1
|
Huang S, Zou S, Gongye R, Xu S. Age at menarche in relation to risk of chronic obstructive pulmonary disease: Results from the China Kadoorie Biobank study of 300,000 Chinese women. Maturitas 2024; 193:108173. [PMID: 39674076 DOI: 10.1016/j.maturitas.2024.108173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 10/01/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Age at menarche has important implications for lung function. However, little is known about its association with chronic obstructive pulmonary disease (COPD). The present study aims to investigate age at menarche in relation to the risk of COPD among Chinese women. METHODS We analyzed data on 302,463 women aged 30-79 years from 10 regions across China during 2004 to 2008, recruited by the China Kadoorie Biobank baseline survey. Multivariable logistic regression was used to examine odds ratios (ORs) with 95 % confidence intervals (CIs) for the association between COPD and age at menarche. RESULTS Overall, the average age at menarche was 15.4 ± 2.0 years. The relationship between age at menarche and adult COPD was J-shaped. Compared with women with menarche at age 13-14 years, the adjusted ORs for COPD for menarche at ≤12, 15-16, and ≥ 17 years of age were 1.21 (95 % CI: 1.07, 1.36), 1.04 (95 % CI: 0.98, 1.11) and 1.11 (95 % CI, 1.03, 1.19), respectively. The patterns of association appeared to be more pronounced among women living in rural areas and those who ever drank alcohol. CONCLUSIONS In Chinese adults, both early menarche and late menarche are associated with increased risk of COPD; as such, age at menarche may be a factor that could help to identify women at higher COPD risk who would benefit from early preventative strategies.
Collapse
Affiliation(s)
- Sha Huang
- School of Environmental Science and Engineering, Hainan University, Haikou, China.
| | - Siyu Zou
- Vanke School of Public Health, Tsinghua University, Beijing, China; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, MD, USA
| | - Ruofan Gongye
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Shunqing Xu
- School of Environmental Science and Engineering, Hainan University, Haikou, China
| |
Collapse
|
2
|
Tagliapietra G, Citherlet T, Raberin A, Bourdillon N, Krumm B, Narang BJ, Giardini G, Pialoux V, Debevec T, Millet GP. Effect of menstrual cycle phase on physiological responses in healthy women at rest and during submaximal exercise at high altitude. Sci Rep 2024; 14:27793. [PMID: 39537750 PMCID: PMC11561278 DOI: 10.1038/s41598-024-79702-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024] Open
Abstract
As more women engage in high-altitude activities, understanding how ovarian hormone fluctuations affect their cardiorespiratory system is essential for optimizing acclimatization to these environments. This study investigates the effects of menstrual cycle (MC) phases on physiological responses at rest, during and after submaximal exercise, at high-altitude (barometric pressure 509 ± 6 mmHg; partial pressure of inspired oxygen 96 ± 1 mmHg; ambient temperature 21 ± 2 °C and relative humidity 27 ± 4%) in 16 eumenorrheic women. Gas exchange, hemodynamic responses, heart rate variability and heart rate recovery (HRR) were monitored at low altitude, and then at 3375 m on the Mont Blanc (following nocturnal exposure) during both the early-follicular (EF) and mid-luteal (ML) phases. Significant differences were observed between low and high-altitude in ventilation, heart rate and cardiac output. Resting ventilation (15.2 ± 1.9 vs. 13.2 ± 2.5 L.min-1; p = 0.039) and tidal volume (812 ± 217 vs. 713 ± 190 mL; p = 0.027) were higher during EF than ML at high-altitude. These differences between EF and ML were no longer evident during exercise, with comparable responses in oxygen uptake kinetics, cycling efficiency and HRR. The MC had negligible effects on physiological responses to high-altitude. An individualized approach, tailored to each woman's specific responses to hypoxia across the MC, may be more beneficial in optimizing high-altitude sojourns than general guidelines.
Collapse
Affiliation(s)
- Guia Tagliapietra
- Institute of Sport Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, 1015, Switzerland.
| | - Tom Citherlet
- Institute of Sport Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, 1015, Switzerland
| | - Antoine Raberin
- Institute of Sport Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, 1015, Switzerland
| | - Nicolas Bourdillon
- Institute of Sport Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, 1015, Switzerland
| | - Bastien Krumm
- Institute of Sport Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, 1015, Switzerland
| | - Benjamin J Narang
- Faculty of Sport, University of Ljubljana, Ljubljana, 1000, Slovenia
- Department of Automatics, Biocybernetics and Robotics, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Guido Giardini
- Mountain Medicine and Neurology Center Valle D'Aosta, Regional Hospital, Aosta, 11100, Italy
| | - Vincent Pialoux
- Interuniversity Laboratory of Motor Biology, Rockefeller Faculty of Medicine, University of Lyon, Lyon, 69008, France
| | - Tadej Debevec
- Faculty of Sport, University of Ljubljana, Ljubljana, 1000, Slovenia
- Department of Automatics, Biocybernetics and Robotics, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Grégoire P Millet
- Institute of Sport Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, 1015, Switzerland
| |
Collapse
|
3
|
Hughes-Fulford M, Carroll DJ, Allaway HCM, Dunbar BJ, Sawyer AJ. Women in space: A review of known physiological adaptations and health perspectives. Exp Physiol 2024. [PMID: 39487998 DOI: 10.1113/ep091527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/08/2024] [Indexed: 11/04/2024]
Abstract
Exposure to the spaceflight environment causes adaptations in most human physiological systems, many of which are thought to affect women differently from men. Since only 11.5% of astronauts worldwide have been female, these issues are largely understudied. The physiological nuances affecting the female body in the spaceflight environment remain inadequately defined since the last thorough published review on the subject. A PubMed literature search yielded over 2200 publications. Using NASA's 2014 review series 'The effects of sex and gender on adaptation to space' as a benchmark, we identified substantive advancements and persistent knowledge gaps in need of further study from the nearly 600 related articles that have been published since the initial review. This review highlights the most critical issues to mitigate medical risk and promote the success of missions to the Moon and Mars. Salient sex-linked differences observed terrestrially should be studied during upcoming missions, including increased levels of inflammatory markers, coagulation factors and leptin levels following sleep deprivation; correlation between body mass and the severity of spaceflight-associated neuro-ocular syndrome; increased incidence of orthostatic intolerance; increased severity of muscle atrophy and bone loss; differences in the incidence of urinary tract infections; and susceptibility to specific cancers after exposure to ionizing radiation. To optimize health and well-being among all astronauts, it is imperative to prioritize research that considers the physiological nuances of the female body. A more robust understanding of female physiology in the spaceflight environment will support crew readiness for Artemis missions and beyond.
Collapse
Affiliation(s)
- Millie Hughes-Fulford
- UC Space Health, University of California San Francisco (UCSF), San Francisco, California, USA
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Danielle J Carroll
- UC Space Health, University of California San Francisco (UCSF), San Francisco, California, USA
- Department of Surgery, UCSF, San Francisco, California, USA
- Department of Bioastronautics, University of Colorado Boulder, Boulder, Colorado, USA
| | - Heather C M Allaway
- Department of Kinesiology, Texas A&M University, College Station, Texas, USA
- School of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bonnie J Dunbar
- Department of Aerospace Engineering, Texas A&M University, College Station, Texas, USA
- Texas A&M Engineering Experiment Station, Texas A&M University, College Station, Texas, USA
| | - Aenor J Sawyer
- UC Space Health, University of California San Francisco (UCSF), San Francisco, California, USA
- Department of Orthopaedic Surgery, UCSF, San Francisco, California, USA
| |
Collapse
|
4
|
Ambhore NS, Balraj P, Kumar A, Reza MI, Ramakrishnan YS, Tesch J, Lohana S, Sathish V. Kiss1 receptor knockout exacerbates airway hyperresponsiveness and remodeling in a mouse model of allergic asthma. Respir Res 2024; 25:387. [PMID: 39468619 PMCID: PMC11520794 DOI: 10.1186/s12931-024-03017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND In asthma, sex-steroids signaling is recognized as a critical regulator of disease pathophysiology. However, the paradoxical role of sex-steroids, especially estrogen, suggests that an upstream mechanism or even independent of estrogen plays an important role in regulating asthma pathophysiology. In this context, in our previous studies, we explored kisspeptin (Kp) and its receptor Kiss1R's signaling in regulating human airway smooth muscle cell remodeling in vitro and airway hyperresponsiveness (AHR) in vivo in a mouse (wild-type, WT) model of asthma. In this study, we evaluated the effect of endogenous Kp in regulating AHR and remodeling using Kiss1R knockout (Kiss1R-/-) mice. METHODS C57BL/6J WT (Kiss1R+/+) and Kiss1R-/- mice, both male and female, were intranasally challenged with mixed-allergen (MA) and/or phosphate-buffered saline (PBS). We used flexiVent analysis to assess airway resistance (Rrs), elastance (Ers), and compliance (Crs). Following this, broncho-alveolar lavage (BAL) was performed for differential leukocyte count (DLC) and cytokine analysis. Histology staining was performed using hematoxylin and eosin (H&E) for morphological analysis and Masson's Trichrome (MT) for collagen deposition. Additionally, lung sections were processed for immunofluorescence (IF) of Ki-67, α-smooth muscle actin (α-SMA), and tenascin-c. RESULTS Interestingly, the loss of Kiss1R exacerbated lung function and airway contractility in mice challenged with MA, with more profound effects in Kiss1R-/- female mice. MA-challenged Kiss1R-/- mice showed a significant increase in immune cell infiltration and proinflammatory cytokine levels. Importantly, the loss of Kiss1R aggravated Th2/Th17 biased cytokines in MA-challenged mice. Furthermore, histology of lung sections from Kiss1R-/- mice showed increased collagen deposition on airway walls and mucin production in airway cells compared to Kiss1R+/+ mice. In addition, immunofluorescence analysis showed loss of Kiss1R significantly aggravated airway remodeling and subsequently AHR. CONCLUSIONS These findings demonstrate the importance of inherent Kiss1R signaling in regulating airway inflammation, AHR, and remodeling in the pathophysiology of asthma.
Collapse
Affiliation(s)
- Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Premanand Balraj
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Ashish Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Mohammad Irshad Reza
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Yogaraj S Ramakrishnan
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Jacob Tesch
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Sahil Lohana
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND, 58102, USA.
| |
Collapse
|
5
|
Gill NK, Sohi SK, Joseph G, Bhatti N. Hormone Replacement Therapy and Pulmonary Hypertension: A Review of the Literature. Cureus 2024; 16:e71908. [PMID: 39564039 PMCID: PMC11576071 DOI: 10.7759/cureus.71908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 11/21/2024] Open
Abstract
Pulmonary hypertension (PH) is a multifactorial condition that encompasses a group of diseases characterized by a progressive increase in pulmonary vascular resistance and pulmonary arterial pressure, ultimately leading to right heart failure and death. The primary goals of PH treatment are to lower pulmonary arterial pressure, alleviate symptoms such as shortness of breath and chest pain, address modifiable risk factors, and manage the underlying cause, often a common advanced disease like chronic obstructive lung disease or left heart disease. While sex is an unchangeable risk factor for PH development, the presence or absence of estrogens has a significant influence on its progression. Hormone replacement therapy (HRT) is the recommended form of estrogen therapy for postmenopausal women, but only in carefully selected cases. However, a paradox arises, as some research suggests HRT benefits women, while other studies highlight its risks. This review provides a comprehensive analysis of the literature on the role of HRT in PH.
Collapse
Affiliation(s)
- Natasha K Gill
- Internal Medicine, University Hospitals Parma Medical Center, Parma, USA
| | - Supreet K Sohi
- Medicine, Rajendra Institute of Medical Sciences, Patiala, IND
| | - Girish Joseph
- Pharmacology, Christian Medical College and Hospital, Ludhiana, IND
| | - Neena Bhatti
- Pharmacology, Christian Medical College and Hospital, Ludhiana, IND
| |
Collapse
|
6
|
Pelizzo G, Calcaterra V, Baldassarre P, Marinaro M, Taranto S, Ceresola M, Capelo G, Gazzola C, Zuccotti G. The impact of hormones on lung development and function: an overlooked aspect to consider from early childhood. Front Endocrinol (Lausanne) 2024; 15:1425149. [PMID: 39371928 PMCID: PMC11449876 DOI: 10.3389/fendo.2024.1425149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/29/2024] [Indexed: 10/08/2024] Open
Abstract
The impact of hormones on the respiratory system constitutes a multifaceted and intricate facet of human biology. We propose a comprehensive review of recent advancements in understanding the interactions between hormones and pulmonary development and function, focusing on pediatric populations. We explore how hormones can influence ventilation, perfusion, and pulmonary function, from regulating airway muscle tone to modulating the inflammatory response. Hormones play an important role in the growth and development of lung tissues, influencing them from early stages through infancy, childhood, adolescence, and into adulthood. Glucocorticoids, thyroid hormones, insulin, ghrelin, leptin, glucagon-like peptide 1 (GLP-1), retinoids, cholecalciferol sex steroids, hormones derived from adipose tissue, factors like insulin, granulocyte-macrophage colony-stimulating factor (GM-CSF) and glucagon are key players in modulating respiratory mechanics and inflammation. While ample evidence underscores the impact of hormones on lung development and function, along with sex-related differences in the prevalence of respiratory disorders, further research is needed to clarify their specific roles in these conditions. Further research into the mechanisms underlying hormonal effects is essential for the development of customizing therapeutic approaches for respiratory diseases. Understanding the impact of hormones on lung function could be valuable for developing personalized monitoring approaches in both medical and surgical pediatric settings, in order to improve outcomes and the quality of care for pediatric patients.
