s Asthma Clinical Research Network. Smoking affects response to inhaled corticosteroids or leukotriene receptor antagonists in asthma. Am. J. Respir. Crit. Care Med. 2007, 175, 783–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Chaudhuri, R.; Livingston, E.; McMahon, A.D.; Thomson, L.; Borland, W.; Thomson, N.C. Cigarette smoking impairs the therapeutic response to oral corticosteroids in chronic asthma. Am. J. Respir. Crit. Care Med. 2003, 168, 1308–1311. [Google Scholar] [CrossRef] [PubMed]
  207. Ricciardolo, F.L.; Di Stefano, A. Corticosteroid resistance in smokers with asthma. Am. J. Respir. Crit. Care Med. 2004, 169, 1252. [Google Scholar] [CrossRef] [PubMed]
  208. Goleva, E.; Li, L.B.; Eves, P.T.; Strand, M.J.; Martin, R.J.; Leung, D.Y. Increased glucocorticoid receptor beta alters steroid response in glucocorticoid-insensitive asthma. Am. J. Respir. Crit. Care Med. 2006, 173, 607–616. [Google Scholar] [CrossRef] [Green Version]
  209. Thomson, N.C.; Chaudhuri, R. Asthma in smokers: Challenges and opportunities. Curr. Opin. Pulm. Med. 2009, 15, 39–45. [Google Scholar] [CrossRef]
  210. Dijkstra, A.; Vonk, J.M.; Jongepier, H.; Koppelman, G.H.; Schouten, J.P.; ten Hacken, N.H.; Timens, W.; Postma, D.S. Lung function decline in asthma: Association with inhaled corticosteroids, smoking and sex. Thorax 2006, 61, 105–110. [Google Scholar] [CrossRef] [Green Version]
  211. Palumbo, M.L.; Prochnik, A.; Wald, M.R.; Genaro, A.M. Chronic Stress and Glucocorticoid Receptor Resistance in Asthma. Clin. Ther. 2020, 42, 993–1006. [Google Scholar] [CrossRef]
  212. Chen, E.; Miller, G.E. Stress and inflammation in exacerbations of asthma. Brain Behav. Immun. 2007, 21, 993–999. [Google Scholar] [CrossRef] [Green Version]
  213. Loerbroks, A.; Gadinger, M.C.; Bosch, J.A.; Stürmer, T.; Amelang, M. Work-related stress, inability to relax after work and risk of adult asthma: A population-based cohort study. Allergy 2010, 65, 1298–1305. [Google Scholar] [CrossRef]
  214. Plourde, A.; Lavoie, K.L.; Raddatz, C.; Bacon, S.L. Effects of acute psychological stress induced in laboratory on physiological responses in asthma populations: A systematic review. Respir. Med. 2017, 127, 21–32. [Google Scholar] [CrossRef] [Green Version]
  215. Miyasaka, T.; Dobashi-Okuyama, K.; Takahashi, T.; Takayanagi, M.; Ohno, I. The interplay between neuroendocrine activity and psychological stress-induced exacerbation of allergic asthma. Allergol. Int. 2018, 67, 32–42. [Google Scholar] [CrossRef] [PubMed]
  216. Li, B.; Duan, X.H.; Wu, J.F.; Liu, B.J.; Luo, Q.L.; Jin, H.L.; Du, Y.J.; Zhang, H.Y.; Cao, Y.X.; Dong, J.C. Impact of psychosocial stress on airway inflammation and its mechanism in a murine model of allergic asthma. Chin. Med. J. 2013, 126, 325–334. [Google Scholar] [PubMed]
  217. Haczku, A.; Panettieri, R. A Jr. Social stress and asthma: The role of corticosteroid insensitivity. J. Allergy Clin. Immunol. 2010, 125, 550–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Eng, A.; Mannetje, A.; Pearce, N.; Douwes, J. Work-related stress and asthma: Results from a workforce survey in New Zealand. J. Asthma 2011, 48, 783–789. [Google Scholar] [CrossRef] [Green Version]