Collapse
Affiliation(s)
- Gloria Pelizzo
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Valeria Calcaterra
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, Milan, Italy
| | | | - Michela Marinaro
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
| | - Silvia Taranto
- Pediatric Department, Buzzi Children’s Hospital, Milan, Italy
| | - Michele Ceresola
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
| | - Gerson Capelo
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
| | | | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
- Pediatric Department, Buzzi Children’s Hospital, Milan, Italy
| |
Collapse
|
7
|
Xie B, Chen Q, Dai Z, Jiang C, Chen X. Progesterone (P4) ameliorates cigarette smoke-induced chronic obstructive pulmonary disease (COPD). Mol Med 2024; 30:123. [PMID: 39138434 PMCID: PMC11323532 DOI: 10.1186/s10020-024-00883-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory lung disease associated with high morbidity and mortality worldwide. Oxidative injury and mitochondrial dysfunction in the airway epithelium are major events in COPD progression. METHODS AND RESULTS The therapeutic effects of Progesterone (P4) were investigated in vivo and in vitro in this study. In vivo, in a cigarette smoke (CS) exposure-induced COPD mouse model, P4 treatment significantly ameliorated CS exposure-induced physiological and pathological characteristics, including inflammatory cell infiltration and oxidative injury, in a dose-dependent manner. The c-MYC/SIRT1/PGC-1α pathway is involved in the protective function of P4 against CS-induced COPD. In vitro, P4 co-treatment significantly ameliorated H2O2-induced oxidative injury and mitochondrial dysfunctions by promoting cell proliferation, increasing mitochondrial membrane potential, decreasing ROS levels and apoptosis, and increasing ATP content. Moreover, P4 co-treatment partially attenuated H2O2-caused inhibition in Nrf1, Tfam, Mfn1, PGR-B, c-MYC, SIRT1, and PGC-1α levels. In BEAS-2B and ASM cells, the c-MYC/SIRT1 axis regulated P4's protective effects against H2O2-induced oxidative injury and mitochondrial dysfunctions. CONCLUSION P4 activates the c-MYC/SIRT1 axis, ameliorating CS-induced COPD and protecting both airway epithelial cells and smooth muscle cells against H2O2-induced oxidative damage. PGC-1α and downstream mitochondrial signaling pathways might be involved.
Collapse
Affiliation(s)
- Bin Xie
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Chen
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziyu Dai
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chen Jiang
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xi Chen
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
8
|
Harvey BJ, McElvaney NG. Sex differences in airway disease: estrogen and airway surface liquid dynamics. Biol Sex Differ 2024; 15:56. [PMID: 39026347 PMCID: PMC11264786 DOI: 10.1186/s13293-024-00633-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
Biological sex differences exist for many airway diseases in which females have either worse or better health outcomes. Inflammatory airway diseases such as cystic fibrosis (CF) and asthma display a clear male advantage in post-puberty while a female benefit is observed in asthma during the pre-puberty years. The influence of menstrual cycle stage and pregnancy on the frequency and severity of pulmonary exacerbations in CF and asthma point to a role for sex steroid hormones, particularly estrogen, in underpinning biological sex differences in these diseases. There are many ways by which estrogen may aggravate asthma and CF involving disturbances in airway surface liquid (ASL) dynamics, inappropriate hyper-immune and allergenic responses, as well as exacerbation of pathogen virulence. The deleterious effect of estrogen on pulmonary function in CF and asthma contrasts with the female advantage observed in airway diseases characterised by pulmonary edema such as pneumonia, acute respiratory distress syndrome (ARDS) and COVID-19. Airway surface liquid hypersecretion and alveolar flooding are hallmarks of ARDS and COVID-19, and contribute to the morbidity and mortality of severe forms of these diseases. ASL dynamics encompasses the intrinsic features of the thin lining of fluid covering the airway epithelium which regulate mucociliary clearance (ciliary beat, ASL height, volume, pH, viscosity, mucins, and channel activating proteases) in addition to innate defence mechanisms (pathogen virulence, cytokines, defensins, specialised pro-resolution lipid mediators, and metabolism). Estrogen regulation of ASL dynamics contributing to biological sex differences in CF, asthma and COVID-19 is a major focus of this review.
Collapse
Affiliation(s)
- Brian J Harvey
- Faculty of Medicine and Health Sciences, Royal College of Surgeons in Ireland, 126 St Stephens Green, Dublin 2, Ireland.
- Department of Medicine, RCSI ERC, Beaumont Hospital, Dublin 2, Ireland.
| | - Noel G McElvaney
- Faculty of Medicine and Health Sciences, Royal College of Surgeons in Ireland, 126 St Stephens Green, Dublin 2, Ireland
| |
Collapse
|
9
|
Wardhani K, Yazzie S, McVeigh C, Edeh O, Grimes M, Jacquez Q, Dixson C, Barr E, Liu R, Bolt AM, Feng C, Zychowski KE. Systemic immunological responses are dependent on sex and ovarian hormone presence following acute inhaled woodsmoke exposure. Part Fibre Toxicol 2024; 21:27. [PMID: 38797836 PMCID: PMC11129474 DOI: 10.1186/s12989-024-00587-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Rural regions of the western United States have experienced a noticeable surge in both the frequency and severity of acute wildfire events, which brings significant challenges to both public safety and environmental conservation efforts, with impacts felt globally. Identifying factors contributing to immune dysfunction, including endocrinological phenotypes, is essential to understanding how hormones may influence toxicological susceptibility. METHODS This exploratory study utilized male and female C57BL/6 mice as in vivo models to investigate distinct responses to acute woodsmoke (WS) exposure with a focus on sex-based differences. In a second set of investigations, two groups were established within the female mouse cohort. In one group, mice experienced ovariectomy (OVX) to simulate an ovarian hormone-deficient state similar to surgical menopause, while the other group received Sham surgery as controls, to investigate the mechanistic role of ovarian hormone presence in driving immune dysregulation following acute WS exposure. Each experimental cohort followed a consecutive 2-day protocol with daily 4-h exposure intervals under two conditions: control HEPA-filtered air (FA) and acute WS to simulate an acute wildfire episode. RESULTS Metals analysis of WS particulate matter (PM) revealed significantly increased levels of 63Cu, 182W, 208Pb, and 238U, compared to filtered air (FA) controls, providing insights into the specific metal components most impacted by the changing dynamics of wildfire occurrences in the region. Male and female mice exhibited diverse patterns in lung mRNA cytokine expression following WS exposure, with males showing downregulation and females displaying upregulation, notably for IL-1β, TNF-α, CXCL-1, CCL-5, TGF-β, and IL-6. After acute WS exposure, there were notable differences in the responses of macrophages, neutrophils, and bronchoalveolar lavage (BAL) cytokines IL-10, IL-6, IL-1β, and TNF-α. Significant diverse alterations were observed in BAL cytokines, specifically IL-1β, IL-10, IL-6, and TNF-α, as well as in the populations of immune cells, such as macrophages and polymorphonuclear leukocytes, in both Sham and OVX mice, following acute WS exposure. These findings elucidated the profound influence of hormonal changes on inflammatory outcomes, delineating substantial sex-related differences in immune activation and revealing altered immune responses in OVX mice due to ovarian hormone deficiency. In addition, the flow cytometry analysis highlighted the complex interaction between OVX surgery, acute WS exposure, and their collective impact on immune cell populations within the hematopoietic bone marrow niche. CONCLUSIONS In summary, both male and female mice, alongside females subjected to OVX and those who had sham surgery, exhibit significant variations in the expression of proinflammatory cytokines, chemokines, lung mRNA gene expression, and related functional networks linked to signaling pathways. These differences potentially act as mediators of sex-specific and hormonal influences in the systemic inflammatory response to acute WS exposure during a wildfire event. Understanding the regulatory roles of genes expressed differentially under environmental stressors holds considerable implications, aiding in identifying sex-specific therapeutic targets for addressing acute lung inflammation and injury.
Collapse
Affiliation(s)
- Kartika Wardhani
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
- Biochemistry and Biotechnology Group (B-TEK), Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Sydnee Yazzie
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Charlotte McVeigh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Onamma Edeh
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Martha Grimes
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Quiteria Jacquez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Connor Dixson
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Edward Barr
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Alicia M Bolt
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Changjian Feng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Katherine E Zychowski
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA.
| |
Collapse
|
10
|
van der Plaat DA, Lenoir A, Dharmage S, Potts J, Gómez Real F, Shaheen SO, Jarvis D, Minelli C, Leynaert B. Effects of testosterone and sex hormone binding globulin on lung function in males and females: a multivariable Mendelian Randomisation study. Thorax 2024; 79:564-572. [PMID: 38418196 DOI: 10.1136/thorax-2023-220374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 01/12/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND Observational studies suggest that total testosterone (TT) and sex hormone-binding globulin (SHBG) may have beneficial effects on lung function, but these findings might be spurious due to confounding and reverse causation. We addressed these limitations by using multivariable Mendelian randomisation (MVMR) to investigate the independent causal effects of TT and SHBG on lung function. METHODS We first identified genetic instruments by performing genome-wide association analyses of TT and SHBG in the large UK Biobank, separately in males and females. We then assessed the independent effects of TT and SHBG on forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and FEV1/FVC using one-sample MVMR. We addressed pleiotropy, which could bias MVMR, using several methods that account for it. We performed subgroup MVMR analyses by obesity, physical activity and menopausal status, and assessed associations between TT and SHBG with lung function decline. Finally, we compared the MVMR results with those of observational analyses in the UK Biobank. FINDINGS In the MVMR analyses, there was evidence of pleiotropy, but results were consistent when accounting for it. We found a strong beneficial effect of TT on FVC and FEV1 in both males and females, but a moderate detrimental effect of SHBG on FEV1 and FEV1/FVC in males only. Subgroup analyses suggested stronger effects of TT among obese and older males. The observational analyses, in line with previous studies, agreed with MRMV for TT, but not for SHBG. INTERPRETATION These findings suggest that testosterone improves lung function in males and females, while SHBG has an opposite independent effect in males.
Collapse
Affiliation(s)
| | - Alexandra Lenoir
- Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Gesundheitsamt Fürstenfeldbruck, Fürstenfeldbruck, Switzerland
| | - Shyamali Dharmage
- Allergy and Lung Health Unit, The University of Melbourne School of Population and Global Health, Melbourne, Victoria, Australia
| | - James Potts
- National Heart and Lung Institute (NHLI), Imperial College London, London, UK
| | - Francisco Gómez Real
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Seif O Shaheen
- Wolfson Institute of Population Health, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, London, UK
| | - Debbie Jarvis
- National Heart and Lung Institute (NHLI), Imperial College London, London, UK
| | - Cosetta Minelli
- National Heart and Lung Institute (NHLI), Imperial College London, London, UK
| | - Bénédicte Leynaert
- Université Paris-Saclay, UVSQ, Université Paris-Sud, Inserm, Équipe d'Épidémiologie Respiratoire Intégrative, CESP, INSERM, Villejuif, France
| |
Collapse
|
11
|
Liang C, Chung HF, Dobson A, Sandin S, Weiderpass E, Mishra GD. Female reproductive histories and the risk of chronic obstructive pulmonary disease. Thorax 2024; 79:508-514. [PMID: 38350732 DOI: 10.1136/thorax-2023-220388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/17/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Female reproductive factors may influence the development of chronic obstructive pulmonary disease (COPD) through the female hormonal environment, but studies on this topic are limited. This study aimed to assess whether age at menarche, number of children, infertility, miscarriage, stillbirth and age at natural menopause were associated with the risk of COPD. METHODS Women from three cohorts with data on reproductive factors, COPD and covariates were included. Cause specific Cox regression models were adjusted for birth year, race, educational level, body mass index and pack years of smoking, stratified by asthma, and incorporating interaction between birth year and time. Between cohort differences and within cohort correlations were taken into account. RESULTS Overall, 2 83 070 women were included and 10 737 (3.8%) developed COPD after a median follow-up of 11 (IQR 10-12) years. Analyses revealed a U shaped association between age at menarche and COPD (≤11 vs 13: HR 1.17, 95% CI 1.11 to 1.23; ≥16 vs 13: HR 1.24, 95% CI 1.21 to 1.27). Women with three or more children (3 vs 2: HR 1.14, 95% CI 1.12 to 1.17; ≥4 vs 2: HR 1.34, 95% CI 1.28 to 1.40), multiple miscarriages (2 vs 0: HR 1.28, 95% CI 1.24 to 1.32; ≥3 vs 0: HR 1.36, 95% CI 1.30 to 1.43) or stillbirth (1 vs 0: HR 1.38, 95% CI 1.25 to 1.53; ≥2 vs 0: HR 1.67, 95% CI 1.32 to 2.10) were at a higher risk of COPD. Among postmenopausal women, earlier age at natural menopause was associated with an increased risk of COPD (<40 vs 50-51: HR 1.69, 95% CI 1.63 to 1.75; 40-44 vs 50-51: HR 1.42, 95% CI 1.38 to 1.47). CONCLUSIONS Multiple female reproductive factors, including age at menarche, number of children, miscarriage, stillbirth, and age at natural menopause were associated with the risk of COPD.
Collapse
Affiliation(s)
- Chen Liang
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| | - Hsin-Fang Chung
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| | - Annette Dobson
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Gita D Mishra
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Carbajal-García A, Reyes-García J, Díaz-Hernández V, Casas-Hernández MF, Flores-Murrieta FJ, Montaño LM. Testosterone Enhances K V Currents and Airway Smooth Muscle Relaxation Induced by ATP and UTP through P2Y 4 Receptors and Adenylyl Cyclase Pathway. Int J Mol Sci 2024; 25:4652. [PMID: 38731872 PMCID: PMC11083821 DOI: 10.3390/ijms25094652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
Numerous studies suggest the involvement of adenosine-5'-triphosphate (ATP) and similar nucleotides in the pathophysiology of asthma. Androgens, such as testosterone (TES), are proposed to alleviate asthma symptoms in young men. ATP and uridine-5'-triphosphate (UTP) relax the airway smooth muscle (ASM) via purinergic P2Y2 and P2Y4 receptors and K+ channel opening. We previously demonstrated that TES increased the expression of voltage-dependent K+ (KV) channels in ASM. This study investigates how TES may potentiate ASM relaxation induced by ATP and UTP. Tracheal tissues treated with or without TES (control group) from young male guinea pigs were used. In organ baths, tracheas exposed to TES (40 nM for 48 h) showed enhanced ATP- and UTP-evoked relaxation. Tetraethylammonium, a K+ channel blocker, annulled this effect. Patch-clamp experiments in tracheal myocytes showed that TES also increased ATP- and UTP-induced K+ currents, and this effect was abolished with flutamide (an androgen receptor antagonist). KV channels were involved in this phenomenon, which was demonstrated by inhibition with 4-aminopyridine. RB2 (an antagonist of almost all P2Y receptors except for P2Y2), as well as N-ethylmaleimide and SQ 22,536 (inhibitors of G proteins and adenylyl cyclase, respectively), attenuated the enhancement of the K+ currents induced by TES. Immunofluorescence and immunohistochemistry studies revealed that TES did not modify the expression of P2Y4 receptors or COX-1 and COX-2, while we have demonstrated that this androgen augmented the expression of KV1.2 and KV1.5 channels in ASM. Thus, TES leads to the upregulation of P2Y4 signaling and KV channels in guinea pig ASM, enhancing ATP and UTP relaxation responses, which likely limits the severity of bronchospasm in young males.
Collapse
Affiliation(s)
- Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Verónica Díaz-Hernández
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - María F. Casas-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| | - Francisco Javier Flores-Murrieta
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.C.-G.); (J.R.-G.); (M.F.C.-H.)
| |
Collapse
|
13
|
Cao J, Ma Y, Zhao W, Feng C. Age at menarche and idiopathic pulmonary fibrosis: a two-sample mendelian randomization study. BMC Pulm Med 2024; 24:117. [PMID: 38448907 PMCID: PMC10916238 DOI: 10.1186/s12890-024-02936-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/27/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Sex difference in the incidence rate of idiopathic pulmonary fibrosis (IPF) indicates that estrogen has a certain protective effect on the disease. Nevertheless, there is a dearth of study investigating the association between factors pertaining to endogenous estrogen exposure level, such as age at menarche (AAM) in women, and IPF. Our study intended to employ Mendelian randomization (MR) method to elucidate the causal association between AAM and IPF. METHODS Our study utilized AAM as a measure of endogenous estrogen exposure and investigated its causal effect on the risk of IPF through MR. We employed the inverse variance weighted (IVW) method to assess the causal relationship between AAM and IPF risk, with supplementary analyses conducted using the weighted median estimator (WME) and MR-Egger method. Several sensitivity analyses were performed to assess the dependability of MR estimates. RESULTS A total of 9 selected single nucleotide polymorphisms (SNPs) significantly associated with AAM were selected as instrumental variables. The IVW method showed that genetically later AAM was associated with an increased risk of IPF (odds ratio [OR] = 1.0014, 95%confidence interval [CI] = 1.0005-1.0023, p = 0.001). The median weighting method and the MR-Egger method obtained similar estimates, and no heterogeneity or pleiotropy was found, indicating that the results were robust. CONCLUSIONS Our MR study suggested a causal relationship between a later onset of menarche and a heightened susceptibility to IPF.
Collapse
Affiliation(s)
- Jiaqi Cao
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Yazhou Ma
- Department of Neurology, Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Wei Zhao
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Chunlai Feng
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
14
|
Kwon SH, Chung H, Seo JW, Kim HS. Genistein alleviates pulmonary fibrosis by inactivating lung fibroblasts. BMB Rep 2024; 57:143-148. [PMID: 37817434 PMCID: PMC10979345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Pulmonary fibrosis is a serious lung disease that occurs predominantly in men. Genistein is an important natural soybeanderived phytoestrogen that affects various biological functions, such as cell migration and fibrosis. However, the antifibrotic effects of genistein on pulmonary fibrosis are largely unknown. The antifibrotic effects of genistein were evaluated using in vitro and in vivo models of lung fibrosis. Proteomic data were analyzed using nano-LC-ESI-MS/MS. Genistein significantly reduced transforming growth factor (TGF)-β1-induced expression of collagen type I and α-smooth muscle actin (SMA) in MRC-5 cells and primary fibroblasts from patients with idiopathic pulmonary fibrosis (IPF). Genistein also reduced TGF-β1-induced expression of p-Smad2/3 and p-p38 MAPK in fibroblast models. Comprehensive protein analysis confirmed that genistein exerted an anti-fibrotic effect by regulating various molecular mechanisms, such as unfolded protein response, epithelial mesenchymal transition (EMT), mammalian target of rapamycin complex 1 (mTORC1) signaling, cell death, and several metabolic pathways. Genistein was also found to decrease hydroxyproline levels in the lungs of BLM-treated mice. Genistein exerted an anti-fibrotic effect by preventing fibroblast activation, suggesting that genistein could be developed as a pharmacological agent for the prevention and treatment of pulmonary fibrosis. [BMB Reports 2024; 57(3): 143-148].
Collapse
Affiliation(s)
- Seung-hyun Kwon
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Korea
| | - Hyunju Chung
- Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea
| | - Jung-Woo Seo
- Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Korea
| | - Hak Su Kim
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Korea
| |
Collapse
|
15
|
Wu X, Xiao X, Fang H, He C, Wang H, Wang M, Lan P, Wang F, Du Q, Yang H. Elucidating shared biomarkers in gastroesophageal reflux disease and idiopathic pulmonary fibrosis: insights into novel therapeutic targets and the role of angelicae sinensis radix. Front Pharmacol 2024; 15:1348708. [PMID: 38414734 PMCID: PMC10897002 DOI: 10.3389/fphar.2024.1348708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Background: The etiological underpinnings of gastroesophageal reflux disease (GERD) and idiopathic pulmonary fibrosis (IPF) remain elusive, coupled with a scarcity of effective therapeutic interventions for IPF. Angelicae sinensis radix (ASR, also named Danggui) is a Chinese herb with potential anti-fibrotic properties, that holds promise as a therapeutic agent for IPF. Objective: This study seeks to elucidate the causal interplay and potential mechanisms underlying the coexistence of GERD and IPF. Furthermore, it aims to investigate the regulatory effect of ASR on this complex relationship. Methods: A two-sample Mendelian randomization (TSMR) approach was employed to delineate the causal connection between gastroesophageal reflux disease and IPF, with Phennoscanner V2 employed to mitigate confounding factors. Utilizing single nucleotide polymorphism (SNPs) and publicly available microarray data, we analyzed potential targets and mechanisms related to IPF in GERD. Network pharmacology and molecular docking were employed to explore the targets and efficacy of ASR in treating GERD-related IPF. External datasets were subsequently utilized to identify potential diagnostic biomarkers for GERD-related IPF. Results: The IVW analysis demonstrated a positive causal relationship between GERD and IPF (IVW: OR = 1.002, 95%CI: 1.001, 1.003; p < 0.001). Twenty-five shared differentially expressed genes (DEGs) were identified. GO functional analysis revealed enrichment in neural, cellular, and brain development processes, concentrated in chromosomes and plasma membranes, with protein binding and activation involvement. KEGG analysis unveiled enrichment in proteoglycan, ERBB, and neuroactive ligand-receptor interaction pathways in cancer. Protein-protein interaction (PPI) analysis identified seven hub genes. Network pharmacology analysis demonstrated that 104 components of ASR targeted five hub genes (PDE4B, DRD2, ERBB4, ESR1, GRM8), with molecular docking confirming their excellent binding efficiency. GRM8 and ESR1 emerged as potential diagnostic biomarkers for GERD-related IPF (ESR1: AUCGERD = 0.762, AUCIPF = 0.725; GRM8: AUCGERD = 0.717, AUCIPF = 0.908). GRM8 and ESR1 emerged as potential diagnostic biomarkers for GERD-related IPF, validated in external datasets. Conclusion: This study establishes a causal link between GERD and IPF, identifying five key targets and two potential diagnostic biomarkers for GERD-related IPF. ASR exhibits intervention efficacy and favorable binding characteristics, positioning it as a promising candidate for treating GERD-related IPF. The potential regulatory mechanisms may involve cell responses to fibroblast growth factor stimulation and steroidal hormone-mediated signaling pathways.
Collapse
Affiliation(s)
- Xuanyu Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hanyu Fang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Cuifang He
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hanyue Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Miao Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peishu Lan
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Quanyu Du
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Ambhore NS, Balraj P, Pabelick CM, Prakash YS, Sathish V. Estrogen receptors differentially modifies lamellipodial and focal adhesion dynamics in airway smooth muscle cell migration. Mol Cell Endocrinol 2024; 579:112087. [PMID: 37827228 PMCID: PMC10842142 DOI: 10.1016/j.mce.2023.112087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/21/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Sex-steroid signaling, especially estrogen, has a paradoxical impact on regulating airway remodeling. In our previous studies, we demonstrated differential effects of 17β-estradiol (E2) towards estrogen receptors (ERs: α and β) in regulating airway smooth muscle (ASM) cell proliferation and extracellular matrix (ECM) production. However, the role of ERs and their signaling on ASM migration is still unexplored. In this study, we examined how ERα versus ERβ affects the mitogen (Platelet-derived growth factor, PDGF)-induced human ASM cell migration as well as the underlying mechanisms involved. We used Lionheart-FX automated microscopy and transwell assays to measure cell migration and found that activating specific ERs had differential effects on PDGF-induced ASM cell migration. Pharmacological activation of ERβ or shRNA mediated knockdown of ERα and specific activation of ERβ blunted PDGF-induced cell migration. Furthermore, specific ERβ activation showed inhibition of actin polymerization by reducing the F/G-actin ratio. Using Zeiss confocal microscopy coupled with three-dimensional algorithmic ZEN-image analysis showed an ERβ-mediated reduction in PDGF-induced expressions of neural Wiskott-Aldrich syndrome protein (N-WASP) and actin-related proteins-2/3 (Arp2/3) complex, thereby inhibiting actin-branching and lamellipodia. In addition, ERβ activation also reduces the clustering of actin-binding proteins (vinculin and paxillin) at the leading edge of ASM cells. However, cells treated with E2 or ERα agonists do not show significant changes in actin/lamellipodial dynamics. Overall, these findings unveil the significance of ERβ activation in regulating lamellipodial and focal adhesion dynamics to regulate ASM cell migration and could be a novel target to blunt airway remodeling.
Collapse
Affiliation(s)
| | - Premanand Balraj
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
17
|
Liang S, Lu Z, Cai L, Zhu M, Zhou H, Zhang J. Multi-Omics analysis reveals molecular insights into the effects of acute ozone exposure on lung tissues of normal and obese male mice. ENVIRONMENT INTERNATIONAL 2024; 183:108436. [PMID: 38219541 DOI: 10.1016/j.envint.2024.108436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/16/2024]
Abstract
Certain sub-groups, including men and obese individuals, are more susceptible to ozone (O3) exposure, but the underlying molecular mechanisms remain unclear. In this study, the male mice were divided into two dietary groups: one fed a high-fat diet (HFD), mimicking obesity conditions, and the other fed a normal diet (ND), then exposed to 0.5 ppm and 2 ppm O3 for 4 h per day over two days. The HFD mice exhibited significantly higher body weight and serum lipid biochemical indicators compared to the ND mice. Obese mice also exhibited more severe pulmonary inflammation and oxidative stress. Using a multi-omics approach including proteomics, metabolomics, and lipidomics, we observed that O3 exposure induced significant pulmonary molecular changes in both obese and normal mice, primarily arachidonic acid metabolism and lipid metabolism. Different molecular biomarker responses to acute O3 exposure were also observed between two dietary groups, with immune-related proteins impacted in obese mice and PPAR pathway-related proteins affected in normal mice. Furthermore, although not statistically significant, O3 exposure tended to aggravate HFD-induced disturbances in lung glycerophospholipid metabolism. Overall, this study provides valuable molecular insights into the responses of lung to O3 exposure and highlights the potential impact of O3 on obesity-induced metabolic changes.