  219. Runeson-Broberg, R.; Norbäck, D. Work-related psychosocial stress as a risk factor for asthma, allergy, and respiratory infections in the Swedish workforce. Psychol. Rep. 2014, 114, 377–389. [Google Scholar] [CrossRef]
  220. Forster, F.; Weinmann, T.; Gerlich, J.; Schlotz, W.; Weinmayr, G.; Genuneit, J.; Windstetter, D.; Vogelberg, C.; von Mutius, E.; Nowak, D.; et al. Work-related stress and incident asthma and rhinitis: Results from the SOLAR study. Int. Arch. Occup. Environ. Health 2019, 92, 673–681. [Google Scholar] [CrossRef]
  221. Loerbroks, A.; Ding, H.; Han, W.; Wang, H.; Wu, J.P.; Yang, L.; Angerer, P.; Li, J. Work stress, family stress and asthma: A cross-sectional study among women in China. Int. Arch. Occup. Environ. Health 2017, 90, 349–356. [Google Scholar] [CrossRef]
  222. Colombo, D.; Zagni, E.; Ferri, F.; Canonica, G.W.; PROXIMA study centers. Gender differences in asthma perception and its impact on quality of life: A post hoc analysis of the PROXIMA (Patient Reported Outcomes and Xolair® In the Management of Asthma) study. Allergy Asthma Clin. Immunol. 2019, 15, 65. [Google Scholar] [CrossRef]
  223. Gautam, Y.; Afanador, Y.; Abebe, T.; López, J.E.; Mersha, T.B. Genome-wide analysis revealed sex-specific gene expression in asthmatics. Hum. Mol. Genet. 2019, 28, 2600–2614. [Google Scholar] [CrossRef]
  224. Espuela-Ortiz, A.; Herrera-Luis, E.; Lorenzo-Díaz, F.; Hu, D.; Eng, C.; Villar, J.; Rodriguez-Santana, J.R.; Burchard, E.G.; Pino-Yanes, M. Role of Sex on the Genetic Susceptibility to Childhood Asthma in Latinos and African Americans. J. Pers. Med. 2021, 11, 1140. [Google Scholar] [CrossRef]
  225. Myers, R.A.; Scott, N.M.; Gauderman, W.J.; Qiu, W.; Mathias, R.A.; Romieu, I.; Levin, A.M.; Pino-Yanes, M.; Graves, P.E.; Villarreal, A.B.; et al. Genome-wide interaction studies reveal sex-specific asthma risk alleles. Hum. Mol. Genet. 2014, 23, 5251–5259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Santillan, A.A.; Camargo, C.A., Jr.; Ramirez-Rivera, A.; Delgado-Enciso, I.; Rojas-Martinez, A.; Cantu-Diaz, F.; Barrera-Saldaña, H.A. Association between beta2-adrenoceptor polymorphisms and asthma diagnosis among Mexican adults. J. Allergy Clin. Immunol. 2003, 112, 1095–1100. [Google Scholar] [CrossRef] [PubMed]
  227. Hunninghake, G.M.; Soto-Quirós, M.E.; Avila, L.; Kim, H.P.; Lasky-Su, J.; Rafaels, N.; Ruczinski, I.; Beaty, T.H.; Mathias, R.A.; Barnes, K.C.; et al. TSLP polymorphisms are associated with asthma in a sex-specific fashion. Allergy 2010, 65, 1566–1575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Birben, E.; Sahiner, U.M.; Karaaslan, C.; Yavuz, T.S.; Cosgun, E.; Kalayci, O.; Sackesen, C. The genetic variants of thymic stromal lymphopoietin protein in children with asthma and allergic rhinitis. Int. Arch. Allergy Immunol. 2014, 163, 185–192. [Google Scholar] [CrossRef] [PubMed]
  229. Syamsu; Yusuf, I.; Budu; Patellongi, I. The effect of polymorphism of the beta-2 adrenergic receptor on the response to beta-2 agonist in bronchial asthma patients. Acta Med. Indones. 2007, 39, 8–12. [Google Scholar]
  230. Labuda, M.; Laberge, S.; Brière, J.; Bérubé, D.; Krajinovic, M. RGS5 gene and therapeutic response to short acting bronchodilators in paediatric asthma patients. Pediatr. Pulmonol. 2013, 48, 970–975. [Google Scholar] [CrossRef]