Collapse
Affiliation(s)
- Shijia Liang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Zhonghua Lu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Lijing Cai
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Miao Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Haixia Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Jie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
18
|
Eliyahu E, Katz MG, Vincek A, Freage-Kahn L, Ravvin S, Tal S, Grage H, Shtraizent N, Barak T, Arkush B. Effects of Hormone Replacement Therapy on Women's Lung Health and Disease. Pulm Ther 2023; 9:461-477. [PMID: 37815696 PMCID: PMC10721592 DOI: 10.1007/s41030-023-00240-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023] Open
Abstract
This review provides an overview of menopausal hormone therapy and pulmonary disease risk, with a focus on the effect of hormone replacement therapy (HRT) on pulmonary function and its relation to lung diseases. This summary is based on authors' knowledge in the field of HRT and supplemented by a PubMed search using the terms "menopause hormone therapy," "asthma", "lung cancer", "chronic obstructive pulmonary disease", "lung function", and "pulmonary hypertension". Available evidence indicates that there is limited research on the role of sex hormones in the susceptibility, severity, and progression of chronic respiratory diseases. However, some studies suggest that the hormonal changes that occur during the menopausal transition may have an impact on pulmonary function and respiratory diseases. Women are in need of convenient access to a safe and effective modality for personalized HRT based on an artificial intelligence (AI)-driven platform that will enable them to receive personalized hormonal treatment through frequent, convenient, and accurate measurements of hormone levels in peripheral blood.
Collapse
Affiliation(s)
- Efrat Eliyahu
- Aveta.Life, Hoboken, NJ, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, Box 1030, New York, NY, 10029-6574, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Michael G Katz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, Box 1030, New York, NY, 10029-6574, USA
- Department of Pediatric Cardiac Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam Vincek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, Box 1030, New York, NY, 10029-6574, USA
| | | | - Shana Ravvin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave, Box 1030, New York, NY, 10029-6574, USA
| | - Smadar Tal
- Department of Animal Sciences, Tel-Hai College, Qiryat Shemona, Israel
| | | | | | | | | |
Collapse
|
19
|
Feng Y, Jiang Y, Zhou Y, Li ZH, Yang QQ, Mo JF, Wen YY, Shen LP. Metabolomics unveils the mechanism of Bufei Huayu decoction in combination with cisplatin against non-small cell lung cancer (NSCLC). Heliyon 2023; 9:e19155. [PMID: 37664700 PMCID: PMC10469573 DOI: 10.1016/j.heliyon.2023.e19155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/28/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023] Open
Abstract
Introduction Bufei Huayu Decoction (BFHY) is a clinical prescription with reported efficacy in enhancing the therapeutic outcomes of chemotherapeutic agents for non-small cell lung cancer (NSCLC). However, the underlying metabolic mechanism of BFHY's action remains unexplored. Objective The objective of this study is to investigate the global metabolic effects of cisplatin and cisplatin plus BFHY on NSCLC. Methods Three groups (NSCLC, cisplatin, and cisplatin + BFHY) underwent a serum metabolomics procedure based on UHPLC-QE-MS. Then, a pathway analysis was carried out using MetaboAnalyst 3.0 to elucidate the therapeutic action routes of cisplatin and cisplatin plus BFHY in NSCLC. Results In the subcutaneous NSCLC model, both cisplatin and cisplatin + BFHY reduced the tumor volume and caused cell death. In comparison to cisplatin alone, cisplatin + BFHY showed a stronger tumor-suppressing impact. Furthermore, the same 16 metabolic signaling pathways were shared by the cisplatin and cisplatin + BFHY treatments. These typical metabolites are mainly involved in amino acid metabolism, lipid mobilization, nucleic acid metabolism and carbohydrate metabolites. Conclusions Potential biomarkers and metabolic networks of cisplatin and cisplatin + BFHY's anti-tumor actions are revealed in our investigation.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Respiratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| | - Ying Jiang
- Department of Neurology, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| | - Ying Zhou
- Department of Radiation Oncology, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| | - Zhan-hua Li
- Department of Respiratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| | - Qi-qian Yang
- Department of Respiratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| | - Jin-feng Mo
- Department of Respiratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| | - Yu-yan Wen
- Department of Respiratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| | - Li-ping Shen
- Department of Respiratory Medicine, Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530011, Guangxi, China
| |
Collapse
|
20
|
Borkar NA, Ambhore NS, Balraj P, Ramakrishnan YS, Sathish V. Kisspeptin regulates airway hyperresponsiveness and remodeling in a mouse model of asthma. J Pathol 2023; 260:339-352. [PMID: 37171283 PMCID: PMC10759912 DOI: 10.1002/path.6086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/06/2023] [Accepted: 03/31/2023] [Indexed: 05/13/2023]
Abstract
Asthma is a multifactorial disease of origin characterized by airway hyperresponsiveness (AHR) and airway remodeling. Several pieces of evidence from other pathologies suggest that Kisspeptins (Kp) regulate cell proliferation, migration, and invasion, mechanisms that are highly relevant to asthma. Our recent in vitro studies show Kp-10 (active peptide of Kp), via its receptor, KISS1R, inhibits human airway smooth muscle cell proliferation. Here, we hypothesize a crucial role for Kp-10 in regulating AHR and airway remodeling in vivo. Utilizing C57BL/6J mice, we assessed the effect of chronic intranasal Kp-10 exposure on mixed allergen (MA)-induced mouse model of asthma. MA-challenged mice showed significant deterioration of lung function compared to those exposed to vehicle (DPBS); Kp-10 treatment significantly improved the MA-altered lung functions. Mice treated with Kp-10 alone did not show any notable changes in lung functions. MA-exposed mice showed a significant reduction in KISS1R expression as compared to vehicle alone. MA-challenged mice showed significant alterations in immune cell infiltration in the airways and remodeling changes. Proinflammatory cytokines were significantly increased upon MA exposure, an effect abrogated by Kp-10 treatment. Furthermore, biochemical and histological studies showed Kp-10 exposure significantly reduced MA-induced smooth muscle mass and soluble collagen in the lung. Overall, our findings highlight the effect of chronic Kp-10 exposure in regulating MA-induced AHR and remodeling. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | | | | | | | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| |
Collapse
|
21
|
Romero-Martínez BS, Sommer B, Solís-Chagoyán H, Calixto E, Aquino-Gálvez A, Jaimez R, Gomez-Verjan JC, González-Avila G, Flores-Soto E, Montaño LM. Estrogenic Modulation of Ionic Channels, Pumps and Exchangers in Airway Smooth Muscle. Int J Mol Sci 2023; 24:ijms24097879. [PMID: 37175587 PMCID: PMC10178541 DOI: 10.3390/ijms24097879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 05/15/2023] Open
Abstract
To preserve ionic homeostasis (primarily Ca2+, K+, Na+, and Cl-), in the airway smooth muscle (ASM) numerous transporters (channels, exchangers, and pumps) regulate the influx and efflux of these ions. Many of intracellular processes depend on continuous ionic permeation, including exocytosis, contraction, metabolism, transcription, fecundation, proliferation, and apoptosis. These mechanisms are precisely regulated, for instance, through hormonal activity. The lipophilic nature of steroidal hormones allows their free transit into the cell where, in most cases, they occupy their cognate receptor to generate genomic actions. In the sense, estrogens can stimulate development, proliferation, migration, and survival of target cells, including in lung physiology. Non-genomic actions on the other hand do not imply estrogen's intracellular receptor occupation, nor do they initiate transcription and are mostly immediate to the stimulus. Among estrogen's non genomic responses regulation of calcium homeostasis and contraction and relaxation processes play paramount roles in ASM. On the other hand, disruption of calcium homeostasis has been closely associated with some ASM pathological mechanism. Thus, this paper intends to summarize the effects of estrogen on ionic handling proteins in ASM. The considerable diversity, range and power of estrogens regulates ionic homeostasis through genomic and non-genomic mechanisms.
Collapse
Affiliation(s)
- Bianca S Romero-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Bettina Sommer
- Laboratorio de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Ciudad de México 14080, Mexico
| | - Héctor Solís-Chagoyán
- Neurociencia Cognitiva Evolutiva, Centro de Investigación en Ciencias Cognitivas, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico
| | - Eduardo Calixto
- Departamento de Neurobiología, Dirección de Investigación en Neurociencias, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Ciudad de México 14370, Mexico
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México City 14080, Mexico
| | - Ruth Jaimez
- Laboratorio de Estrógenos y Hemostasis, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Juan C Gomez-Verjan
- Dirección de Investigación, Instituto Nacional de Geriatría (INGER), Ciudad de México 10200, Mexico
| | - Georgina González-Avila
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", México City 14080, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
22
|
Miller RAJ, Williams AP, Kovats S. Sex chromosome complement and sex steroid signaling underlie sex differences in immunity to respiratory virus infection. Front Pharmacol 2023; 14:1150282. [PMID: 37063266 PMCID: PMC10097973 DOI: 10.3389/fphar.2023.1150282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/08/2023] [Indexed: 04/18/2023] Open
Abstract
Epidemiological studies have revealed sex differences in the incidence and morbidity of respiratory virus infection in the human population, and often these observations are correlated with sex differences in the quality or magnitude of the immune response. Sex differences in immunity and morbidity also are observed in animal models of respiratory virus infection, suggesting differential dominance of specific immune mechanisms. Emerging research shows intrinsic sex differences in immune cell transcriptomes, epigenomes, and proteomes that may regulate human immunity when challenged by viral infection. Here, we highlight recent research into the role(s) of sex steroids and X chromosome complement in immune cells and describe how these findings provide insight into immunity during respiratory virus infection. We focus on the regulation of innate and adaptive immune cells by receptors for androgen and estrogens, as well as genes with a propensity to escape X chromosome inactivation. A deeper mechanistic knowledge of these pathways will help us to understand the often significant sex differences in immunity to endemic or pandemic respiratory pathogens such as influenza viruses, respiratory syncytial viruses and pathogenic coronaviruses.
Collapse
Affiliation(s)
- Reegan A. J. Miller
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Abigael P. Williams
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Susan Kovats
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
23
|
Ford ML, Ruwanpathirana A, Lewis BW, Britt RD. Aging-Related Mechanisms Contribute to Corticosteroid Insensitivity in Elderly Asthma. Int J Mol Sci 2023; 24:6347. [PMID: 37047327 PMCID: PMC10093993 DOI: 10.3390/ijms24076347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Asthma in elderly populations is an increasing health problem that is accompanied by diminished lung function and frequent exacerbations. As potent anti-inflammatory drugs, corticosteroids are commonly used to reduce lung inflammation, improve lung function, and manage disease symptoms in asthma. Although effective for most individuals, older patients are more insensitive to corticosteroids, making it difficult to manage asthma in this population. With the number of individuals older than 65 continuing to increase, it is important to understand the distinct mechanisms that promote corticosteroid insensitivity in the aging lung. In this review, we discuss corticosteroid insensitivity in asthma with an emphasis on mechanisms that contribute to persistent inflammation and diminished lung function in older individuals.
Collapse
Affiliation(s)
- Maria L. Ford
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA; (M.L.F.); (A.R.)
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anushka Ruwanpathirana
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA; (M.L.F.); (A.R.)
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Brandon W. Lewis
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA; (M.L.F.); (A.R.)
| | - Rodney D. Britt
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA; (M.L.F.); (A.R.)
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
24
|
Zhou R, Jin S, Jin C, Sun Y, Zhang L, Yan J, Jiang H. Association of urinary bisphenol A with chronic obstructive pulmonary disease-related diseases: evidence from the National Health and Nutrition Examination Survey database (2005-2016). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:33170-33180. [PMID: 36474039 DOI: 10.1007/s11356-022-24572-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/30/2022] [Indexed: 06/17/2023]
Abstract
In vivo and in vitro studies have found that exposure to bisphenol A (BPA) can lead to pulmonary diseases, but there exists little evidence regarding the association between urinary BPA level and chronic obstructive pulmonary disease (COPD)-related diseases in the population. National Health and Nutrition Examination Survey data, from 2005 to 2016, were utilized in this study. Participants who self-reported having emphysema, chronic bronchitis, or COPD were defined as having COPD-related diseases. A multivariate logistic regression analysis was used to analyze the association of urinary BPA with COPD-related diseases in the overall population and according to sex. Three separate models including different covariates were used in our analyses. The association of urinary BPA with COPD-related diseases in different subgroups (age, smoking status, participants belonging to "ever had asthma" and "ever had cardiovascular disease") other than sex was also analyzed. Based on the different models, 9189, 7006, and 6946 participants were used in our study. BPA was found to be significantly associated with COPD-related diseases in all models. The concentration of BPA in urine was significantly associated with COPD-related diseases regardless of whether BPA concentration was treated as a continuous variable (odds ratio (OR): 1.24, 95% confidence interval (CI): 1.11-1.38, in model 3) or as tertiles (OR: 2.01, 95% CI: 1.49-2.72, between the lowest group and highest group, in model 3). This association was significant among male but not female participants. BPA also demonstrated a significant association with emphysema and chronic bronchitis in adults, particularly in males. No significant interaction was found for all the other subgroup analyses. Urinary BPA was associated with COPD-related diseases in adult participants, especially males.