  231. Pua, H.H.; Ansel, K.M. MicroRNA regulation of allergic inflammation and asthma. Curr. Opin. Immunol. 2015, 36, 101–118. [Google Scholar] [CrossRef] [Green Version]
  232. Kho, A.T.; Sharma, S.; Davis, J.S.; Spina, J.; Howard, D.; McEnroy, K.; Moore, K.; Sylvia, J.; Qiu, W. Circulating MicroRNAs: Association with Lung Function in Asthma. PLoS ONE 2016, 11, e0157998. [Google Scholar] [CrossRef] [Green Version]
  233. Weidner, J.; Bartel, S.; Kılıç, A.; Zissler, U.M.; Renz, H.; Schwarze, J.; Schmidt-Weber, C.B.; Maes, T.; Rebane, A.; Krauss-Etschmann, S.; et al. Spotlight on microRNAs in allergy and asthma. Allergy 2021, 76, 1661–1678. [Google Scholar] [CrossRef]
  234. Weidner, J.; Ekerljung, L.; Malmhäll, C.; Miron, N.; Rådinger, M. Circulating microRNAs correlate to clinical parameters in individuals with allergic and non-allergic asthma. Respir. Res. 2020, 21, 107. [Google Scholar] [CrossRef]
  235. Li, J.; Panganiban, R.; Kho, A.T.; McGeachie, M.J.; Farnam, L.; Chase, R.P.; Weiss, S.T.; Lu, Q.; Tantisira, K.G. Circulating MicroRNAs and Treatment Response in Childhood Asthma. Am. J. Respir. Crit. Care Med. 2020, 202, 65–72. [Google Scholar] [CrossRef] [PubMed]
  236. Jacobsen, H.; Klein, S.L. Sex Differences in Immunity to Viral Infections. Front. Immunol. 2021, 12, 720952. [Google Scholar] [CrossRef] [PubMed]
  237. Dias, S.P.; Brouwer, M.C.; van de Beek, D. Sex and Gender Differences in Bacterial Infections. Infect. Immun. 2022, 90, e0028322. [Google Scholar] [CrossRef] [PubMed]
  238. Ngo, S.T.; Steyn, F.J.; McCombe, P.A. Gender differences in autoimmune disease. Front. Neuroendocrinol. 2014, 35, 347–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Ruggieri, A.; Anticoli, S.; D’Ambrosio, A.; Giordani, L.; Viora, M. The influence of sex and gender on immunity, infection and vaccination. Ann. Ist. Super. Sanita 2016, 52, 198–204. [Google Scholar] [CrossRef] [PubMed]
  240. Viveiros, A.; Rasmuson, J.; Vu, J.; Mulvagh, S.L.; Yip, C.Y.Y.; Norris, C.M.; Oudit, G.Y. Sex differences in COVID-19: Candidate pathways, genetics of ACE2, and sex hormones. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H296–H304. [Google Scholar] [CrossRef] [PubMed]
  241. Chanana, N.; Palmo, T.; Sharma, K.; Kumar, R.; Graham, B.B.; Pasha, Q. Sex-derived attributes contributing to SARS-CoV-2 mortality. Am. J. Physiol. Endocrinol. Metab. 2020, 319, E562–E567. [Google Scholar] [CrossRef]
  242. Wark, P.A.B.; Pathinayake, P.S.; Kaiko, G.; Nichol, K.; Ali, A.; Chen, L.; Sutanto, E.N.; Garratt, L.W.; Sohal, S.S.; Lu, W.; et al. ACE2 expression is elevated in airway epithelial cells from older and male healthy individuals but reduced in asthma. Respirology 2021, 26, 442–451. [Google Scholar] [CrossRef]
  243. Ferastraoaru, D.; Hudes, G.; Jerschow, E.; Jariwala, S.; Karagic, M.; de Vos, G.; Rosenstreich, D.; Ramesh, M. Eosinophilia in Asthma Patients Is Protective Against Severe COVID-19 Illness. J. Allergy Clin. Immunol. Pract. 2021, 9, 1152–1162.e3. [Google Scholar] [CrossRef]
  244. Lipworth, B.; Chan, R.; RuiWen Kuo, C. Type 2 Asthma Inflammation and COVID-19: A Double Edged Sword. J. Allergy Clin. Immunol. Pract. 2021, 9, 1163–1165. [Google Scholar] [CrossRef]