Collapse
Affiliation(s)
- Ren Zhou
- Department of Anaesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Shanliang Jin
- Department of Anaesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Chenyu Jin
- Department of Anaesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Yu Sun
- Department of Anaesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Lei Zhang
- Department of Anaesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Jia Yan
- Department of Anaesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Hong Jiang
- Department of Anaesthesiology, The Ninth People's Hospital of Shanghai, Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
25
|
Warren KJ, Deering-Rice C, Huecksteadt T, Trivedi S, Venosa A, Reilly C, Sanders K, Clayton F, Wyatt TA, Poole JA, Heller NM, Leung D, Paine R. Steady-state estradiol triggers a unique innate immune response to allergen resulting in increased airway resistance. Biol Sex Differ 2023; 14:2. [PMID: 36609358 PMCID: PMC9817388 DOI: 10.1186/s13293-022-00483-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/11/2022] [Indexed: 01/07/2023] Open
Abstract
RATIONALE Asthma is a chronic airway condition that occurs more often in women than men during reproductive years. Population studies have collectively shown that long-term use of oral contraceptives decreased the onset of asthma in women of reproductive age. In the current study, we hypothesized that steady-state levels of estrogen would reduce airway inflammation and airway hyperresponsiveness to methacholine challenge. METHODS Ovariectomized BALB/c mice (Ovx) were implanted with subcutaneous hormone pellets (estrogen, OVX-E2) that deliver consistent levels of estrogen [68 ± 2 pg/mL], or placebo pellets (OVX-Placebo), followed by ovalbumin sensitization and challenge. In conjunction with methacholine challenge, immune phenotyping was performed to correlate inflammatory proteins and immune populations with better or worse pulmonary outcomes measured by invasive pulmonary mechanics techniques. RESULTS Histologic analysis showed an increase in total cell infiltration and mucus staining around the airways leading to an increased inflammatory score in ovarectomized (OVX) animals with steady-state estrogen pellets (OVX-E2-OVA) as compared to other groups including female-sham operated (F-INTACT-OVA) and OVX implanted with a placebo pellet (OVX-Pl-OVA). Airway resistance (Rrs) and lung elastance (Ers) were increased in OVX-E2-OVA in comparison to F-INTACT-OVA following aerosolized intratracheal methacholine challenges. Immune phenotyping revealed that steady-state estrogen reduced CD3+ T cells, CD19+ B cells, ILC2 and eosinophils in the BAL across all experiments. While these commonly described allergic cells were reduced in the BAL, or airways, we found no changes in neutrophils, CD3+ T cells or CD19+ B cells in the remaining lung tissue. Similarly, inflammatory cytokines (IL-5 and IL-13) were also decreased in OVX-E2-OVA-treated animals in comparison to Female-INTACT-OVA mice in the BAL, but in the lung tissue IL-5, IL-13 and IL-33 were comparable in OVX-E2-OVA and F-INTACT OVA mice. ILC2 were sorted from the lungs and stimulated with exogenous IL-33. These ILC2 had reduced cytokine and chemokine expression when they were isolated from OVX-E2-OVA animals, indicating that steady-state estrogen suppresses IL-33-mediated activation of ILC2. CONCLUSIONS Therapeutically targeting estrogen receptors may have a limiting effect on eosinophils, ILC2 and potentially other immune populations that may improve asthma symptoms in those females that experience perimenstrual worsening of asthma, with the caveat, that long-term use of estrogens or hormone receptor modulators may be detrimental to the lung microenvironment over time.
Collapse
Affiliation(s)
- Kristi J Warren
- George E Wahlen Salt Lake City VA Medical Center, 500 Foothill Dr., Salt Lake City, UT, USA.
- The Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.
- Division of Allergy and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Cassandra Deering-Rice
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Tom Huecksteadt
- George E Wahlen Salt Lake City VA Medical Center, 500 Foothill Dr., Salt Lake City, UT, USA
| | - Shubhanshi Trivedi
- George E Wahlen Salt Lake City VA Medical Center, 500 Foothill Dr., Salt Lake City, UT, USA
- The Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- Division of Allergy and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Christopher Reilly
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Karl Sanders
- George E Wahlen Salt Lake City VA Medical Center, 500 Foothill Dr., Salt Lake City, UT, USA
- The Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- Division of Allergy and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Frederic Clayton
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Todd A Wyatt
- Division of Allergy and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Jill A Poole
- Division of Allergy and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, USA
| | - Daniel Leung
- The Division of Infectious Diseases, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Robert Paine
- George E Wahlen Salt Lake City VA Medical Center, 500 Foothill Dr., Salt Lake City, UT, USA
- The Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
26
|
Micera A, Di Zazzo A, De Piano M, Sharma S, Mori T, De Gregorio C, Coassin M, Fernandes M. Tissue remodeling in adult vernal keratoconjunctivitis. Exp Eye Res 2022; 225:109301. [PMID: 36336099 DOI: 10.1016/j.exer.2022.109301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/30/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Our aim is to describe local tissue remodeling in a cohort of adult VKC patients. Male patients diagnosed with active VKC were enrolled in an open pilot study into two groups according disease onset: childhood classic VKC and adult VKC. Visual acuity and ocular surface clinical examination focusing on chronic inflammatory sequelae and impression cytology were performed in all enrolled subjects. Conjunctival imprints were processed for molecular, biochemical and immunofluorescent analysis for tissue remodeling (TGFβ1,2,3 and αSMA) and epigenetic (DNMT3a, Keap1; Nrf2) markers as well as androgen receptors were investigated and compared between groups. Clinical assessment showed increased conjunctival scarring in adult VKC compared to classic VKC. Immunoreactivity for αSMA and expression of TGFβ were higher in adult VKC group. Significantly higher levels of TGFβ3 (3.44 ± 1.66; p < 0.05) were detected in adult VKC compared to childhood VKC, associated with an increasing trend of TGFβ1 (1.58 ± 0.25) and TGFβ2 (1.65 ± 0.20) isoforms levels. Molecular analysis showed a relative increase in tissue remodeling/fibrogenic transcripts (TGFβ isoforms and αSMA) associated to a significant increase of selective epigenetic targets (DNMT3, Nrf2 and keap1) in adult VKC phenotype. Increased local conjunctival androgen receptors was detected in patients with adult variants compared to classic childhood VKC and healthy subjects. Finally, a direct correlation between TGFβ and androgen receptor expression was also detected. A pro-fibrotic clinical and biomolecular trait was unveiled in adult variant of VKC, which causes ocular surface disease and visual impairment.
Collapse
Affiliation(s)
- Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS - Fondazione Bietti, Rome, Italy
| | - Antonio Di Zazzo
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Maria De Piano
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS - Fondazione Bietti, Rome, Italy
| | - Savitri Sharma
- Jhaveri Microbiology Centre, LV Prasad Eye Institute, Kallam Anji Reddy Campus, Hyderabad, India
| | - Tommaso Mori
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Chiara De Gregorio
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Marco Coassin
- Ophthalmology Operative Complex Unit, University Campus Bio-Medico, Rome, Italy
| | - Merle Fernandes
- Cornea and Anterior Segment Services, LV Prasad Eye Institute, GMR Varalakshmi Campus, Visakhapatnam, India.
| |
Collapse
|
27
|
Nguyen TT, Smith CY, Gazzuola Rocca L, Rocca WA, Vassallo R, Dulohery Scrodin MM. A population-based cohort study on the risk of obstructive lung disease after bilateral oophorectomy. NPJ Prim Care Respir Med 2022; 32:52. [PMCID: PMC9663719 DOI: 10.1038/s41533-022-00317-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
AbstractThere is increasing evidence that sex hormones may impact the development of obstructive lung disease (OLD). Therefore, we studied the effect of bilateral oophorectomy (oophorectomy) on the development of OLD. Women were identified from the Mayo Clinic Cohort Study of Oophorectomy and Aging-2. Data were collected using the Rochester Epidemiology Project records-linkage system. A total of 1653 women who underwent oophorectomy and 1653 referent women of similar age were assessed for OLD using diagnostic codes and medical record abstraction. Women who underwent oophorectomy had an overall higher risk of all OLD, all chronic obstructive pulmonary disease (COPD), emphysema, and chronic bronchitis but not of all asthma, confirmed asthma, or confirmed COPD. The association with all OLD was stronger in women who were age ≤45 years at oophorectomy, never smokers, non-obese, and in women with benign indications; however, the interactions were not statistically significant. There was an increased risk of all asthma in women age ≤45 years at oophorectomy who took estrogen therapy. Never smokers of all ages had a stronger association of oophorectomy with all asthma and all COPD, whereas smokers had a stronger association of oophorectomy with emphysema and chronic bronchitis. Non-obese women of all ages had a stronger association of oophorectomy with all COPD, emphysema, and chronic bronchitis. The results of this study combined with the increased risk of several chronic diseases reported in previous studies suggest that oophorectomy in premenopausal women should be avoided unless there is clear evidence of a high genetic risk of ovarian cancer.
Collapse
|
28
|
Pilling D, Sahlberg K, Chen W, Gomer RH. Changes in lung sialidases in male and female mice after bleomycin aspiration. Exp Lung Res 2022; 48:291-304. [PMID: 36382835 PMCID: PMC10084762 DOI: 10.1080/01902148.2022.2144548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/17/2022]
Abstract
Aim of the study: Sialidases, also called neuraminidases, are enzymes that cleave terminal sialic acids from glycoconjugates. In humans and mice, lung fibrosis is associated with desialylation of glycoconjugates and upregulation of sialidases. There are four mammalian sialidases, and it is unclear when the four mammalian sialidases are elevated over the course of inflammatory and fibrotic responses, whether tissue resident and inflammatory cells express different sialidases, and if sialidases are differentially expressed in male and females. Materials and Methods: To determine the time course of sialidase expression and the identity of sialidase expressing cells, we used the bleomycin model of pulmonary fibrosis in mice to examine levels of sialidases during inflammation (days 3 - 10) and fibrosis (days 10 - 21). Results: Bleomycin aspiration increased sialidase NEU1 at days 14 and 21 in male mice and day 10 in female mice. NEU2 levels increased at day 7 in male and day 10 in female mice. NEU3 appears to have a biphasic response in male mice with increased levels at day 7 and then at days 14 and 21, whereas in female mice NEU3 levels increased over 21 days. In control mice, the sialidases were mainly expressed by EpCAM positive epithelial cells, but after bleomycin, epithelial cells, CD45 positive immune cells, and alveolar cells expressed NEU1, NEU2, and NEU3. Sialidase expression was higher in male compared to female mice. There was little expression of NEU4 in murine lung tissue. Conclusions: These results suggest that sialidases are dynamically expressed following bleomycin, that sialidases are differentially expressed in male and females, and that of the four sialidases only NEU3 upregulation is associated with fibrosis in both male and female mice.
Collapse
|
29
|
Debieuvre D, Molinier O, Falchero L, Locher C, Templement-Grangerat D, Meyer N, Morel H, Duval Y, Asselain B, Letierce A, Trédaniel J, Auliac JB, Bylicki O, Moreau L, Fore M, Corre R, Couraud S, Cortot A. Lung cancer trends and tumor characteristic changes over 20 years (2000–2020): Results of three French consecutive nationwide prospective cohorts’ studies. Lancet Reg Health Eur 2022; 22:100492. [PMID: 36108315 PMCID: PMC9445429 DOI: 10.1016/j.lanepe.2022.100492] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Long-term changes in lung cancer (LC) patients are difficult to evaluate. We report results from the French KBP-2020 real-life cohort. Methods KBP-2020 was a prospective cohort that included all patients diagnosed with LC in 2020, in nonacademic public hospital in France. Patient and tumour characteristics were described and compared with similarly designed cohorts in 2000 and 2010. Findings In 2020, 82 centers included 8,999 patients diagnosed with LC. The proportion of women increased: 34·6% (3114/8999) compared to, 24·3% (1711/7051) and 16·0% (904/5667) in 2010 and 2000 (p<0·0001). The proportion of non-smokers was higher in 2020 (12·6%, 1129/8983) than in previous cohorts (10·9% (762/7008) in 2010; 7·2% (402/5586) in 2000, p<0·0001). In 2020, at diagnosis, 57·6% (4405/7648) of patients had a metastatic/disseminated stage non-small-cell lung cancer (NSCLC) (58·3% (3522/6046) in 2010; 42·6% (1879/4411) in 2000, p<0·0001). Compared with 2000 and 2010 data, early survival improved slightly. In 2020, 3-month mortality of NSCLC varied from 3·0% [2·2 – 3·8] for localized to 9·6% [8·1 – 11·0] for locally advanced to 29·2% [27·8 – 30·6] for metastatic and was 24·8% [22·3 – 27·3] for SCLC. Interpretation To our knowledge KBP cohorts have been the largest, prospective, real-world cohort studies involving LC patients conducted in worldwide. The trend found in our study shows an increase in LC in women and still a large proportion of patients diagnosed at metastatic or disseminated stage. Funding The study was promoted by the French College of General Hospital Pulmonologists with financial support of industrials laboratories.