  245. Camiolo, M.; Gauthier, M.; Kaminski, N.; Ray, A.; Wenzel, S.E. Expression of SARS-CoV-2 receptor ACE2 and coincident host response signature varies by asthma inflammatory phenotype. J. Allergy Clin. Immunol. 2020, 146, 315–324.e7. [Google Scholar] [CrossRef] [PubMed]
  246. Rogliani, P.; Cavalli, F.; Ritondo, B.L.; Cazzola, M.; Calzetta, L. Sex differences in adult asthma and COPD therapy: A systematic review. Respir. Res. 2022, 23, 222. [Google Scholar] [CrossRef] [PubMed]
  247. Pace, S.; Pergola, C.; Dehm, F.; Rossi, A.; Gerstmeier, J.; Troisi, F.; Pein, H.; Schaible, A.M.; Weinigel, C.; Rummler, S.; et al. Androgen-mediated sex bias impairs efficiency of leukotriene biosynthesis inhibitors in males. J. Clin. Investig. 2017, 127, 3167–3176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Nerpin, E.; Ferreira, D.S.; Weyler, J.; Schlunnsen, V.; Jogi, R.; Raherison Semjen, C.; Gislasson, T.; Demoly, P.; Heinrich, J.; Nowak, D.; et al. Bronchodilator response and lung function decline: Associations with exhaled nitric oxide with regard to sex and smoking status. World Allergy Organ. J. 2021, 14, 100544. [Google Scholar] [CrossRef] [PubMed]
  249. Lima, J.J.; Mohamed, M.H.; Self, T.H.; Eberle, L.V.; Johnson, J.A. Importance of beta(2)adrenergic receptor genotype, gender and race on albuterol-evoked bronchodilation in asthmatics. Pulm. Pharmacol. Ther. 2000, 13, 127–134. [Google Scholar] [CrossRef]
  250. Harvey, E.S.; Langton, D.; Katelaris, C.; Stevens, S.; Farah, C.S.; Gillman, A.; Harrington, J.; Hew, M.; Kritikos, V.; Radhakrishna, N.; et al. Mepolizumab effectiveness and identification of super-responders in severe asthma. Eur. Respir. J. 2020, 55, 1902420. [Google Scholar] [CrossRef]
  251. Kerstjens, H.A.; Moroni-Zentgraf, P.; Tashkin, D.P.; Dahl, R.; Paggiaro, P.; Vandewalker, M.; Schmidt, H.; Engel, M.; Bateman, E.D. Tiotropium improves lung function, exacerbation rate, and asthma control, independent of baseline characteristics including age, degree of airway obstruction, and allergic status. Respir. Med. 2016, 117, 198–206. [Google Scholar] [CrossRef] [Green Version]
  252. Bousquet, J.; Cabrera, P.; Berkman, N.; Buhl, R.; Holgate, S.; Wenzel, S.; Fox, H.; Hedgecock, S.; Blogg, M.; Cioppa, G.D. The effect of treatment with omalizumab, an anti-IgE antibody, on asthma exacerbations and emergency medical visits in patients with severe persistent asthma. Allergy 2005, 60, 302–308. [Google Scholar] [CrossRef]
Figure 1. Outline of sex hormone synthesis (modified from [112,126,127,128,129]).
Figure 1. Outline of sex hormone synthesis (modified from [112,126,127,128,129]).
Applsci 13 02694 g001
Figure 2. Outline of the actions of estrogens, progesterone, and testosterone in bronchial asthma. Abbreviations: AHR: airway hyperreactivity, ER-α and -β: estrogen receptors, IgE: immunoglobulin E, IL: interleukin, IFN-γ: interferon gamma, ILC2: group 2 innate lymphoid cell, MCP-1: monocyte chemoattractant protein-1, NF-κB: nuclear factor kappa B, NO: nitric oxide, PAI: plasminogen activator inhibitor, TGF-β1: transforming growth factor beta 1, TLR: Toll-like receptor, TNFα: tumor necrosis factor alpha, TSLP: thymic stromal lymphopoietin, ↑: increased, ↓: decreased.