Collapse
Affiliation(s)
- Didier Debieuvre
- Respiratory Medicine Department, Groupe Hospitalier de la Région Mulhouse Sud-Alsace, Hôpital Emile Muller, Mulhouse, France
- Corresponding author at: Service de Pneumologie, GHRMSA, Hôpital Emile Muller, 20 rue du Dr Laënnec, BP 1370, 68070 Mulhouse CEDEX, France.
| | - Olivier Molinier
- Respiratory Medicine Department, Centre Hospitalier Le Mans, Le Mans, France
| | - Lionel Falchero
- Respiratory Medicine Department, L'Hôpital Nord-Ouest, Villefranche-Sur-Saône, France
| | - Chrystèle Locher
- Respiratory Medicine Department, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| | | | - Nicolas Meyer
- Biostatistician, Public Health Department, CHU de Strasbourg, GMRC, Strasbourg, France
| | - Hugues Morel
- Respiratory Medicine Department, Centre Hospitalier Régional D'Orléans Hôpital de La Source, Orléans, France
| | - Yannick Duval
- Respiratory Medicine Department, Hôpital de Cannes Simone Veil, Cannes, France
| | - Bernard Asselain
- Methodologist, Groupe Statistique, ARCAGY - GINECO, Paris, France
| | | | - Jean Trédaniel
- Department of Thoracic Oncology, Groupe hospitalier Paris-Saint Joseph, Paris, France
| | - Jean-Bernard Auliac
- Respiratory Medicine Department, Centre Hospitalier Intercommunal Créteil, Créteil, France
| | - Olivier Bylicki
- Respiratory Medicine Department, Hôpital d'Instruction des Armées Sainte-Anne, Toulon, France
| | - Lionel Moreau
- Respiratory Medicine Department, Centre Hospitalier de Colmar, Colmar, France
| | - Mathieu Fore
- Respiratory Medicine Department, Groupe Hospitalier de la Région Mulhouse Sud-Alsace, Hôpital Emile Muller, Mulhouse, France
| | - Romain Corre
- Respiratory Medicine Department, Centre Hospitalier de Cornouaille, Quimper, France
| | - Sébastien Couraud
- Respiratory Medicine Department, Centre Hospitalier de Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Alexis Cortot
- Department of Thoracic Oncology, CHU de Lille, CNRS, Inserm, Institut Pasteur de Lille, UMR9020-U1277-CANTHER, Lille, France
| | | |
Collapse
|
30
|
Sehsah R, Wu W, Ichihara S, Hashimoto N, Zong C, Yamazaki K, Sato H, Itoh K, Yamamoto M, Elsayed AA, El-Bestar S, Kamel E, Ichihara G. Protective role of Nrf2 in zinc oxide nanoparticles-induced lung inflammation in female mice and sexual dimorphism in susceptibility. Toxicol Lett 2022; 370:24-34. [PMID: 36100149 DOI: 10.1016/j.toxlet.2022.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/14/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Zinc oxide nanoparticles (ZnO-NPs) are currently employed in various products such as rubber, paint, and cosmetics. Our group reported recently that Nrf2 protein provides protection against pulmonary inflammation induced by ZnO-NPs in male mice. The current study investigated the effect of Nrf2 deletion on the lung inflammatory response in female mice exposed to ZnO-NPs. METHODS An equal number of female Nrf2-/- mice and female Nrf2+/+ mice (24 each) were allocated into three equal groups, and each was exposed to ZnO-NPs at either 0, 10 or 30 µg ZnO-NPs/mouse through pharyngeal aspiration. Bronchoalveolar lavage fluid (BALF) and lungs were examined 14 days later to determine the number of inflammatory cells, the protein level, and for scoring inflammation histopathologically. The mRNA levels of Nrf2-dependent antioxidant enzymes and proinflammatory cytokine in lung tissue were also measured. RESULTS Exposure to ZnO-NPs increased all types of BALF cells and lung inflammation scores in both of female Nrf2-null (Nrf2-/-) and wild-type (Nrf2+/+) mice, and Nrf2 deletion enhanced ZnO-NPs-induced increase in the number of eosinophils in BALF. Exposure to ZnO-NPs dose-dependently increased the level of oxidized glutathione (GSSG), and mRNA levels of proinflammatory cytokines/chemokines; KC, MIP-2, IL-6, IL-1β and MCP-1 only in wild-type mice. Nrf2 deletion decreased total glutathione levels and basal mRNA levels of SOD1 and NQO1, and increased the basal mRNA level of above proinflammatory cytokines/chemokines. Nrf2 deletion enhanced ZnO-NPs-induced downregulation of GcLc, GR and TGF-β and upregulation of HO-1 and TNF-α. Taken together with our previous results in male mice, our results showed a lower susceptibility of females to lung tissue inflammation, relative to males, irrespective of Nrf2 deletion, and that enhancement of ZnO-NPs-induced upregulation of HO-1 and TNF-α and downregulation of GcLc, GR and TGF-β by deletion of Nrf2 is specific to female mice. CONCLUSION We conclude that Nrf2 provides protection in female mice against increase in BALF eosinophils, probably through down-regulation of proinflammatory cytokines/chemokines and upregulation of oxidative stress-related genes. The study also suggests lower susceptibility to lung tissue inflammation in female mice relative to their male counterparts and the synergistic effects of Nrf2 and exposure to ZnO-NPs on mRNA expression of GcLc, GR, HO-1, TGF-β or TNF-α in female mice.
Collapse
Affiliation(s)
- Radwa Sehsah
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Public Health and Community Medicine, Mansoura Faculty of Medicine, Mansoura, Egypt.
| | - Wenting Wu
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University, Shimotsuke, Japan.
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Cai Zong
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.
| | - Kyoka Yamazaki
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.
| | - Harue Sato
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.
| | - Ken Itoh
- Department of Stress Response Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.
| | - Masayuki Yamamoto
- Department of Molecular Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Ahmed Ali Elsayed
- Department of Pathology, Mansoura Faculty of Medicine, Mansoura, Egypt.
| | - Soheir El-Bestar
- Department of Public Health and Community Medicine, Mansoura Faculty of Medicine, Mansoura, Egypt.
| | - Emily Kamel
- Department of Public Health and Community Medicine, Mansoura Faculty of Medicine, Mansoura, Egypt.
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan; Center for Health Management, Tokyo University of Science, Shinjuku, Tokyo.
| |
Collapse
|
31
|
Sex Steroids Effects on Asthma: A Network Perspective of Immune and Airway Cells. Cells 2022; 11:cells11142238. [PMID: 35883681 PMCID: PMC9318292 DOI: 10.3390/cells11142238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/17/2022] Open
Abstract
A multitude of evidence has suggested the differential incidence, prevalence and severity of asthma between males and females. A compilation of recent literature recognized sex differences as a significant non-modifiable risk factor in asthma pathogenesis. Understanding the cellular and mechanistic basis of sex differences remains complex and the pivotal point of this ever elusive quest, which remains to be clarified in the current scenario. Sex steroids are an integral part of human development and evolution while also playing a critical role in the conditioning of the immune system and thereby influencing the function of peripheral organs. Classical perspectives suggest a pre-defined effect of sex steroids, generalizing estrogens popularly under the “estrogen paradox” due to conflicting reports associating estrogen with a pro- and anti-inflammatory role. On the other hand, androgens are classified as “anti-inflammatory,” serving a protective role in mitigating inflammation. Although considered mainstream and simplistic, this observation remains valid for numerous reasons, as elaborated in the current review. Women appear immune-favored with stronger and more responsive immune elements than men. However, the remarkable female predominance of diverse autoimmune and allergic diseases contradicts this observation suggesting that hormonal differences between the sexes might modulate the normal and dysfunctional regulation of the immune system. This review illustrates the potential relationship between key elements of the immune cell system and their interplay with sex steroids, relevant to structural cells in the pathophysiology of asthma and many other lung diseases. Here, we discuss established and emerging paradigms in the clarification of observed sex differences in asthma in the context of the immune system, which will deepen our understanding of asthma etiopathology.
Collapse
|
32
|
Zhang B, Gao D, Xu G, Zhu W, Liu J, Sun R, Wang L, Zhang C, Ding Q, Shi Y. Integrated multicomponent analysis based on UHPLC-Q-Exactive Orbitrap-MS and network pharmacology to elucidate the potential mechanism of Baoyuan decoction against idiopathic pulmonary fibrosis. PHYTOCHEMICAL ANALYSIS : PCA 2022; 33:678-695. [PMID: 35396886 DOI: 10.1002/pca.3120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a serious lung disease with a high mortality rate. Baoyuan decoction (BYD), a classic medicinal food homology recipe, has anti-apoptotic effects, enhances immune function, and alleviates fibrosis, suggesting that it may be a potential therapeutic drug for IPF. OBJECTIVES We aimed to identify the main active ingredients of BYD, determine the basis of its efficacy, prove its anti-IPF effects, and explore the mechanisms underlying its anti-IPF effects. MATERIALS AND METHODS In this study, the active components of BYD were detected and analysed by ultra-high-performance liquid chromatography coupled with hybrid quadrupole Orbitrap mass spectrometry (UHPLC-Q-Exactive Orbitrap-MS). A network pharmacology analysis was performed to determine the potential targets and relevant pathways of BYD in treating IPF. Western blotting and quantitative real-time polymerase chain reaction (qPCR) were conducted to verify the efficacy of BYD against IPF. Finally, molecular docking and qPCR were performed to identify the central targets of BYD. RESULTS A total of 39 components of BYD were identified. After performing the network pharmacology analysis, 35 active components and eight presumptive targets of BYD were found to play a central role in its anti-IPF effects. The molecular docking results indicated that most of the active components of BYD exhibited good binding activity with these eight central target proteins. In addition, the expression of collagen, α-SMA, and these eight targets in human pulmonary fibroblast (HPF) cells was suppressed from treatment with BYD. CONCLUSION This study determined the efficacy of BYD against IPF and clarified its multiple-target and multiple-pathway mechanisms. Furthermore, the study also provides a new method for exploring the chemical and pharmacological bases of other traditional Chinese medicine (TCM).
Collapse
Affiliation(s)
- Binbin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Dongyang Gao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Gonghao Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Wenxiang Zhu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Jing Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rui Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chen Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
33
|
Ciereszko A, Dietrich MA, Słowińska M, Nynca J, Ciborowski M, Kaczmarek MM, Myszczyński K, Kiśluk J, Majewska A, Michalska-Falkowska A, Kodzik N, Reszeć J, Sierko E, Nikliński J. Application of two-dimensional difference gel electrophoresis to identify protein changes between center, margin, and adjacent non-tumor tissues obtained from non-small-cell lung cancer with adenocarcinoma or squamous cell carcinoma subtype. PLoS One 2022; 17:e0268073. [PMID: 35512017 PMCID: PMC9071164 DOI: 10.1371/journal.pone.0268073] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is responsible for the most cancer-related mortality worldwide and the mechanism of its development is poorly understood. Proteomics has become a powerful tool offering vital knowledge related to cancer development. Using a two-dimensional difference gel electrophoresis (2D-DIGE) approach, we sought to compare tissue samples from non-small-cell lung cancer (NSCLC) patients taken from the tumor center and tumor margin. Two subtypes of NSCLC, adenocarcinoma (ADC) and squamous cell carcinoma (SCC) were compared. Data are available via ProteomeXchange with identifier PXD032736 and PXD032962 for ADC and SCC, respectively. For ADC proteins, 26 significant canonical pathways were identified, including Rho signaling pathways, a semaphorin neuronal repulsive signaling pathway, and epithelial adherens junction signaling. For SCC proteins, nine significant canonical pathways were identified, including hypoxia-inducible factor-1α signaling, thyroid hormone biosynthesis, and phagosome maturation. Proteins differentiating the tumor center and tumor margin were linked to cancer invasion and progression, including cell migration, adhesion and invasion, cytoskeletal structure, protein folding, anaerobic metabolism, tumor angiogenesis, EMC transition, epithelial adherens junctions, and inflammatory responses. In conclusion, we identified several proteins that are important for the better characterization of tumor development and molecular specificity of both lung cancer subtypes. We also identified proteins that may be important as biomarkers and/or targets for anticancer therapy.
Collapse
Affiliation(s)
- Andrzej Ciereszko
- Department of Gametes and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- * E-mail:
| | - Mariola A. Dietrich
- Department of Gametes and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Mariola Słowińska
- Department of Gametes and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Nynca
- Department of Gametes and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Michał Ciborowski
- Metabolomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Monika M. Kaczmarek
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research Polish Academy of Sciences, Olsztyn, Poland
| | - Kamil Myszczyński
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Kiśluk
- Department of Clinical Molecular Biology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Majewska
- Department of Gametes and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | | | - Natalia Kodzik
- Department of Gametes and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Reszeć
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Sierko
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Jacek Nikliński
- Department of Clinical Molecular Biology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
34
|
Modulation of innate immune response to viruses including SARS-CoV-2 by progesterone. Signal Transduct Target Ther 2022; 7:137. [PMID: 35468896 PMCID: PMC9035769 DOI: 10.1038/s41392-022-00981-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/14/2022] Open
Abstract
Whether and how innate antiviral response is regulated by humoral metabolism remains enigmatic. We show that viral infection induces progesterone via the hypothalamic-pituitary-adrenal axis in mice. Progesterone induces downstream antiviral genes and promotes innate antiviral response in cells and mice, whereas knockout of the progesterone receptor PGR has opposite effects. Mechanistically, stimulation of PGR by progesterone activates the tyrosine kinase SRC, which phosphorylates the transcriptional factor IRF3 at Y107, leading to its activation and induction of antiviral genes. SARS-CoV-2-infected patients have increased progesterone levels, and which are co-related with decreased severity of COVID-19. Our findings reveal how progesterone modulates host innate antiviral response, and point to progesterone as a potential immunomodulatory reagent for infectious and inflammatory diseases.
Collapse
|
35
|
Borkar NA, Ambhore NS, Kalidhindi RSR, Pabelick CM, Prakash YS, Sathish V. Kisspeptins inhibit human airway smooth muscle proliferation. JCI Insight 2022; 7:152762. [PMID: 35420998 DOI: 10.1172/jci.insight.152762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Sex/gender disparity in asthma is recognized, and suggests a modulatory role for sex-steroids, particularly estrogen. However, studies including our own show a dichotomous role for estrogen in airway remodeling, making it unclear whether sex hormones are protective or detrimental in asthma, and suggesting a need to explore mechanisms upstream or independent of estrogen. We hypothesize that Kisspeptin (Kp)/KISS1R signaling serves this role. Airway smooth muscle (ASM) is a key structural cell type that contributes to remodeling in asthma. We explored the role of Kp/KISS1R in regulating ASM proliferation. We report novel data that Kp and KISS1R are expressed in human airways, especially ASM, with lower expression in ASM from females compared to males, and asthmatics showing lowest expression compared to non-asthmatics. Proliferation studies showed that cleaved forms of Kp, particularly Kp-10 mitigates PDGF-induced ASM proliferation. Pharmacological inhibition and shRNA knockdown of KISS1R increased basal ASM proliferation, further amplified by PDGF. The anti-proliferative effect of Kp-10 in ASM was found to be mediated by inhibition of MAPK-ERK-Akt pathways, with altered expression of PCNA, C/EBP-alpha, Ki-67, Cyclin-D1, and Cyclin-E leading to cell-cycle arrest at G0/G1 phase. Overall, we demonstrate the importance of Kp/KISS1R signaling in regulating ASM proliferation and a potentially novel therapeutic avenue to blunt remodeling in asthma.
Collapse
Affiliation(s)
- Niyati A Borkar
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, United States of America
| | - Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, United States of America
| | | | - Christina M Pabelick
- Department of Anesthesiology and Physiology, Mayo Clinic, Rochester, United States of America
| | - Y S Prakash
- Department of Anesthesiology and Physiology, Mayo Clinic, Rochester, United States of America
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, United States of America
| |
Collapse
|
36
|
Y It Matters—Sex Differences in Fetal Lung Development. Biomolecules 2022; 12:biom12030437. [PMID: 35327629 PMCID: PMC8946560 DOI: 10.3390/biom12030437] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 02/01/2023] Open
Abstract
Within this review, sex-specific differences in alveolar epithelial functions are discussed with special focus on preterm infants and the respiratory disorders associated with premature birth. First, a short overview about fetal lung development, the challenges the lung faces during perinatal lung transition to air breathing and respiratory distress in preterm infants is given. Next, clinical observations concerning sex-specific differences in pulmonary morbidity of human preterm infants are noted. The second part discusses potential sex-specific causes of pulmonary complications, including pulmonary steroid receptors and local lung steroid metabolism. With regard to pulmonary steroid metabolism, it is important to highlight which steroidogenic enzymes are expressed at which stage during fetal lung development. Thereafter, we review the knowledge concerning sex-specific aspects of lung growth and maturation. Special focus is given to alveolar epithelial Na+ transport as a driver of perinatal lung transition and the sex differences that were noted in this process.
Collapse
|
37
|
The Relationship between Metal Exposure and Chronic Obstructive Pulmonary Disease in the General US Population: NHANES 2015–2016. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042085. [PMID: 35206273 PMCID: PMC8871875 DOI: 10.3390/ijerph19042085] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022]
Abstract
The effects of metal on pulmonary function are inconsistent, and abnormal distribution of metals can decrease lung function. However, the effects of metals exposure on chronic obstructive pulmonary disease (COPD) are still unclear. This study aims to explore the relationship between metal exposure and COPD risk. Cross-sectional data from the National Health and Nutrition Survey (NHANES) 2015–2016 was analyzed. Inductively coupled plasma dynamic reaction cell mass spectrometry (ICP-DRC-MS) was used to measure the metals concentration in the blood. The multiple linear regression and restricted cubic spline (RCS) were used to analyze the relationship between metals exposure and COPD risk. In this study, 1399 participants were included, of which 107 participants were diagnosed with COPD using self-reported chronic bronchitis, emphysema, and COPD. The second and third tertiles of copper increased the COPD risk by 1.98-fold (95% CI: 1.08–3.62) and 2.43-fold (95% CI: 1.32–4.48) compared with the first tertile, using p = 0.005 for the trend after adjusting for the covariates. RCS showed a positive linear correlation between copper and COPD risk (p = 0.006 for overall association) in all participants. When stratified by sex, the multi-factor analysis showed that the third tertile of copper increased male’s COPD risk by 3.42-fold (95% CI: 1.52–7.76), with p = 0.003 for the trend, and RCS also showed a positive linear correlation (p = 0.013 for overall association). Although RCS showed that selenium can reduce the COPD risk (p = 0.008 for overall association) in males, an association between selenium and COPD was not observed (p > 0.05). Our findings suggest that a high concentration of copper may increase COPD risk in males in the general US population, and more research is needed to explore its possible mechanism of action.
Collapse
|
38
|
Ulland TK, Ewald AC, Knutson AO, Marino KM, Smith SMC, Watters JJ. Alzheimer's Disease, Sleep Disordered Breathing, and Microglia: Puzzling out a Common Link. Cells 2021; 10:2907. [PMID: 34831129 PMCID: PMC8616348 DOI: 10.3390/cells10112907] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/14/2022] Open
Abstract
Sleep Disordered Breathing (SDB) and Alzheimer's Disease (AD) are strongly associated clinically, but it is unknown if they are mechanistically associated. Here, we review data covering both the cellular and molecular responses in SDB and AD with an emphasis on the overlapping neuroimmune responses in both diseases. We extensively discuss the use of animal models of both diseases and their relative utilities in modeling human disease. Data presented here from mice exposed to intermittent hypoxia indicate that microglia become more activated following exposure to hypoxia. This also supports the idea that intermittent hypoxia can activate the neuroimmune system in a manner like that seen in AD. Finally, we highlight similarities in the cellular and neuroimmune responses between SDB and AD and propose that these similarities may lead to a pathological synergy between SDB and AD.
Collapse
Affiliation(s)
- Tyler K. Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI 53705, USA; (T.K.U.); (K.M.M.)
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Andrea C. Ewald
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Andrew O. Knutson
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Kaitlyn M. Marino
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI 53705, USA; (T.K.U.); (K.M.M.)
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Stephanie M. C. Smith
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Jyoti J. Watters
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| |
Collapse
|
39
|
Investigation of Combination Treatment With an Aromatase Inhibitor Exemestane and Carboplatin-Based Therapy for Postmenopausal Women With Advanced NSCLC. JTO Clin Res Rep 2021; 2:100150. [PMID: 34590007 PMCID: PMC8474426 DOI: 10.1016/j.jtocrr.2021.100150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 11/24/2022] Open
Abstract
Introduction Estrogen receptors (ER) (ERα, ERβ) and aromatase (key enzyme for estrogen synthesis) are expressed in most human NSCLCs. High intratumoral estrogen levels and elevated aromatase expression in NSCLC predict poor outcome. This open-label, phase 1b, single-center study evaluated the safety and tolerability of escalating doses of the aromatase inhibitor, exemestane, in combination with carboplatin and pemetrexed in postmenopausal women with stage IV nonsquamous NSCLC. Methods Patients received exemestane (starting 1-wk before chemotherapy) at 25 mg orally (PO) daily (cohort 1) or 50 mg PO daily (cohort 2) combined with carboplatin (area under the curve 6 mg × min/mL) and pemetrexed (500 mg/m2) intravenously every 3 weeks for four cycles. Thereafter, patients were eligible for continued therapy with exemestane and pemetrexed or pemetrexed alone. Results A total of 10 patients consented for therapy, and two patients failed in the screening. Four patients completed the therapy in cohort 1 and four patients in cohort 2. The median number of cycles administered was 15 (range: 1-54). Maximum tolerated dose was exemestane 50 mg PO daily with combination chemotherapy. Intention-to-treat analysis revealed an objective response rate (ORR) of 62.5% (five of eight patients with partial response) and a clinical benefit rate of 87.5% (seven of eight patients with either stable disease or partial response). ORR was associated with aromatase expression (p = 0.02). Circulating estrogen levels decreased with exemestane use, and quality of life measurements did not significantly change during the treatment. There were no adverse events. Conclusions The combination of carboplatin, pemetrexed, and exemestane in postmenopausal women with metastatic NSCLC is safe and well tolerated. Biomarker studies revealed that ORR correlates with tumor aromatase expression. These findings support future clinical trials to confirm the antitumor efficacy with this combination therapy.
Collapse
|
40
|
Yang Z, Wang F, Tan F, Cao W, Xu Y, Qin C, Yu Y, Zhao L, Wen Y, Wu Z, Zheng Y, Liu Y, Yu L, Wei D, Dong D, Cao J, Zhang S, Yan S, Wang N, Liao X, Du L, Li J, Li N, Chen W, He J. Menstrual factors, reproductive history, and risk of lung cancer: a multi-center population-based cohort study in Chinese females. Transl Lung Cancer Res 2021; 10:3912-3928. [PMID: 34858781 PMCID: PMC8577983 DOI: 10.21037/tlcr-21-552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Female menstrual and reproductive factors, as remarkable indicators of hormone effect, were hypothesized to be associated with lung cancer risk, whereas the existed epidemiological evidence was inconsistent. Our study aims to investigate the association between menstrual and reproductive factors and lung cancer risk based on the Chinese Lung Cancer Screening Program. METHODS This study was based on a large-scale multi-center population cohort across China recruiting individuals aged 40-74 years old between 2013-2018. Cox regression model was applied to estimate the HRs and 95% CIs. Restricted cubic spline (RCS) analysis was used to estimate dose-response relationships and test for nonlinear associations. RESULTS Among 553,434 female participants, 1,529 incident lung cancer cases were identified with a median follow-up of 3.61 years. With adjustment for multiple covariates and all significant hormonal factors, elevated lung cancer risk was associated with later age (15, or ≥16 years) at menarche (HR =1.27, 95% CI: 1.04-1.56; HR =1.45, 95% CI: 1.19-1.76), later age (25-29, or ≥30 years) at first live birth (HR =1.27, 95% CI: 1.13-1.43; HR =1.23, 95% CI: 1.00-1.51), and benign breast disease history (HR =1.25, 95% CI: 1.10-1.41). For postmenopausal females specifically, surgical menopause (HR =1.62; 95% CI: 1.29-2.05) and other surgeries on the reproductive system (HR =1.19; 95% CI: 1.01-1.40) both appeared to be predictive of elevated lung cancer risk. Concerning age at menopause, a nonlinear association was observed (P-nonlinear =0.0126). Increased lung cancer risk was observed among females with age at menopause especially above 50. Although we observed no significant associations between longer time (≥13 months) of breastfeeding and lung cancer risk among all participants (HR =0.86; 95% CI: 0.71-1.04), significant decreased adenocarcinoma risk (HR =0.65; 95% CI: 0.53-0.81) was noted among nonsmoking females. CONCLUSIONS Our findings add some support for the role of menstrual and reproductive factors in lung carcinogenesis. However, these relationships were complex, and required further investigations addressing the biological mechanisms.
Collapse
Affiliation(s)
- Zhuoyu Yang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Wang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongjie Xu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Qin
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiwen Yu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Zhao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Wen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Wu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yadi Zheng
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunyong Liu
- Department for Cancer Prevention and Treatment of Liaoning Province/Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- Division of Pneumoconiosis School of Public Health, China Medical University, Shenyang, China
| | - Lianzheng Yu
- Department of Noncommunicable Chronic Disease Prevention, Liaoning Center for Disease Control and Prevention, Shenyang, China
| | - Donghua Wei
- Cancer Department of Physical Examination, Anhui Provincial Cancer Hospital, Hefei, China
| | - Dong Dong
- Office of Cancer Prevention and Treatment, Xuzhou Cancer Hospital, Xuzhou, China
| | - Ji Cao
- Department of Experimental Pathology, Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Shaokai Zhang
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Shipeng Yan
- Department of Cancer Prevention and Control, Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ning Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xianzhen Liao
- Department of Cancer Prevention and Control, Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Lingbin Du
- Department of Cancer Prevention, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)/IBMC, Chinese Academy of Sciences, Hangzhou, China
| | - Jiang Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ni Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences Key Laboratory for National Cancer Big Data Analysis and Implement, Beijing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
41
|
Abstract
Lung cancer represents the world's leading cause of cancer deaths. Sex differences in the incidence and mortality rates for various types of lung cancers have been identified, but the biological and endocrine mechanisms implicated in these disparities have not yet been determined. While some cancers such as lung adenocarcinoma are more commonly found among women than men, others like squamous cell carcinoma display the opposite pattern or show no sex differences. Associations of tobacco product use rates, susceptibility to carcinogens, occupational exposures, and indoor and outdoor air pollution have also been linked to differential rates of lung cancer occurrence and mortality between sexes. While roles for sex hormones in other types of cancers affecting women or men have been identified and described, little is known about the influence of sex hormones in lung cancer. One potential mechanism identified to date is the synergism between estrogen and some tobacco compounds, and oncogene mutations, in inducing the expression of metabolic enzymes, leading to enhanced formation of reactive oxygen species and DNA adducts, and subsequent lung carcinogenesis. In this review, we present the literature available regarding sex differences in cancer rates, associations of male and female sex hormones with lung cancer, the influence of exogenous hormone therapy in women, and potential mechanisms mediated by male and female sex hormone receptors in lung carcinogenesis. The influence of biological sex on lung disease has recently been established, thus new research incorporating this variable will shed light on the mechanisms behind the observed disparities in lung cancer rates, and potentially lead to the development of new therapeutics to treat this devastating disease.
Collapse
Affiliation(s)
- Nathalie Fuentes
- National Institute of Allergy and Infectious Diseases, Bethesda, MD 20852, USA
| | - Miguel Silva Rodriguez
- Department of Environmental and Occupational Health, Indiana University, School of Public Health, Bloomington, IN 47405, USA
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, Indiana University, School of Public Health, Bloomington, IN 47405, USA
| |
Collapse
|
42
|
Lachowicz-Scroggins ME, Vuga LJ, Laposky AD, Brown M, Banerjee K, Croxton TL, Kiley JP. The intersection of women's health, lung health, and disease. Am J Physiol Lung Cell Mol Physiol 2021; 321:L624-L627. [PMID: 34431414 DOI: 10.1152/ajplung.00333.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Marrah E Lachowicz-Scroggins
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Louis J Vuga
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Aaron D Laposky
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Marishka Brown
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Koyeli Banerjee
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Thomas L Croxton
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - James P Kiley
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
43
|
Karampitsakos T, Papaioannou O, Katsaras M, Sampsonas F, Tzouvelekis A. Interstitial Lung Diseases and the Impact of Gender. Clin Chest Med 2021; 42:531-541. [PMID: 34353457 DOI: 10.1016/j.ccm.2021.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Interstitial lung diseases encompass an amalgamated group of heterogeneous lung disorders, characterized by variable clinical and radiologic patterns. Despite an increase in our knowledge, pathogenesis of interstitial lung diseases remains largely unknown. Experimental evidence on the role of sex hormones in lung development and epidemiologic associations of gender differences with interstitial lung diseases prevalence fueled studies investigating the role of gender and sex hormones in the pathogenesis and treatment of pulmonary fibrosis. This review summarizes experimental and clinical data for the impact of gender and sex hormones on interstitial lung diseases and highlights future perspectives in the field.
Collapse
Affiliation(s)
| | | | - Matthaios Katsaras
- Department of Respiratory Medicine, University Hospital of Patras, Greece
| | - Fotios Sampsonas
- Department of Respiratory Medicine, University Hospital of Patras, Greece
| | | |
Collapse
|
44
|
Abstract
Disparities in health care have risen to the forefront of medicine in the past several years. One of the most notable disparities in the research and delivery of health care relates to sex and gender. Sex and gender affect the epidemiology, pathophysiology, and outcomes of disease and social determinants of health and access to medical care. This article discusses some of the history of considering sex as a biologic variable in medical research and clinical care. It also clarifies the definitions and terminology necessary for understanding the biologic and social underpinnings of sex and gender.
Collapse
Affiliation(s)
- Shannon Kay
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, PO Box 208057, New Haven, CT 06519, USA
| | - Margaret A Pisani
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, PO Box 208057, New Haven, CT 06519, USA.
| |
Collapse
|
45
|
Lopes ACR, Zavan B, Corrêa YJC, Vieira TM, Severs LJ, Oliveira LM, Soncini R. Impact of obesity and ovariectomy on respiratory function in female mice. Respir Physiol Neurobiol 2021; 294:103775. [PMID: 34416380 DOI: 10.1016/j.resp.2021.103775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
Obesity and the corresponding variations in female sex hormones are associated with severe lung disease. We determined the potential effects of obesity and sex hormones in female mice by investigating changes in lung structure and respiratory function in an obesity model induced by postnatal overnutrition. Obese female mice exhibited pronounced weight gain, abdominal fat accumulation and collagen type I deposition in the airways. However, neither elastic tissue nor estrogen receptors-α/-β were affected in obese female mice after ovariectomy or sham-operated mice. Bronchoconstriction in response to methacholine challenge in obese sham-operated mice was higher than in the obese group after ovariectomy. Our results suggest that the coexistence of obesity and ovariectomy impacted on respiratory system and airway resistance (attenuates bronchoconstriction after methacholine), on collagen I deposition and on airway estrogen β-receptors of mice.
Collapse
Affiliation(s)
- Ana C R Lopes
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Bruno Zavan
- Integrative Animal Biology Laboratory, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Yuri J C Corrêa
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Tânia M Vieira
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil
| | - Liza J Severs
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, JMB10, Seattle, WA, 98101, USA
| | - Luiz M Oliveira
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, JMB10, Seattle, WA, 98101, USA
| | - Roseli Soncini
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, 37130-000, Alfenas, MG, Brazil.
| |
Collapse
|
46
|
Lallai V, Manca L, Fowler CD. E-cigarette vape and lung ACE2 expression: Implications for coronavirus vulnerability. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 86:103656. [PMID: 33838329 PMCID: PMC8025581 DOI: 10.1016/j.etap.2021.103656] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 05/15/2023]
Abstract
Evidence in humans suggests a correlation between nicotine smoking and severe respiratory symptoms with COVID-19 infection. In lung tissue, angiotensin-converting enzyme 2 (ACE2) appears to mechanistically underlie viral entry. Here, we investigated whether e-cigarette vapor inhalation alters ACE2 and nicotinic acetylcholine receptor (nAChR) expression in male and female mice. In male lung, nicotine vapor inhalation induced a significant increase in ACE2 mRNA and protein, but surprisingly, these differences were not found in females. Further, both vehicle and nicotine vapor inhalation downregulated α5 nAChR subunits in both sexes, while differences were not found in α7 nAChR subunit expression. Finally, blood ACE2 levels did not differ with exposure, indicating that blood sampling is not a sufficient indicator of lung ACE2 changes. Together, these data indicate a direct link between e-cigarette vaping and increased ACE2 expression in male lung tissue, which thereby reveals an underlying mechanism of increased vulnerability to coronavirus infection in individuals vaping nicotine.
Collapse
Affiliation(s)
- Valeria Lallai
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
| | - Letizia Manca
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
| | - Christie D Fowler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
47
|
Zein JG, McManus JM, Sharifi N, Erzurum SC, Marozkina N, Lahm T, Giddings O, Davis MD, DeBoer MD, Comhair SA, Bazeley P, Kim HJ, Busse W, Calhoun W, Castro M, Chung KF, Fahy JV, Israel E, Jarjour NN, Levy BD, Mauger DT, Moore WC, Ortega VE, Peters M, Bleecker ER, Meyers DA, Zhao Y, Wenzel SE, Gaston B. Benefits of Airway Androgen Receptor Expression in Human Asthma. Am J Respir Crit Care Med 2021; 204:285-293. [PMID: 33779531 DOI: 10.1164/rccm.202009-3720oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Androgens are potentially beneficial in asthma, but AR (androgen receptor) has not been studied in human airways.Objectives: To measure whether AR and its ligands are associated with human asthma outcomes.Methods: We compared the effects of AR expression on lung function, symptom scores, and fractional exhaled nitric oxide (FeNO) in adults enrolled in SARP (Severe Asthma Research Program). The impact of sex and of androgens on asthma outcomes was also evaluated in the SARP with validation studies in the Cleveland Clinic Health System and the NHANES (U.S. National Health and Nutrition Examination Survey).Measurements and Main Results: In SARP (n = 128), AR gene expression from bronchoscopic epithelial brushings was positively associated with both FEV1/FVC ratio (R2 = 0.135, P = 0.0002) and the total Asthma Quality of Life Questionnaire score (R2 = 0.056, P = 0.016) and was negatively associated with FeNO (R2 = 0.178, P = 9.8 × 10-6) and NOS2 (nitric oxide synthase gene) expression (R2 = 0.281, P = 1.2 × 10-10). In SARP (n = 1,659), the Cleveland Clinic Health System (n = 32,527), and the NHANES (n = 2,629), women had more asthma exacerbations and emergency department visits than men. The levels of the AR ligand precursor dehydroepiandrosterone sulfate correlated positively with the FEV1 in both women and men.Conclusions: Higher bronchial AR expression and higher androgen levels are associated with better lung function, fewer symptoms, and a lower FeNO in human asthma. The role of androgens should be considered in asthma management.
Collapse
Affiliation(s)
- Joe G Zein
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | - Serpil C Erzurum
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | | | | | - Mark D DeBoer
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Suzy A Comhair
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Peter Bazeley
- Lerner Research Institute and.,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Hyun Jo Kim
- Department of Systems Biology and Bioinformatics, Case Western Reserve University, Cleveland, Ohio
| | - William Busse
- Department of Medicine, School of Medicine, University of Wisconsin, Madison, Wisconsin
| | - William Calhoun
- Department of Medicine, University of Texas Medical Branch, University of Texas, Galveston, Texas
| | - Mario Castro
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, University of Kansas, Kansas City, Kansas
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - John V Fahy
- Division of Pulmonary, Critical Care, and Sleep Medicine, School of Medicine, University of Kansas, Kansas City, Kansas.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California at San Francisco, San Francisco, California
| | - Elliot Israel
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Nizar N Jarjour
- Department of Medicine, School of Medicine, University of Wisconsin, Madison, Wisconsin
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts
| | - David T Mauger
- Center for Biostatistics and Epidemiology, School of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Wendy C Moore
- Section on Pulmonary, Critical Care, Allergic, and Immunologic Disease, Department of Internal Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Victor E Ortega
- Section on Pulmonary, Critical Care, Allergic, and Immunologic Disease, Department of Internal Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Michael Peters
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California at San Francisco, San Francisco, California
| | - Eugene R Bleecker
- Division of Genetics, Genomics, and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Arizona; and
| | - Deborah A Meyers
- Division of Genetics, Genomics, and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Arizona; and
| | - Yi Zhao
- Department of Biostatistics and Health Science Data, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
48
|
Abstract
Kidney pathophysiology is influenced by gender. Evidence suggests that kidney damage is more severe in males than in females and that sexual hormones contribute to this. Elevated prolactin concentration is common in renal impairment patients and is associated with an unfavorable prognosis. However, PRL is involved in the osmoregulatory process and promotes endothelial proliferation, dilatation, and permeability in blood vessels. Several proteinases cleavage its structure, forming vasoinhibins. These fragments have antagonistic PRL effects on endothelium and might be associated with renal endothelial dysfunction, but its role in the kidneys has not been enough investigated. Therefore, the purpose of this review is to describe the influence of sexual dimorphism and gonadal hormones on kidney damage, emphasizing the role of the hormone prolactin and its cleavage products, the vasoinhibins.
Collapse
|
49
|
Chiarella SE, Cardet JC, Prakash YS. Sex, Cells, and Asthma. Mayo Clin Proc 2021; 96:1955-1969. [PMID: 34218868 PMCID: PMC8262071 DOI: 10.1016/j.mayocp.2020.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/19/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
There are marked sex differences in asthma prevalence and severity. Sex hormones play a central role in these sex biases and directly interact with multiple key cells involved in the pathogenesis of asthma. Here we review the known effects of estrogen, progesterone, and testosterone on airway epithelial cells, airway smooth muscle cells, the mononuclear phagocyte system, innate lymphoid cells, eosinophils, mast cells, T cells, and B cells, all in the context of asthma. Furthermore, we explore unresolved clinical questions, such as the role of sex hormones in the link between asthma and obesity.
Collapse
Affiliation(s)
- Sergio E Chiarella
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Juan Carlos Cardet
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida, Tampa
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN.
| |
Collapse
|
50
|
Kim NN. Close Encounters in the World of Women's Sexual Health: An Alien's Journey. Sex Med Rev 2021; 9:359-364. [PMID: 34246428 DOI: 10.1016/j.sxmr.2021.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|