Figure 2. Outline of the actions of estrogens, progesterone, and testosterone in bronchial asthma. Abbreviations: AHR: airway hyperreactivity, ER-α and -β: estrogen receptors, IgE: immunoglobulin E, IL: interleukin, IFN-γ: interferon gamma, ILC2: group 2 innate lymphoid cell, MCP-1: monocyte chemoattractant protein-1, NF-κB: nuclear factor kappa B, NO: nitric oxide, PAI: plasminogen activator inhibitor, TGF-β1: transforming growth factor beta 1, TLR: Toll-like receptor, TNFα: tumor necrosis factor alpha, TSLP: thymic stromal lymphopoietin, ↑: increased, ↓: decreased.
Applsci 13 02694 g002
Table 1. Features of major asthma endotypes (modified from [1,2,3,8]).
Table 1. Features of major asthma endotypes (modified from [1,2,3,8]).
Eosinophilic AsthmaNon-Eosinophilic AsthmaMixed Granulocytic Asthma
AllergicNon-allergicPaucigranulocyticNeutrophilic
Triggering agentsAllergensPollutants, microbesPollutants,
oxidative stress
Pollutants, oxidative stress, microbes, obesity
Involvement of eosinophils+++++
Sputum eosinophils>3%>3%<3%<3%>3%
Blood eosinophils++
Atopy+
Involvement of neutrophils+++
Sputum neutrophils<61%<61%<61%≥61%≥61%
Epithelial damage+++++++++
Mucus+++/−++++
Reticular membrane basement thickening+++++/−++
Airway smooth muscle mass+++++++
Notes: ++: high increase, +: moderate increase, +/−: inconsistent changes, −: no increase.
Table 2. Potential clinical and biochemical biomarkers for the eosinophilic, neutrophilic, and paucigranulocytic endotypes of bronchial asthma (modified from [1,2,3,8]).
Table 2. Potential clinical and biochemical biomarkers for the eosinophilic, neutrophilic, and paucigranulocytic endotypes of bronchial asthma (modified from [1,2,3,8]).
Eosinophilic AsthmaNeutrophilic AsthmaPaucigranulocytic Asthma
FeNO+++/−+
AHR+++/−+
Endotype-specific changes in biochemical biomarkers↑ serum periostin
↑ serum IgE (in atopy)
↑ sputum supernatant ECP, EDN, EPO, eotaxin-2, IL-5, IL-13, GM-CSF, osteopontin, angiopoietin-I, Grx1; ↑ MMP-9
↑ serum CRP and IL-6
↑ sputum supernatant IL-8, IL-17, MPO, NE, TNFα, CXCR1, and CXCR2
↓ sputum gal-3
↑ gal-3BP and IL-1β
↓ gal-3/gal-3BP and IL-1Ra/IL-1β
↑ Grx1
Abbreviations: AHR: airway hyperresponsiveness, CRP: C-reactive protein, CXCR: chemokine (C-X-C motif) receptor, ECP: eosinophil cationic protein, EDN: eosinophil-derived neurotoxin, EPO: eosinophil peroxidase, FeNO: fractional exhaled nitric oxide, gal-3: galectin-3, gal-3BP: galectin-3 binding protein, GM-CSF: granulocyte-macrophage colony-stimulating factor, Grx1: glutaredoxin 1, IgE: immunoglobulin E, IL: interleukin, IL-1Ra: interleukin-1 receptor antagonist, MMP: matrix metalloproteinase, MPO: myeloperoxidase, NE: neutrophil elastase, TNFα: tumor necrosis factor alpha, ++: high increase, +: moderate increase, +/−: low values, ↑: increase, ↓: decrease.
Table 3. Currently available and prospective treatments for eosinophilic, neutrophilic, and paucigranulocytic endotypes of bronchial asthma.
Table 3. Currently available and prospective treatments for eosinophilic, neutrophilic, and paucigranulocytic endotypes of bronchial asthma.
Classified DrugsExamples of Drugs UsedReferences
Eosinophilic asthma
Currently available treatments:
Inhaled corticosteroids (ICSs)Budesonide, fluticasone[12,14,15]
Oral corticosteroids (OCSs)Prednisolone[12,14]
Bronchodilators (SABA, LABA, SAMA, LAMA)Salbutamol, formoterol, ipratropium, tiotropium[12,15]
Xanthine derivativesTheophylline[16,17]
Anti-IgEOmalizumab[18,19]
Anti-IL-5/IL-5RMepolizumab, benralizumab[20,21]
Anti-IL-4RDupilumab[22,23]
Anti-TSLPTezepelumab[23,24]
Anti-leukotrienesMontelukast[25,26]
Prospective treatments:
Anti-IL-13Tralokinumab[27]
Tyrosine kinase inhibitorsDasatinib[28,29]
StatinsSimvastatin, atorvastatin[30,31]
Macrolide antibioticsAzithromycin[32]
PGD2 receptor 2 antagonistFevipiprant[33]
Neutrophilic asthma
Currently available treatments:
Macrolide antibioticsAzithromycin[32,34]
Bronchodilators (LAMA)Tiotropium[35,36]
Xanthine derivativesTheophylline[16,17]
Prospective treatments:
PDE4 inhibitorsRoflumilast[37,38]
StatinsSimvastatin, atorvastatin[30,31]
Tyrosine kinase inhibitorsDasatinib[28,29]
CXCR2 antagonistsAZD5069[39,40]
Anti-TNFαEtanercept[41,42]
Anti-IL-17Brodalumab[43,44]
Paucigranulocytic asthma
Prospective treatments:
Bronchial thermoplasty [45,46]
Bronchodilators (LAMA)Tiotropium[35,36]
Mast-cell directed therapy [47,48]
Abbreviations: CXCR: chemokine (C-X-C motif) receptor, ICSs: inhaled corticosteroids, IgE: immunoglobulin E, IL: interleukin, IL-4R: interleukin-4 receptor, IL-5R: interleukin-5 receptor, LABA: long-acting β2-agonist, LAMA: long-acting muscarinic antagonist, OCSs: oral corticosteroids, PDE: phosphodiesterase, PGD2: prostaglandin D2, SABA: short-acting β2-agonist, SAMA: short-acting muscarinic antagonist, TNFα: tumor necrosis factor alpha, TSLP: thymic stromal lymphopoietin.
Table 4. Sex-based differences in lung-related features and asthma. For more details, see the text above.
Table 4. Sex-based differences in lung-related features and asthma. For more details, see the text above.
MalesFemales
Lung maturation in neonatesDelayed compared to female neonates
Lung dysanapsisRetarded AW growth in boysSmaller AWs vs. lungs in adult women
Excitability for cough
Clinical course of asthma ↑ severity and hospital admissions
Type 2 inflammation
Non-type 2 inflammation
General immune response
Effectiveness of response to viral infections, including COVID-19
Effectiveness of response to bacterial infections
Relation to smoking↑ exposure to smoke↑ vulnerability to smoke
Occupation-associated asthma↑ exposure to organic dust, diesel, flour/bakery products, wood/wood-component dust↑ exposure to inorganic dust, hair products, ozone
Obesity-associated asthma↑ risk in boys↑ risk in adolescent and adult women
Stress-associated asthma↑ risk↑ risk
Sex-specific genes associated with asthmaFBXL7, ITPR3, ALOX15; ↓ RAD51BHLA_DQA1
Sex-specific alleles associated with asthma risk5q31.1, 10q26.12q23.3, 2q34, 6q27, 17213.3
Response to corticosteroids
Response to bronchodilators
Response to anti-leukotrienes
Response to anti-IgE
Response to anti-IL-5
Abbreviations: AW: airway, IgE: immunoglobulin E, IL: interleukin, ↑: increase, ↓: decrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mokra, D.; Barosova, R.; Mokry, J. Sex-Based Differences in Bronchial Asthma: What Are the Mechanisms behind Them? Appl. Sci. 2023, 13, 2694. https://doi.org/10.3390/app13042694

AMA Style

Mokra D, Barosova R, Mokry J. Sex-Based Differences in Bronchial Asthma: What Are the Mechanisms behind Them? Applied Sciences. 2023; 13(4):2694. https://doi.org/10.3390/app13042694

Chicago/Turabian Style

Mokra, Daniela, Romana Barosova, and Juraj Mokry. 2023. "Sex-Based Differences in Bronchial Asthma: What Are the Mechanisms behind Them?" Applied Sciences 13, no. 4: 2694. https://doi.org/10.3390/app13042694

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop