1
|
Yang JJ, Li Z, Wang LN, Huang BX, Ng JPL, Xu XF, Wang YP, Zhang DW, Qin B, Zhang DQ, Liu C, Luo WD, Law BYK, Wang HM, Liu MH, Yun XY, Chan JTW, Wu WY, Li YT, Cheung PKF, Pou MC, Ha KS, Ao Ieong WF, Leong CH, Leong KI, Lei CW, Cheang LH, Wong VKW. X-chromosome-linked miR-542-5p as a key regulator of sex disparity in rats with adjuvant-induced arthritis by promoting Th17 differentiation. Biomark Res 2025; 13:36. [PMID: 40025567 PMCID: PMC11872315 DOI: 10.1186/s40364-025-00741-x] [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: 12/16/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Studies have indicated that X-linked microRNAs (miRNAs) play a role in the pathogenesis of rheumatoid arthritis (RA) and its gender-specific differences. However, research on specific miRNAs remains limited. This study aims to investigate the possible role of X-linked miR-542-5p in RA pathogenesis and gender differences. METHODS We investigated the impact of miR-542-5p on RA pathogenesis and gender differences by manipulating its expression in various rat models. RESULTS Our findings revealed a significant overexpression of miR-542-5p in RA patients compared with healthy individuals, with a notable gender difference among RA patients. In vivo experiments confirmed that upregulation of miR-542-5p could accelerate RA pathogenesis. Further analysis showed that the onset of adjuvant-induced arthritis (AIA) in rats exhibited significant gender differences, with more severe clinical phenotypes found in female rats. This may be attributed to their stronger immune responses and elevated levels of miR-542-5p. Subsequent in vitro and in vivo experiments demonstrated that miR-542-5p contributes to the regulation of gender differences in RA pathogenesis by promoting the differentiation of Th17 cells. CONCLUSIONS This study offers new insights into the sex-specific nature of RA, suggesting X-linked miR-542-5p as a potential target for both diagnostic and therapeutic purposes. These findings lay the groundwork for the development of gender-specific therapeutic strategies for RA and underscore the importance of gender consideration in RA research.
Collapse
Affiliation(s)
- Jiu Jie Yang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- Macau Medical Science and Technology Research Association, Macao SAR, China
| | - Zhi Li
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Lin Na Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Bai Xiong Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Jerome P L Ng
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Xiong Fei Xu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Yu Ping Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - David Wei Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Bo Qin
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Ding Qi Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Chang Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Wei Dan Luo
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Betty Yuen Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Hui Miao Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Meng Han Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Xiao Yun Yun
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Joyce Tsz Wai Chan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Wan Yu Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Yi Ting Li
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Peter Kam Fai Cheung
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Man Chon Pou
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Kat Sang Ha
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Wang Fai Ao Ieong
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Chi Hou Leong
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Kit Ieng Leong
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Chan Wang Lei
- Macau Medical Science and Technology Research Association, Macao SAR, China
- Centro Hospitalar Conde de São Januário, Macau SAR, China
| | - Lek Hang Cheang
- Macau Medical Science and Technology Research Association, Macao SAR, China.
- Centro Hospitalar Conde de São Januário, Macau SAR, China.
| | - Vincent Kam Wai Wong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China.
| |
Collapse
|
2
|
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
|
3
|
Commodore S, Ekpruke CD, Rousselle D, Alford R, Babayev M, Sharma S, Buechlein A, Rusch DB, Silveyra P. Lung proinflammatory microRNA and cytokine expression in a mouse model of allergic inflammation: role of sex chromosome complement and gonadal hormones. Physiol Genomics 2024; 56:179-193. [PMID: 38047312 PMCID: PMC11281810 DOI: 10.1152/physiolgenomics.00049.2023] [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: 06/01/2023] [Revised: 10/13/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023] Open
Abstract
Epigenetic alterations such as dysregulation of miRNAs have been reported to play important roles in interactions between genetic and environmental factors. In this study, we tested the hypothesis that induction of lung inflammation by inhaled allergens triggers a sex-specific miRNA regulation that is dependent on chromosome complement and hormonal milieu. We challenged the four core genotypes (FCGs) model through intranasal sensitization with a house dust mite (HDM) solution (or PBS as a control) for 5 wk. The FCG model allows four combinations of gonads and sex chromosomes: 1) XX mice with ovaries (XXF), 2) XY mice with testes (XYM), 3) XX mice with testes (XXM), and 4) XY mice with ovaries (XYF). Following the challenge (n = 5-7/group), we assessed the expression of 84 inflammatory miRNAs in lung tissue using a PCR array and cytokine levels in bronchoalveolar lavage fluid (BAL) by a multiplex protein assay (n = 4-7 animals/group). Our results showed higher levels of the chemokine KC (an Il-8 homolog) and IL-7 in BAL from XYF mice challenged with HDM. In addition, IL-17A was significantly higher in BAL from both XXF and XYF mice. A three-way interaction among treatment, gonads, and sex chromosome revealed 60 of 64 miRNAs that differed in expression depending on genotype; XXF, XXM, XYF, and XYM mice had 45, 32, 4, and 52 differentially expressed miRNAs, respectively. Regulatory networks of miRNAs identified in this study were implicated in pathways associated with asthma. Female gonadal hormonal effects may alter miRNA expression and contribute to the higher susceptibility of females to asthma.NEW & NOTEWORTHY miRNAs play important roles in regulating gene and environmental interactions. However, their role in mediating sex differences in allergic responses and lung diseases has not been elucidated. Our study used a targeted omics approach to characterize the contributions of gonadal hormones and chromosomal components to lung responses to an allergen challenge. Our results point to the influence of sex hormones in miRNA expression and proinflammatory markers in allergic airway inflammation.
Collapse
Affiliation(s)
- Sarah Commodore
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States
| | - Carolyn Damilola Ekpruke
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States
| | - Dustin Rousselle
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States
| | - Rachel Alford
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States
| | - Maksat Babayev
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States
| | - Shikha Sharma
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States
| | - Aaron Buechlein
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, Indiana, United States
| | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, Indiana, United States
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States
| |
Collapse
|
4
|
Saleem A, Awan T, Akhtar MF. A comprehensive review on endocrine toxicity of gaseous components and particulate matter in smog. Front Endocrinol (Lausanne) 2024; 15:1294205. [PMID: 38352708 PMCID: PMC10863453 DOI: 10.3389/fendo.2024.1294205] [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: 09/14/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Smog is a form of extreme air pollution which comprises of gases such as ozone, sulfur dioxide, nitrogen and carbon oxides, and solid particles including particulate matter (PM2.5 and PM10). Different types of smog include acidic, photochemical, and Polish. Smog and its constituents are hazardaous to human, animals, and plants. Smog leads to plethora of morbidities such as cancer, endocrine disruption, and respiratory and cardiovascular disorders. Smog components alter the activity of various hormones including thyroid, pituitary, gonads and adrenal hormones by altering regulatory genes, oxidation status and the hypothalamus-pituitary axis. Furthermore, these toxicants are responsible for the development of metabolic disorders, teratogenicity, insulin resistance, infertility, and carcinogenicity of endocrine glands. Avoiding fossil fuel, using renewable sources of energy, and limiting gaseous discharge from industries can be helpful to avoid endocrine disruption and other toxicities of smog. This review focuses on the toxic implications of smog and its constituents on endocrine system, their toxicodynamics and preventive measures to avoid hazardous health effects.
Collapse
Affiliation(s)
- Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Tanzeela Awan
- Department of Pharmacy, The Women University Multan, Multan, Pakistan
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| |
Collapse
|
5
|
Sundar IK, Duraisamy SK, Choudhary I, Saini Y, Silveyra P. Acute and Repeated Ozone Exposures Differentially Affect Circadian Clock Gene Expression in Mice. Adv Biol (Weinh) 2023; 7:e2300045. [PMID: 37204107 PMCID: PMC10657336 DOI: 10.1002/adbi.202300045] [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: 01/30/2023] [Revised: 04/20/2023] [Indexed: 05/20/2023]
Abstract
Circadian rhythms have an established role in regulating physiological processes, such as inflammation, immunity, and metabolism. Ozone, a common environmental pollutant with strong oxidative potential, is implicated in lung inflammation/injury in asthmatics. However, whether O3 exposure affects the expression of circadian clock genes in the lungs is not known. In this study, changes in the expression of core clock genes are analyzed in the lungs of adult female and male mice exposed to filtered air (FA) or O3 using qRT-PCR. The findings are confirmed using an existing RNA-sequencing dataset from repeated FA- and O3 -exposed mouse lungs and validated by qRT-PCR. Acute O3 exposure significantly alters the expression of clock genes in the lungs of females (Per1, Cry1, and Rora) and males (Per1). RNA-seq data revealing sex-based differences in clock gene expression in the airway of males (decreased Nr1d1/Rev-erbα) and females (increased Skp1), parenchyma of females and males (decreased Nr1d1 and Fbxl3 and increased Bhlhe40 and Skp1), and alveolar macrophages of males (decreased Arntl/Bmal1, Per1, Per2, Prkab1, and Prkab2) and females (increased Cry2, Per1, Per2, Csnk1d, Csnk1e, Prkab2, and Fbxl3). These findings suggest that lung inflammation caused by O3 exposure affects clock genes which may regulate key signaling pathways.
Collapse
Affiliation(s)
- Isaac Kirubakaran Sundar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Santhosh Kumar Duraisamy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, Indiana University, School of Public Health, Bloomington, IN, USA
| |
Collapse
|
6
|
Deng Y, Wang J, Sun L, Wang Y, Chen J, Zhao Z, Wang T, Xiang Y, Wang Y, Chen J, He M. Effects of Ambient O 3 on Respiratory Mortality, Especially the Combined Effects of PM 2.5 and O 3. TOXICS 2023; 11:892. [PMID: 37999544 PMCID: PMC10675328 DOI: 10.3390/toxics11110892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND In China, the increasing concentration of ozone (O3) has emerged as a significant air pollution issue, leading to adverse effects on public health, particularly the respiratory system. Despite the progress made in managing air pollution in China, it is crucial to address the problem of environmental O3 pollution at present. METHODS The connection between O3 exposure and respiratory mortality in Shenyang, China, from 2014 to 2018 was analyzed by a time-series generalized additive regression model (GAM) with quasi-Poisson regression. Additionally, the potential combined effects of fine particulate matter (PM2.5) and O3 were investigated using the synergy index (SI). RESULTS Our findings indicate that each 10 μg/m3 increase in O3 at lag 2 days was associated with a maximum relative risk (RR) of 1.0150 (95% CI: 1.0098-1.0202) for respiratory mortality in the total population. For individuals aged ≥55 years, unmarried individuals, those engaged in indoor occupations, and those with low educational attainment, each 10 μg/m3 increase in O3 at lag 07 days was linked to RR values of 1.0301 (95% CI: 1.0187-1.0417), 1.0437 (95% CI: 1.0266-1.0610), 1.0317 (95% CI: 1.0186-1.0450), and 1.0346 (95% CI: 1.0222-1.0471), respectively. Importantly, we discovered a synergistic effect of PM2.5 and O3, resulting in an SI of 2.372 on the occurrence of respiratory mortality. CONCLUSIONS This study confirmed a positive association between O3 exposure and respiratory mortality. Furthermore, it highlighted the interaction between O3 and PM2.5 in exacerbating respiratory deaths.
Collapse
Affiliation(s)
- Ye Deng
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Junlong Wang
- Liaoning Provincial Center for Disease Control and Prevention, Shenyang 110005, China
| | - Li Sun
- Liaoning Provincial Center for Disease Control and Prevention, Shenyang 110005, China
| | - Yue Wang
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Jiaoyang Chen
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Zhixin Zhao
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Tianyun Wang
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Yuting Xiang
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Yuting Wang
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Jiamei Chen
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
| | - Miao He
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Department of Environmental Health, School of Public Health, Ministry of Education, China Medical University, Shenyang 110122, China
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
| |
Collapse
|
7
|
Siddaiah R, Oji‐Mmuo C, Aluquin VPR, Kawasawa YI, Donnelly A, Rousselle D, Fuentes N, Austin ED, Silveyra P. Multiomics endotyping of preterm infants with bronchopulmonary dysplasia and pulmonary hypertension-A pilot study. Pulm Circ 2023; 13:e12232. [PMID: 37123538 PMCID: PMC10142061 DOI: 10.1002/pul2.12232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023] Open
Abstract
Pulmonary hypertension associated with bronchopulmonary dysplasia is a severe complication of preterm birth resulting in high mortality of up to 50% within the first 2 years of life. There is a direct relationship between bronchopulmonary dysplasia severity and incidence of associated pulmonary hypertension. However, it is challenging to clinically characterize severe bronchopulmonary dysplasia with and without pulmonary hypertension and there is need for better understanding of the two entities. Our main objective is to identify markers to help understand biological processes and characterize infants with pulmonary hypertension associated with bronchopulmonary dysplasia using tracheal aspirates. We conducted an unbiased multiomic analysis of tracheal aspirates via microRNA (miRNA) polymerase chain reaction arrays, RNA sequencing, and mass spectrometry proteomics in preterm infants with severe bronchopulmonary dysplasia with and without pulmonary hypertension (n = 46). Our pilot study analysis revealed 12 miRNAs (hsa-miR-29a, has-miR-542-3p, has-miR-624, has-miR-183, hsa-miR-501-3p, hsa-miR-101, hsa-miR-3131, hsa-miR-3683, hsa-miR-3193, hsa-miR-3672, hsa-miR-3128, and hsa-miR-1287), 6 transcripts (IL6, RPL35P5, HSD3B7, RNA5SP215, OR2A1-AS1, and RNVU1-19), and 5 proteins (CAPS, AAT, KRT5, SFTPB, and LGALS3BP) with significant differential expression in preterm infants with severe lung disease with pulmonary hypertension when compared with infants with severe lung disease but no pulmonary hypertension. Pathway analysis of the integrated multiomic expression signatures revealed NFkB, VEGF, SERPINA1, IL6, and ERK1/2 as target molecules and cellular development, cellular growth and proliferation, and cellular movement as key affected molecular functions. Our multiomic analysis of tracheal aspirates revealed a comprehensive thumbprint of miRNAs, mRNAs, and proteins that could help endotype infants with severe lung disease and pulmonary hypertension.
Collapse
Affiliation(s)
- Roopa Siddaiah
- Department of PediatricsPenn State Health Children's HospitalHersheyPennsylvaniaUSA
| | - Christiana Oji‐Mmuo
- Department of PediatricsPenn State Health Children's HospitalHersheyPennsylvaniaUSA
| | | | - Yuka Imamura Kawasawa
- Department of PharmacologyPenn State Health Children's HospitalHersheyPennsylvaniaUSA
| | - Ann Donnelly
- Department of Respiratory TherapyPenn State Health Children's HospitalHersheyPennsylvaniaUSA
| | - Dustin Rousselle
- Department of Environmental and Occupational HealthIndiana University School of Public HealthBloomingtonIndianaUSA
| | - Nathalie Fuentes
- Department of PediatricsPenn State Health Children's HospitalHersheyPennsylvaniaUSA
| | - Eric D. Austin
- Department of PediatricsVanderbilt UniversityNashvilleTennesseeUSA
| | - Patricia Silveyra
- Department of PediatricsPenn State Health Children's HospitalHersheyPennsylvaniaUSA
- Department of Environmental and Occupational HealthIndiana University School of Public HealthBloomingtonIndianaUSA
| |
Collapse
|
8
|
Mohamed HA, Abdelkafy AE, Khairy RMM, Abdelraheim SR, Kamel BA, Marey H. MicroRNAs and cytokines as potential predictive biomarkers for COVID-19 disease progression. Sci Rep 2023; 13:3531. [PMID: 36864077 PMCID: PMC9979137 DOI: 10.1038/s41598-023-30474-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Host microRNAs can influence the cytokine storm associated SARS-CoV-2 infection and proposed as biomarkers for COVID-19 disease. In the present study, serum MiRNA-106a and miRNA-20a were quantified by real time-PCR in 50 COVID-19 patients hospitalized at Minia university hospital and 30 healthy volunteers. Profiles of serum inflammatory cytokines (TNF-α, IFN-γ, and IL-10) and TLR4 were analyzed by Eliza in patients and controls. A highly significant decrease (P value = 0.0001) in the expressions of miRNA-106a and miRNA-20a was reported in COVID-19 patients compared to controls. A significant decrease in the levels of miRNA-20a was also reported in patients with lymphopenia, patients having chest CT severity score (CSS) > 19 and in patients having O2 saturation less than 90%. Significantly higher levels of TNF-α, IFN-γ, IL-10 and TLR4 were reported in patients compared to controls. IL-10 and TLR4 levels were significantly higher in patients having lymphopenia. TLR-4 level was higher in patients with CSS > 19 and in patients with hypoxia. Using univariate logistic regression analysis, miRNA-106a, miRNA-20a, TNF-α, IFN-γ, IL-10 and TLR4 were identified as good predictors of disease. Receiver operating curve showed that the downregulation of miRNA-20a in patients having lymphopenia, patients with CSS > 19 and patients with hypoxia could be a potential biomarker with AUC = 0.68 ± 0.08, AUC = 0.73 ± 0.07 and AUC = 0.68 ± 0.07 respectively. Also, ROC curve showed accurate association between the increase of serum IL-10 and TLR-4 and lymphopenia among COVID-19 patients with AUC = 0.66 ± 0.08 and AUC = 0.73 ± 0.07 respectively. ROC curve showed also that serum TLR-4 could be a potential marker for high CSS with AUC = 0.78 ± 0.06. A negative correlation was detected between miRNA-20a with TLR-4 (r = - 0.30, P value = 0.03). We concluded that, miR-20a, is a potential biomarker of COVID-19 severity and blockade of IL-10 and TLR4 may constitute a novel therapy for COVID-19 patients.
Collapse
Affiliation(s)
- Hatem A. Mohamed
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Aya Eid Abdelkafy
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Rasha M. M. Khairy
- grid.411806.a0000 0000 8999 4945Department of Microbiology and Immunology, Faculty of Medicine, Minia University, Minia, 61511 Egypt
| | - Salama R. Abdelraheim
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Bothina Ahmed Kamel
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Heba Marey
- grid.411806.a0000 0000 8999 4945Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
9
|
Ho K, Weimar D, Torres-Matias G, Lee H, Shamsi S, Shalosky E, Yaeger M, Hartzler-Lovins H, Dunigan-Russell K, Jelic D, Novak CM, Gowdy KM, Englert JA, Ballinger MN. Ozone impairs endogenous compensatory responses in allergic asthma. Toxicol Appl Pharmacol 2023; 459:116341. [PMID: 36502870 PMCID: PMC9840700 DOI: 10.1016/j.taap.2022.116341] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
Asthma is a chronic inflammatory airway disease characterized by acute exacerbations triggered by inhaled allergens, respiratory infections, or air pollution. Ozone (O3), a major component of air pollution, can damage the lung epithelium in healthy individuals. Despite this association, little is known about the effects of O3 and its impact on chronic lung disease. Epidemiological data have demonstrated that elevations in ambient O3 are associated with increased asthma exacerbations. To identify mechanisms by which O3 exposure leads to asthma exacerbations, we developed a two-hit mouse model where mice were sensitized and challenged with three common allergens (dust mite, ragweed and Aspergillus fumigates, DRA) to induce allergic inflammation prior to exposure to O3 (DRAO3). Changes in lung physiology, inflammatory cells, and inflammation were measured. Exposure to O3 following DRA significantly increased airway hyperreactivity (AHR), which was independent of TLR4. DRA exposure resulted in increased BAL eosinophilia while O3 exposure resulted in neutrophilia. Additionally, O3 exposure following DRA blunted anti-inflammatory and antioxidant responses. Finally, there were significantly less monocytes and innate lymphoid type 2 cells (ILC2s) in the dual challenged DRA-O3 group suggesting that the lack of these immune cells may influence O3-induced AHR in the setting of allergic inflammation. In summary, we developed a mouse model that mirrors some aspects of the clinical course of asthma exacerbations due to air pollution and identified that O3 exposure in the asthmatic lung leads to impaired endogenous anti-inflammatory and antioxidant responses and alterations inflammatory cell populations.
Collapse
Affiliation(s)
- Kevin Ho
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - David Weimar
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Gina Torres-Matias
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States of America
| | - Hyunwook Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Saaleha Shamsi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Emily Shalosky
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Michael Yaeger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States of America
| | - Hannah Hartzler-Lovins
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America; Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, United States of America
| | - Katelyn Dunigan-Russell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Daria Jelic
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Caymen M Novak
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Joshua A Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Megan N Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States of America.
| |
Collapse
|
10
|
Identification of Potential microRNA Panels for Male Non-Small Cell Lung Cancer Identification Using Microarray Datasets and Bioinformatics Methods. J Pers Med 2022; 12:jpm12122056. [PMID: 36556276 PMCID: PMC9780989 DOI: 10.3390/jpm12122056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Non-small cell lung cancer (NSCLC) is still one of the types of cancer with the highest death rates. MicroRNAs (miRNAs) play essential roles in NSCLC development. This study evaluates miRNA expression patterns and specific mechanisms in male patients with NSCLC. Methods: We report an integrated microarray analysis of miRNAs for eight matched samples of males with NSCLC compared to the study of public datasets of males with NSCLC from TCGA, followed by qRT-PCR validation. Results: For the TCGA dataset, we identified 385 overexpressed and 75 underexpressed miRNAs. Our cohort identified 54 overexpressed and 77 underexpressed miRNAs, considering a fold-change (FC) of ±1.5 and p < 0.05 as the cutoff value. The common miRNA signature consisted of eight overexpressed and nine underexpressed miRNAs. Validation was performed using qRT-PCR on the tissue samples for miR-183-3p and miR-34c-5p and on plasma samples for miR-34c-5p. We also created mRNA-miRNA regulatory networks to identify critical molecules, revealing NSCLC signaling pathways related to underexpressed and overexpressed transcripts. The genes targeted by these transcripts were correlated with overall survival. Conclusions: miRNAs and some of their target genes could play essential roles in investigating the mechanisms involved in NSCLC evolution and provide opportunities to identify potential therapeutic targets.
Collapse
|
11
|
Jackson TW, Henriquez AR, Snow SJ, Schladweiler MC, Fisher AA, Alewel DI, House JS, Kodavanti UP. Adrenal Stress Hormone Regulation of Hepatic Homeostatic Function After an Acute Ozone Exposure in Wistar-Kyoto Male Rats. Toxicol Sci 2022; 189:73-90. [PMID: 35737395 PMCID: PMC9609881 DOI: 10.1093/toxsci/kfac065] [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] [Indexed: 11/13/2022] Open
Abstract
Ozone-induced lung injury, inflammation, and pulmonary/hypothalamus gene expression changes are diminished in adrenalectomized (AD) rats. Acute ozone exposure induces metabolic alterations concomitant with increases in epinephrine and corticosterone. We hypothesized that adrenal hormones are responsible for observed hepatic ozone effects, and in AD rats, these changes would be diminished. In total, 5-7 days after sham (SH) or AD surgeries, male Wistar-Kyoto rats were exposed to air or 0.8-ppm ozone for 4 h. Serum samples were analyzed for metabolites and liver for transcriptional changes immediately post-exposure. Ozone increased circulating triglycerides, cholesterol, free fatty-acids, and leptin in SH but not AD rats. Ozone-induced inhibition of glucose-mediated insulin release was absent in AD rats. Unlike diminution of ozone-induced hypothalamus and lung mRNA expression changes, AD in air-exposed rats (AD-air/SH-air) caused differential hepatic expression of ∼1000 genes. Likewise, ozone in AD rats caused differential expression of ∼1000 genes (AD-ozone/AD-air). Ozone-induced hepatic changes in SH rats reflected enrichment for pathways involving metabolic processes, including acetyl-CoA biosynthesis, TCA cycle, and sirtuins. Upstream predictor analysis identified similarity to responses produced by glucocorticoids and pathways involving forskolin. These changes were absent in AD rats exposed to ozone. However, ozone caused unique changes in AD liver mRNA reflecting activation of synaptogenesis, neurovascular coupling, neuroinflammation, and insulin signaling with inhibition of senescence pathways. In these rats, upstream predictor analysis identified numerous microRNAs likely involved in glucocorticoid insufficiency. These data demonstrate the critical role of adrenal stress hormones in ozone-induced hepatic homeostasis and necessitate further research elucidating their role in propagating environmentally driven diseases.
Collapse
Affiliation(s)
- Thomas W Jackson
- Oak Ridge Institute for Science and Education Research Participation Program
| | - Andres R Henriquez
- Oak Ridge Institute for Science and Education Research Participation Program
| | - Samantha J Snow
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Mette C Schladweiler
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Anna A Fisher
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Devin I Alewel
- Oak Ridge Institute for Science and Education Research Participation Program
| | - John S House
- Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| |
Collapse
|
12
|
Guo H, Yu H, Feng Y, Cheng W, Li Y, Wang Y. The role of estrogen receptor β in fine particulate matter (PM 2.5) organic extract-induced pulmonary inflammation in female and male mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:60922-60932. [PMID: 35435549 DOI: 10.1007/s11356-022-20055-x] [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: 01/13/2022] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
Fine particulate matter organic extract (Po) was reported to promote inflammation in the lung. Sex differences were reported in many inflammatory diseases. In this study, we investigated the effects of Po exposure on pulmonary inflammatory response and evaluated the role of sex in this process. While mice were exposed to 100 µg/m3 Po for 12 weeks by an inhalation exposure system, the lung histopathological analysis shown obvious inflammation, the cell numbers in bronchoalveolar lavage fluid (BALF) were significantly increased, and most inflammatory cytokines in BALF were upregulated. The results of factorial analysis of variance shown that there was an interaction between sex and Po exposure in the inflammatory cell numbers and the levels of tumor necrosis factor-α (TNF-α), interleukin-5 (IL-5), and growth-related oncogene/keratinocyte chemoattractant (GRO/KC). Notably, these changes and interactions were diminished while Po-exposed mice were administered with the estrogen receptor β (ERβ) antagonist. We speculated that sex might affect the levels of inflammatory indicators in BALF of Po-exposed mice and female mice were more prone to inflammation while exposed to Po. Moreover, ERβ was involved in these processes. To our knowledge, this is the first investigation about the role of sex in Po-induced adverse effects.
Collapse
Affiliation(s)
- Huaqi Guo
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
- The Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, no. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Hengyi Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yan Feng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Wei Cheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yan Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yan Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
- The Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, no. 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
13
|
Tuazon JA, Kilburg-Basnyat B, Oldfield LM, Wiscovitch-Russo R, Dunigan-Russell K, Fedulov AV, Oestreich KJ, Gowdy KM. Emerging Insights into the Impact of Air Pollution on Immune-Mediated Asthma Pathogenesis. Curr Allergy Asthma Rep 2022; 22:77-92. [PMID: 35394608 PMCID: PMC9246904 DOI: 10.1007/s11882-022-01034-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Increases in ambient levels of air pollutants have been linked to lung inflammation and remodeling, processes that lead to the development and exacerbation of allergic asthma. Conventional research has focused on the role of CD4+ T helper 2 (TH2) cells in the pathogenesis of air pollution-induced asthma. However, much work in the past decade has uncovered an array of air pollution-induced non-TH2 immune mechanisms that contribute to allergic airway inflammation and disease. RECENT FINDINGS In this article, we review current research demonstrating the connection between common air pollutants and their downstream effects on non-TH2 immune responses emerging as key players in asthma, including PRRs, ILCs, and non-TH2 T cell subsets. We also discuss the proposed mechanisms by which air pollution increases immune-mediated asthma risk, including pre-existing genetic risk, epigenetic alterations in immune cells, and perturbation of the composition and function of the lung and gut microbiomes. Together, these studies reveal the multifaceted impacts of various air pollutants on innate and adaptive immune functions via genetic, epigenetic, and microbiome-based mechanisms that facilitate the induction and worsening of asthma.
Collapse
Affiliation(s)
- J A Tuazon
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, 43210, USA
| | - B Kilburg-Basnyat
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, 27858, USA
| | - L M Oldfield
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
- Department of Synthetic Genomics, Replay Holdings LLC, San Diego, 92121, USA
| | - R Wiscovitch-Russo
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
| | - K Dunigan-Russell
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210, USA
| | - A V Fedulov
- Division of Surgical Research, Department of Surgery, Alpert Medical School, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| | - K J Oestreich
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, The James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - K M Gowdy
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210, USA.
| |
Collapse
|
14
|
Enquobahrie DA, MacDonald J, Hussey M, Bammler TK, Loftus CT, Paquette AG, Byington N, Marsit CJ, Szpiro A, Kaufman JD, LeWinn KZ, Bush NR, Tylavsky F, Karr CJ, Sathyanarayana S. Prenatal exposure to particulate matter and placental gene expression. ENVIRONMENT INTERNATIONAL 2022; 165:107310. [PMID: 35653832 PMCID: PMC9235522 DOI: 10.1016/j.envint.2022.107310] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/13/2022] [Accepted: 05/16/2022] [Indexed: 05/30/2023]
Abstract
BACKGROUND While strong evidence supports adverse maternal and offspring consequences of air pollution, mechanisms that involve the placenta, a key part of the intrauterine environment, are largely unknown. Previous studies of air pollution and placental gene expression were small candidate gene studies that rarely considered prenatal windows of exposure or the potential role of offspring sex. We examined overall and sex-specific associations of prenatal exposure to fine particulate matter (PM2.5) with genome-wide placental gene expression. METHODS Participants with placenta samples, collected at birth, and childhood health outcomes from CANDLE (Memphis, TN) (n = 776) and GAPPS (Seattle, WA) (n = 205) cohorts of the ECHO-PATHWAYS Consortium were included in this study. PM2.5 exposures during trimesters 1, 2, 3, and the first and last months of pregnancy, were estimated using a spatiotemporal model. Cohort-specific linear adjusted models were fit for each exposure window and expression of >11,000 protein coding genes from paired end RNA sequencing data. Models with interaction terms were used to examine PM2.5-offspring sex interactions. False discovery rate (FDR < 0.10) was used to correct for multiple testing. RESULTS Mean PM2.5 estimate was 10.5-10.7 μg/m3 for CANDLE and 6.0-6.3 μg/m3 for GAPPS participants. In CANDLE, expression of 13 (11 upregulated and 2 downregulated), 20 (11 upregulated and 9 downregulated) and 3 (2 upregulated and 1 downregulated) genes was associated with PM2.5 in the first trimester, second trimester, and first month, respectively. While we did not find any statistically significant association, overall, between PM2.5 and gene expression in GAPPS, we found offspring sex and first month PM2.5 interaction for DDHD1 expression (positive association among males and inverse association among females). We did not observe PM2.5 and offspring sex interactions in CANDLE. CONCLUSION In CANDLE, but not GAPPS, we found that prenatal PM2.5 exposure during the first half of pregnancy is associated with placental gene expression.
Collapse
Affiliation(s)
- Daniel A Enquobahrie
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States; Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, WA, United States.
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Michael Hussey
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Christine T Loftus
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Alison G Paquette
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, United States; Seattle Children's Research Institute, Seattle, WA, United States
| | - Nora Byington
- Seattle Children's Research Institute, Seattle, WA, United States
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Adam Szpiro
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, United States
| | - Joel D Kaufman
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States; Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States
| | - Kaja Z LeWinn
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA, United States; Department of Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Frances Tylavsky
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Catherine J Karr
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States; Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States; Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, United States
| | - Sheela Sathyanarayana
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, United States; Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, United States; Seattle Children's Research Institute, Seattle, WA, United States
| |
Collapse
|
15
|
Ekpruke CD, Silveyra P. Sex Differences in Airway Remodeling and Inflammation: Clinical and Biological Factors. FRONTIERS IN ALLERGY 2022; 3:875295. [PMID: 35769576 PMCID: PMC9234861 DOI: 10.3389/falgy.2022.875295] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Asthma is characterized by an increase in the contraction and inflammation of airway muscles, resulting in airflow obstruction. The prevalence of asthma is lower in females than in males until the start of puberty, and higher in adult women than men. This sex disparity and switch at the onset of puberty has been an object of debate among many researchers. Hence, in this review, we have summarized these observations to pinpoint areas needing more research work and to provide better sex-specific diagnosis and management of asthma. While some researchers have attributed it to the anatomical and physiological differences in the male and female respiratory systems, the influences of hormonal interplay after puberty have also been stressed. Other hormones such as leptin have been linked to the sex differences in asthma in both obese and non-obese patients. Recently, many scientists have also demonstrated the influence of the sex-specific genomic framework as a key player, and others have linked it to environmental, social lifestyle, and occupational exposures. The majority of studies concluded that adult men are less susceptible to developing asthma than women and that women display more severe forms of the disease. Therefore, the understanding of the roles played by sex- and gender-specific factors, and the biological mechanisms involved will help develop novel and more accurate diagnostic and therapeutic plans for sex-specific asthma management.
Collapse
Affiliation(s)
- Carolyn Damilola Ekpruke
- Department of Environmental and Occupational Health, Indiana University Bloomington School of Public Health, Bloomington, IN, United States
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, Indiana University Bloomington School of Public Health, Bloomington, IN, United States
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Patricia Silveyra
| |
Collapse
|
16
|
Carnino JM, Lee H, Smith LC, Sunil VR, Rancourt RC, Vayas K, Cervelli J, Kwok ZH, Ni K, Laskin JD, Jin Y, Laskin DL. Microvesicle-Derived miRNAs Regulate Proinflammatory Macrophage Activation in the Lung Following Ozone Exposure. Toxicol Sci 2022; 187:162-174. [PMID: 35201360 PMCID: PMC9041552 DOI: 10.1093/toxsci/kfac025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Ozone is a ubiquitous air pollutant that causes lung damage and altered functioning. Evidence suggests that proinflammatory macrophages contribute to ozone toxicity. Herein, we analyzed the role of extracellular vesicles (EVs) and microRNA (miRNA) cargo in ozone-induced macrophage activation. Exposure of mice to ozone (0.8 ppm, 3 h) resulted in increases in bronchoalveolar lavage fluid EVs, which were comprised predominantly of microvesicles (MVs). NanoFACS analysis revealed that MVs generated following both air and ozone exposure was largely from CD45+ myeloid cells; these MVs were readily taken up by macrophages. Functionally, MVs from ozone, but not air treated mice, upregulated mRNA expression of inflammatory proteins in macrophages including inducible nitric oxide synthase (iNOS), CXCL-1, CXCL-2, and interleukin (IL)-1β. The miRNA profile of MVs in bronchoalveolar lavage fluid (BALF) was altered after ozone exposure; thus, increases in miR-21, miR-145, miR320a, miR-155, let-7b, miR744, miR181, miR-17, miR-92a, and miR-199a-3p were observed, whereas miR-24-3p and miR-20 were reduced. Ingenuity pathway analysis revealed that these miRNAs regulate pathways that promote inflammatory macrophage activation, and predicted that let-7a-5p/let-7b, miR-24-3p, miR-21-5p, miR-17, and miR-181a-5p are key upstream regulators of inflammatory proteins. After ozone exposure, miR-199a-3p, but not precursor miR-199a-3p, was increased in lung macrophages, indicating that it is derived from MV-mediated delivery. Furthermore, lung macrophage mRNA expression of IL-1β was upregulated after administration of MVs containing miR-199a-3p mimic but downregulated by miR-199a-3p inhibitor. Collectively, these data suggest that MVs generated following ozone exposure contribute to proinflammatory macrophage activation via MV-derived miRNAs including miR-199a-3p. These findings identify a novel pathway regulating macrophage inflammatory responses to inhaled ozone.
Collapse
Affiliation(s)
- Jonathan M Carnino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Boston University, Boston, Massachusetts 02118, USA
| | - Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Boston University, Boston, Massachusetts 02118, USA
| | - Ley Cody Smith
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Vasanthi R Sunil
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Raymond C Rancourt
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Kinal Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Jessica Cervelli
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Zhi Hao Kwok
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Boston University, Boston, Massachusetts 02118, USA
| | - Kareemah Ni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Boston University, Boston, Massachusetts 02118, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Boston University, Boston, Massachusetts 02118, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
17
|
Tovar A, Crouse WL, Smith GJ, Thomas JM, Keith BP, McFadden KM, Moran TP, Furey TS, Kelada SNP. Integrative analysis reveals mouse strain-dependent responses to acute ozone exposure associated with airway macrophage transcriptional activity. Am J Physiol Lung Cell Mol Physiol 2022; 322:L33-L49. [PMID: 34755540 PMCID: PMC8721896 DOI: 10.1152/ajplung.00237.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 01/03/2023] Open
Abstract
Acute ozone (O3) exposure is associated with multiple adverse cardiorespiratory outcomes, the severity of which varies across individuals in human populations and inbred mouse strains. However, molecular determinants of response, including susceptibility biomarkers that distinguish who will develop severe injury and inflammation, are not well characterized. We and others have demonstrated that airway macrophages (AMs) are an important resident immune cell type that are functionally and transcriptionally responsive to O3 inhalation. Here, we sought to explore influences of strain, exposure, and strain-by-O3 exposure interactions on AM gene expression and identify transcriptional correlates of O3-induced inflammation and injury across six mouse strains, including five Collaborative Cross (CC) strains. We exposed adult mice of both sexes to filtered air (FA) or 2 ppm O3 for 3 h and measured inflammatory and injury parameters 21 h later. Mice exposed to O3 developed airway neutrophilia and lung injury with strain-dependent severity. In AMs, we identified a common core O3 transcriptional response signature across all strains, as well as a set of genes exhibiting strain-by-O3 exposure interactions. In particular, a prominent gene expression contrast emerged between a low- (CC017/Unc) and high-responding (CC003/Unc) strain, as reflected by cellular inflammation and injury. Further inspection indicated that differences in their baseline gene expression and chromatin accessibility profiles likely contribute to their divergent post-O3 exposure transcriptional responses. Together, these results suggest that aspects of O3-induced respiratory responses are mediated through altered AM transcriptional signatures and further confirm the importance of gene-environment interactions in mediating differential responsiveness to environmental agents.
Collapse
Affiliation(s)
- Adelaide Tovar
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Genetics & Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Wesley L Crouse
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Gregory J Smith
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph M Thomas
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin P Keith
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathryn M McFadden
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Timothy P Moran
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Terrence S Furey
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Genetics & Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Samir N P Kelada
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Genetics & Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Bioinformatics & Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
18
|
Kanjanapruthipong T, Sukphopetch P, Reamtong O, Isarangkul D, Muangkaew W, Thiangtrongjit T, Sansurin N, Fongsodsri K, Ampawong S. Cytoskeletal Alteration Is an Early Cellular Response in Pulmonary Epithelium Infected with Aspergillus fumigatus Rather than Scedosporium apiospermum. MICROBIAL ECOLOGY 2022; 83:216-235. [PMID: 33890146 DOI: 10.1007/s00248-021-01750-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/02/2021] [Indexed: 06/12/2023]
Abstract
Invasive aspergillosis and scedosporiosis are life-threatening fungal infections with similar clinical manifestations in immunocompromised patients. Contrarily, Scedosporium apiospermum is susceptible to some azole derivative but often resistant to amphotericin B. Histopathological examination alone cannot diagnose these two fungal species. Pathogenesis studies could contribute to explore candidate protein markers for new diagnosis and treatment methods leading to a decrease in mortality. In the present study, proteomics was conducted to identify significantly altered proteins in A549 cells infected with or without Aspergillus fumigatus and S. apiospermum as measured at initial invasion. Protein validation was performed with immunogold labelling alongside immunohistochemical techniques in infected A549 cells and lungs from murine models. Further, cytokine production was measured, using the Bio-Plex-Multiplex immunoassay. The cytoskeletal proteins HSPA9, PA2G4, VAT1, PSMA2, PEX1, PTGES3, KRT1, KRT9, CLIP1 and CLEC20A were mainly changed during A. fumigatus infection, while the immunologically activated proteins WNT7A, GAPDH and ANXA2 were principally altered during S. apiospermum infection. These proteins are involved in fungal internalisation and structural destruction leading to pulmonary disorders. Interleukin (IL)-21, IL-1α, IL-22, IL-2, IL-8, IL-12, IL-17A, interferon-γ and tumour necrosis factor-α were upregulated in both aspergillosis and scedosporiosis, although more predominately in the latter, in accordance with chitin synthase-1 and matrix metalloproteinase levels. Our results demonstrated that during invasion, A. fumigatus primarily altered host cellular integrity, whereas S. apiospermum chiefly induced and extensively modulated host immune responses.
Collapse
Affiliation(s)
- Tapanee Kanjanapruthipong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Passanesh Sukphopetch
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetic, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Duangnate Isarangkul
- Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Watcharamat Muangkaew
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Tipparat Thiangtrongjit
- Department of Molecular Tropical Medicine and Genetic, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Nichapa Sansurin
- Northeast Laboratory Animal Center, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kamonpan Fongsodsri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
19
|
Relationships between Long-Term Ozone Exposure and Allergic Rhinitis and Bronchitic Symptoms in Chinese Children. TOXICS 2021; 9:toxics9090221. [PMID: 34564372 PMCID: PMC8472948 DOI: 10.3390/toxics9090221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
Numerous studies have demonstrated that exposure to ambient ozone (O3) could have adverse effects on children's respiratory health. However, previous studies mainly focused on asthma and wheezing. Evidence for allergic rhinitis and bronchitic symptoms (e.g., persistent cough and phlegm) associated with O3 is limited, and results from existing studies are inconsistent. This study included a total of 59,754 children from the seven northeastern cities study (SNEC), who were aged 2 to 17 years and from 94 kindergarten, elementary and middle schools. Information on doctor-diagnosed allergic rhinitis (AR), persistent cough, and persistent phlegm was collected during 2012-2013 using a standardized questionnaire developed by the American Thoracic Society (ATS). Information for potential confounders was also collected via questionnaire. Individuals' exposure to ambient ozone (O3) during the four years before the investigation was estimated using a satellite-based random forest model. A higher level of O3 was significantly associated with increased risk of AR and bronchitic symptoms. After controlling for potential confounders, the OR (95% CI) were 1.13 (1.07-1.18), 1.10 (1.06-1.16), and 1.12 (1.05-1.20) for AR, persistent cough, and persistent phlegm, respectively, associated with each interquartile range (IQR) rise in O3 concentration. Interaction analyses showed stronger adverse effects of O3 on AR in children aged 7-17 years than those aged 2-6 years, while the adverse association of O3 with cough was more prominent in females and children aged 7-12 years than in males and children aged 2-6 and 13-17 years. This study showed that long-term exposure to ambient O3 was significantly associated with higher risk of AR and bronchitic symptoms in children, and the association varies across age and gender. Our findings contribute additional evidence for the importance of controlling O3 pollution and protecting children from O3 exposure.
Collapse
|
20
|
Binda MM, Riiskjaer M, Koninckx PR. Pneumoperitoneum induced mesothelial cell changes in a laparoscopic mouse model. Eur J Obstet Gynecol Reprod Biol 2021; 265:107-112. [PMID: 34482234 DOI: 10.1016/j.ejogrb.2021.08.016] [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: 04/06/2021] [Revised: 07/03/2021] [Accepted: 08/19/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND CO2 pneumoperitoneum (PP) during laparoscopic surgery, can cause hypoxia and desiccation in the peritoneal mesothelial cell, resulting in a time-dependent retraction and bulging of these cells, an acute inflammatory reaction and enhanced adhesion formation. Since hypoxia is prevented by adding 4% of oxygen (O2) to the CO2 PP, the aim of this study was to evaluate the effect of adding 4% O2 to the CO2 PP on mesothelial cell morphology. METHODS In a standardized laparoscopic mouse model (n=8 mice per group), a control group with a 30- or 60-min PP with humidified CO2 + 4% of O2 (groups I and II) was compared to a hypoxic group with 30- or 60-min humidified pure CO2 (groups III and IV) and a desiccation group with 60-min of dry CO2 PP (group V). The effect upon the peritoneum morphology was evaluated by scanning electron microscopy (SEM) of abdominal wall peritoneal biopsies. Biopsies, taken immediately (n=4) and 24 hrs (n=4) after surgery, were compared to a group without PP (group VI, n=4). SEM pictures were blindly scored for cell retraction, deletion of microvilli, fibrin deposition, holes in the epithelial layer and visibility of cell borders using a semi-quantitative scoring system. RESULTS PP Hypoxia (CO2 PP) has a deleterious effect upon mesothelial morphology, immediately (holes: p= 0.04) and 24 hrs later (cell retraction: p=0.005; total score: p=0.03) . Desiccation has also a deleterious effect immediately (microvilli p=0.0090; fibrin deposition p=0.05) and 24 hrs after surgery (cell retraction: p=0.0036; holes: p=0.0004; microvilli: p< 0.0001, fibrin deposition: p=0.0225; borders: p=0.0007). This deleterious effect increases with duration of CO2 PP, affecting cell retraction (p=0.016), holes (p=0.0441), and the total score (p=0.0488). The addition of 4% of O2 to the CO2 PP failed to reach statistical significance. CONCLUSIONS These data confirm that CO2 PP and dry gas have a deleterious effect on mesothelial cell morphology. Humidification of the insufflation gas reduces this deleterious effect. The hypothesis of a protective effect of adding O2 failed to reach significance.
Collapse
Affiliation(s)
- Maria Mercedes Binda
- Department of Obstetrics and Gynaecology, University Hospital Gasthuisberg, Katholieke Universiteit Leuven (KULeuven), Leuven, Belgium.
| | - Mads Riiskjaer
- Department of Obstetrics and Gynaecology, University Hospital Gasthuisberg, Katholieke Universiteit Leuven (KULeuven), Leuven, Belgium.
| | - Philippe Robert Koninckx
- Department of Obstetrics and Gynaecology, University Hospital Gasthuisberg, Katholieke Universiteit Leuven (KULeuven), Leuven, Belgium.
| |
Collapse
|
21
|
Yaeger MJ, Reece SW, Kilburg-Basnyat B, Hodge MX, Pal A, Dunigan-Russell K, Luo B, You DJ, Bonner JC, Spangenburg EE, Tokarz D, Hannan J, Armstrong M, Manke J, Reisdorph N, Tighe RM, Shaikh SR, Gowdy KM. Sex Differences in Pulmonary Eicosanoids and Specialized Pro-Resolving Mediators in Response to Ozone Exposure. Toxicol Sci 2021; 183:170-183. [PMID: 34175951 DOI: 10.1093/toxsci/kfab081] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ozone (O3) is a criteria air pollutant known to increase the morbidity and mortality of cardiopulmonary diseases. This occurs through a pulmonary inflammatory response characterized by increased recruitment of immune cells into the airspace, pro-inflammatory cytokines, and pro-inflammatory lipid mediators. Recent evidence has demonstrated sex-dependent differences in the O3-induced pulmonary inflammatory response. However, it is unknown if this dimorphic response is evident in pulmonary lipid mediator metabolism. We hypothesized that there are sex-dependent differences in lipid mediator production following acute O3 exposure. Male and female C57BL/6J mice were exposed to 1 part per million O3 for 3 hours and were necropsied at 6 or 24 hours following exposure. Lung lavage was collected for cell differential and total protein analysis, and lung tissue was collected for mRNA analysis, metabololipidomics, and immunohistochemistry. Compared to males, O3-exposed female mice had increases in airspace neutrophilia, neutrophil chemokine mRNA, pro-inflammatory eicosanoids such as prostaglandin E2, and specialized pro-resolving mediators (SPMs) such as resolvin D5 in lung tissue. Likewise, precursor fatty acids (arachidonic and docosahexaenoic acid; DHA) were increased in female lung tissue following O3 exposure compared to males. Experiments with ovariectomized females revealed that loss of ovarian hormones exacerbates pulmonary inflammation and injury. However, eicosanoid and SPM production were not altered by ovariectomy despite depleted pulmonary DHA concentrations. Taken together, these data indicate that O3 drives an increased pulmonary inflammatory and bioactive lipid mediator response in females. Furthermore, ovariectomy increases susceptibility to O3-induced pulmonary inflammation and injury, as well as decreases pulmonary DHA concentrations.
Collapse
Affiliation(s)
- M J Yaeger
- Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210
| | - S W Reece
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, 27858
| | - B Kilburg-Basnyat
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, 27858
| | - M X Hodge
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, 27858
| | - A Pal
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599
| | - K Dunigan-Russell
- Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210
| | - B Luo
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, 27858
| | - D J You
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27107
| | - J C Bonner
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27107
| | - E E Spangenburg
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27858
| | - D Tokarz
- Experimental Pathology Laboratories, Inc, Research Triangle Park, NC, 27709
| | - J Hannan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27858
| | - M Armstrong
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO, 80045
| | - J Manke
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO, 80045
| | - N Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO, 80045
| | - R M Tighe
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710
| | - S R Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599
| | - K M Gowdy
- Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210
| |
Collapse
|
22
|
Duran-Ortiz S, List EO, Basu R, Kopchick JJ. Extending lifespan by modulating the growth hormone/insulin-like growth factor-1 axis: coming of age. Pituitary 2021; 24:438-456. [PMID: 33459974 PMCID: PMC8122064 DOI: 10.1007/s11102-020-01117-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Progress made in the years of aging research have allowed the opportunity to explore potential interventions to slow aging and extend healthy lifespan. Studies performed in yeast, worms, flies and mice subjected to genetic and pharmacological interventions have given insight into the cellular and molecular mechanisms associated with longevity. Furthermore, it is now possible to effectively modulate pathways that slow aging at different stages of life (early life or at an adult age). Interestingly, interventions that extend longevity in adult mice have had sex-specific success, suggesting a potential link between particular pathways that modulate aging and sex. For example, reduction of the growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis at an adult age extends lifespan preferentially in females. Moreover, several postnatal dietary interventions tested by the 'Intervention Testing Program (ITP)' from the National Institute of Aging (NIA) have shown that while pharmacological interventions like rapamycin affect the IGF-1/insulin pathway and preferentially extend lifespan in females; dietary compounds that target other cellular pathways are effective only in male mice-indicating mutually exclusive sex-specific pathways. Therefore, a combination of interventions that target non-overlapping aging-related pathways appears to be an effective approach to further extend healthy lifespan in both sexes. Here, we review the germline and postnatal mouse lines that target the GH/IGF-1 axis as a mechanism to extend longevity as well as the dietary compounds that tested positive in the NIA program to increase lifespan. We believe that the interventions reviewed in this paper could constitute feasible combinations for an extended healthy lifespan in both male and female mice.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, USA.
- Molecular and Cellular Biology Program, Ohio University, Athens, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
23
|
Guttenberg MA, Vose AT, Tighe RM. Role of Innate Immune System in Environmental Lung Diseases. Curr Allergy Asthma Rep 2021; 21:34. [PMID: 33970346 PMCID: PMC8311569 DOI: 10.1007/s11882-021-01011-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2021] [Indexed: 01/07/2023]
Abstract
The lung mucosa functions as a principal barrier between the body and inhaled environmental irritants and pathogens. Precise and targeted surveillance mechanisms are required at this lung-environment interface to maintain homeostasis and preserve gas exchange. This is performed by the innate immune system, a germline-encoded system that regulates initial responses to foreign irritants and pathogens. Environmental pollutants, such as particulate matter (PM), ozone (O3), and other products of combustion (NO2, SO3, etc.), both stimulate and disrupt the function of the innate immune system of the lung, leading to the potential for pathologic consequences. PURPOSE OF REVIEW: The purpose of this review is to explore recent discoveries and investigations into the role of the innate immune system in responding to environmental exposures. This focuses on mechanisms by which the normal function of the innate immune system is modified by environmental agents leading to disruptions in respiratory function. RECENT FINDINGS: This is a narrative review of mechanisms of pulmonary innate immunity and the impact of environmental exposures on these responses. Recent findings highlighted in this review are categorized by specific components of innate immunity including epithelial function, macrophages, pattern recognition receptors, and the microbiome. Overall, the review supports broad impacts of environmental exposures to alterations to normal innate immune functions and has important implications for incidence and exacerbations of lung disease. The innate immune system plays a critical role in maintaining pulmonary homeostasis in response to inhaled air pollutants. As many of these agents are unable to be mitigated, understanding their mechanistic impact is critical to develop future interventions to limit their pathologic consequences.
Collapse
Affiliation(s)
| | | | - Robert M. Tighe
- Department of Medicine, Duke University, Durham, NC,Corresponding Author: Robert M Tighe, MD, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Box 2969, Durham, North Carolina 27710, Telephone: 919-684-4894, Fax: 919-684-5266,
| |
Collapse
|
24
|
Tsamou M, Vrijens K, Wang C, Winckelmans E, Neven KY, Madhloum N, de Kok TM, Nawrot TS. Genome-wide microRNA expression analysis in human placenta reveals sex-specific patterns: an ENVIR ONAGE birth cohort study. Epigenetics 2021; 16:373-388. [PMID: 32892695 PMCID: PMC7993149 DOI: 10.1080/15592294.2020.1803467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 07/03/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022] Open
Abstract
There is an increasing interest in microRNAs (miRNAs) as they are of utmost importance in gene regulation at the posttranscriptional level. Sex-related susceptibility for non-communicable diseases later in life could originate in early life. Until now, no data on sex-specific miRNA expression are available for the placenta. Therefore, we investigated the difference by sex of newborn's miRNA expression in human placental tissue. Within the ENVIRONAGE birth cohort, miRNA and mRNA expression profiling was performed in 60 placentae (50% boys) using Agilent (8 × 60 K) microarrays. The distribution of chromosome locations was studied and pathway analysis of the identified sex-specific miRNAs in the placenta was carried out. Of the total 2558 miRNAs on the array, 597 miRNAs were expressed in over 70% of the samples and were included for further analyses. A total of 142 miRNAs were significantly (FDR<0.05) associated with the newborn's sex. In newborn girls, 76 miRNAs had higher expression (hsa-miR-361-5p as most significant) and 66 miRNAs had lower expression (hsa-miR-4646-5p as most significant) than in newborn boys. In the same study population, placental differentially expressed genes by sex were also identified using a whole genome approach. The placental gene expression revealed 27 differentially expressed genes by comparing girls to boys. Ultimately, we studied the miRNA-RNA interactome and identified 14 miRNA-mRNA interactions as sex-specific. Sex differences in placental m(i)RNA expression may reveal sex-specific patterns already present during pregnancy, which may influence physiological conditions in early or later life. These molecular processes might play a role in sex-specific disease susceptibility in later life.
Collapse
Affiliation(s)
- Maria Tsamou
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Karen Vrijens
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Congrong Wang
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Ellen Winckelmans
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Kristof Y. Neven
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Narjes Madhloum
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Theo M. de Kok
- Department of Toxicogenomics, GROW Institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Tim S. Nawrot
- Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Public Health, Environment & Health Unit, Leuven University (KU Leuven), Leuven, Belgium
| |
Collapse
|
25
|
Siddaiah R, Oji-Mmuo CN, Montes DT, Fuentes N, Spear D, Donnelly A, Silveyra P. MicroRNA Signatures Associated with Bronchopulmonary Dysplasia Severity in Tracheal Aspirates of Preterm Infants. Biomedicines 2021; 9:biomedicines9030257. [PMID: 33807742 PMCID: PMC8000397 DOI: 10.3390/biomedicines9030257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/04/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a form of chronic lung disease that develops in neonates as a consequence of preterm birth, arrested fetal lung development, and inflammation. The incidence of BPD remains on the rise as a result of increasing survival of extremely preterm infants. Severe BPD contributes to significant health care costs and is associated with prolonged hospitalizations, respiratory infections, and neurodevelopmental deficits. In this study, we aimed to detect novel biomarkers of BPD severity. We collected tracheal aspirates (TAs) from preterm babies with mild/moderate (n = 8) and severe (n = 17) BPD, and we profiled the expression of 1048 miRNAs using a PCR array. Associations with biological pathways were determined with the Ingenuity Pathway Analysis (IPA) software. We found 31 miRNAs differentially expressed between the two disease groups (2-fold change, false discovery rate (FDR) < 0.05). Of these, 4 miRNAs displayed significantly higher expression levels, and 27 miRNAs had significantly lower expression levels in the severe BPD group when compared to the mild/moderate BPD group. IPA identified cell signaling and inflammation pathways associated with miRNA signatures. We conclude that TAs of extremely premature infants contain miRNA signatures associated with severe BPD. These may serve as potential biomarkers of disease severity in infants with BPD.
Collapse
Affiliation(s)
- Roopa Siddaiah
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.S.); (C.N.O.-M.); (D.S.); (A.D.)
| | - Christiana N. Oji-Mmuo
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.S.); (C.N.O.-M.); (D.S.); (A.D.)
| | - Deborah T. Montes
- Biobehavioral Laboratory, School of Nursing, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Nathalie Fuentes
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Debra Spear
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.S.); (C.N.O.-M.); (D.S.); (A.D.)
| | - Ann Donnelly
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.S.); (C.N.O.-M.); (D.S.); (A.D.)
| | - Patricia Silveyra
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (R.S.); (C.N.O.-M.); (D.S.); (A.D.)
- Biobehavioral Laboratory, School of Nursing, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA
- Correspondence:
| |
Collapse
|
26
|
Oji-Mmuo CN, Siddaiah R, Montes DT, Pham MA, Spear D, Donnelly A, Fuentes N, Imamura-Kawasawa Y, Howrylak JA, Thomas NJ, Silveyra P. Tracheal aspirate transcriptomic and miRNA signatures of extreme premature birth with bronchopulmonary dysplasia. J Perinatol 2021; 41:551-561. [PMID: 33177681 DOI: 10.1038/s41372-020-00868-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/06/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Extreme preterm infants are a growing population in neonatal intensive care units who carry a high mortality and morbidity. Multiple factors play a role in preterm birth, resulting in major impact on organogenesis leading to complications including bronchopulmonary dysplasia (BPD). The goal of this study was to identify biomarker signatures associated with prematurity and BPD. STUDY DESIGN We analyzed miRNA and mRNA profiles in tracheal aspirates (TAs) from 55 infants receiving invasive mechanical ventilation. Twenty-eight infants were extremely preterm and diagnosed with BPD, and 27 were term babies receiving invasive mechanical ventilation for elective procedures. RESULT We found 22 miRNAs and 33 genes differentially expressed (FDR < 0.05) in TAs of extreme preterm infants with BPD vs. term babies without BPD. Pathway analysis showed associations with inflammatory response, cellular growth/proliferation, and tissue development. CONCLUSIONS Specific mRNA-miRNA signatures in TAs may serve as biomarkers for BPD pathogenesis, a consequence of extreme prematurity.
Collapse
Affiliation(s)
| | - Roopa Siddaiah
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Deborah T Montes
- Biobehavioral Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melody A Pham
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Debra Spear
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Ann Donnelly
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Nathalie Fuentes
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Yuka Imamura-Kawasawa
- Institute for Personalized Medicine, Departments of Biochemistry and Molecular Biology and Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Judie A Howrylak
- Division of Pulmonary and Critical Care Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Neal J Thomas
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Patricia Silveyra
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA. .,Biobehavioral Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
27
|
Kim SY, Bang M, Wee JH, Min C, Yoo DM, Han SM, Kim S, Choi HG. Short- and long-term exposure to air pollution and lack of sunlight are associated with an increased risk of depression: A nested case-control study using meteorological data and national sample cohort data. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 757:143960. [PMID: 33321334 DOI: 10.1016/j.scitotenv.2020.143960] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/13/2020] [Accepted: 11/14/2020] [Indexed: 06/12/2023]
Abstract
Previous studies have suggested an increased risk of depression related to air pollutants. This study investigated the relationship of air pollutant exposure and meteorological factors with depression. The Korean National Health Insurance Service-Health Screening Cohort from 2002 to 2013 was analyzed. In total, 25,589 depression participants were 1:4 matched with 102,356 control participants for age, sex, income, and region of residence. Depression was defined based on a diagnosis (ICD-10: F31-33) by a psychiatric physician. Meteorological factors and air pollutants including sulfur dioxide (SO2) (ppm), nitrogen dioxide (NO2) (ppm), ozone (O3) (ppm), carbon monoxide (CO) (ppm), and particulate matter with an aerodynamic diameter <10 μm (PM10) (μg/m3) during the 30 days and 365 days before the index date were analyzed for associations with depression using conditional logistic regression. Subgroup analyses were performed according to age, sex, income, and region of residence. The odds ratios (ORs) for depression were 1.05 (95% CI = 1.02-1.08) at 365 days for 1 h less of sunshine. The ORs for depression were 1.02 (95% CI = 1.01-1.03) and 1.03 (95% CI = 1.00-1.05) at 30 days and 365 days for PM10 (10 μg/m3), respectively. The ORs for depression were 1.18 (95% CI = 1.04-1.35) and 1.25 (95% CI = 1.07-1.47) at 30 days and 365 days for CO (ppm), respectively. In the subgroup analyses, the overall results were consistent. However, statistical significance diminished in the younger, high-income, and urban resident subgroups. Both short- and long-term exposure to PM10 and CO and a reduced duration of sunshine were related to an increased risk of depression.
Collapse
Affiliation(s)
- So Young Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Minji Bang
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Jee Hye Wee
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Chanyang Min
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang, Republic of Korea; Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Dae Myoung Yoo
- Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Sang-Min Han
- Political Science (Climate and Environmental Policy), Graduate School of Global Cooperation, Hallym University, Chuncheon, Republic of Korea
| | - Seungdo Kim
- Research Center for Climate Change and Energy, Hallym University, Chuncheon, Republic of Korea
| | - Hyo Geun Choi
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Anyang, Republic of Korea; Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang, Republic of Korea; Hallym Institute for Environmental Diseases (HIED), Chuncheon, Republic of Korea.
| |
Collapse
|
28
|
A novel and distinctive mode of cell death revealed by using non-thermal atmospheric pressure plasma: The involvements of reactive oxygen species and the translation inhibitor Pdcd4. Chem Biol Interact 2021; 338:109403. [PMID: 33582111 DOI: 10.1016/j.cbi.2021.109403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/22/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022]
Abstract
Cells death is indispensable for embryonic development, tissue homeostasis, and the elimination of cancer, virally infected, or degenerated cells in multicellular organisms. It occurs not only via existing modes but also via unidentified modes, whose elucidation requires. Exposure to non-thermal atmospheric pressure plasma (NTAPP) has been demonstrated to induce cell death, probably because of its ability to generate reactive oxygen species (ROS). However, the mode of this cell death and its underlying mechanism remained elusive. Here we show cell death occurring in a novel and distinctive mode different from apoptosis and necrosis/necroptosis through a mechanism that ROS mediate the loss of the translation inhibitor Programmed cell death 4 (Pdcd4) when cells are cultured in solutions activated by NTAPP irradiation. Thus, our study performed with NTAPP-activated solutions may provide insight into the existence of the atypical cell death in cells and some features of its distinguishing mode and underlying mechanism.
Collapse
|
29
|
Wang X, Zhou X, Xia X, Zhang Y. Estradiol attenuates LPS-induced acute lung injury via induction of aquaporins AQP1 and AQP5. EUR J INFLAMM 2021; 19:205873922110491. [DOI: 10.1177/20587392211049197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
Abstract
Background Acute lung injury (ALI) is associated with increased inflammation and oxidative stress. Estradiol is produced by the ovaries and is the most active hormone of estrogen. Our aim was to investigate whether estradiol contributes to protect against lipopolysaccharide (LPS)-induced ALI via induction of aquaporins AQP1 and AQP5 and the underlying mechanisms. Methods and results For induction of ALI, LPS was applied once by intraperitoneal injection in SD rats 14 days after oophorectomy. To assess the therapeutic effects of estradiol on LPS-induced ALI, estradiol was subcutaneously injected for 1 h prior to LPS challenge. Estradiol can significantly attenuate the lung edema reflected by decreasing wet-to-dry weight ratio and permeability of lung and total protein concentration of bronchial lavage fluid (BALF). Results of histological detection showed that estradiol attenuated the lung injury reflected by reducing edema, congestion, and thickening pulmonary septal of lung tissues. In addition, estradiol attenuated TNF-α, IL-1β, and IL-6 and oxidative stress in lung tissues. Estradiol was more effective than estradiol associated with ERα antagonist or ERβ antagonist in protecting against LPS-induced ALI in rats. Mechanistically, we investigate whether estradiol regulates the expression of AQP1 and AQP5 in lung tissues. Of interest, estradiol upregulates AQP1 and AQP5 mRNA and protein expression. Taken together, these results demonstrate that estradiol can increase the expression of AQP1 and AQP5, which plays a critical role in ameliorating oxidative stress and downregulating inflammatory responses induced by LPS.Conclusion Therefore, these findings strongly suggest that AQP1 and AQP5 mediate the anti-inflammatory and antioxidant effects of estradiol.
Collapse
Affiliation(s)
- Xiaobo Wang
- Internal Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiuyun Zhou
- Blood Purification Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiumei Xia
- Department of Imaging Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yili Zhang
- Health Management Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
30
|
Wei Y, Han B, Dai W, Guo S, Zhang C, Zhao L, Gao Y, Jiang Y, Kong X. Exposure to ozone impacted Th1/Th2 imbalance of CD 4+ T cells and apoptosis of ASMCs underlying asthmatic progression by activating lncRNA PVT1-miR-15a-5p/miR-29c-3p signaling. Aging (Albany NY) 2020; 12:25229-25255. [PMID: 33223504 PMCID: PMC7803560 DOI: 10.18632/aging.104124] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
This investigation attempted to elucidate whether lncRNA PVT1-led miRNA axes participated in aggravating ozone-triggered asthma progression. One hundred and sixty-eight BALB/c mice were evenly divided into saline+air group, ovalbumin+air group, saline+ozone group and ovalbumin+ozone group. Correlations were evaluated between PVT1 expression and airway smooth muscle function/inflammatory cytokine release among the mice models. Furthermore, pcDNA3.1-PVT1 and si-PVT1 were, respectively, transfected into CD4+T cells and airway smooth muscle cells (ASMCs), and activities of the cells were observed. Ultimately, a cohort of asthma patients was recruited to estimate the diagnostic performance of PVT1. It was demonstrated that mice of ovalbumin+ozone group were associated with higher PVT1 expression, thicker trachea/airway smooth muscle and smaller ratio of Th1/Th2-like cytokines than mice of ovalbumin+air group and saline+ozone group (P<0.05). Moreover, pcDNA3.1-PVT1 significantly brought down Th1/Th2 ratio in CD4+ T cells by depressing miR-15a-5p expression and activating PI3K-Akt-mTOR signaling (P<0.05). The PVT1 also facilitated ASMC proliferation by sponging miR-29c-3p and motivating PI3K-Akt-mTOR signaling (P<0.05). Additionally, PVT1 seemed promising in diagnosis of asthma, with favorable sensitivity (i.e. 0.844) and specificity (i.e. 0.978). Conclusively, lncRNA PVT1-miR-15a-5p/miR-29c-3p-PI3K-Akt-mTOR axis was implicated in ozone-induced asthma development by promoting ASMC proliferation and Th1/Th2 imbalance.
Collapse
Affiliation(s)
- Yangyang Wei
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Baofen Han
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Wenjuan Dai
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Shufang Guo
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Caiping Zhang
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Lixuan Zhao
- Department of Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Yan Gao
- Department of Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Yi Jiang
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xiaomei Kong
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
31
|
Transcriptomic analysis reveals the role of a peptide derived from CRYAB on the CoCl 2-induced hypoxic HL-1 cardiomyocytes. J Thromb Thrombolysis 2020; 51:265-276. [PMID: 32621152 DOI: 10.1007/s11239-020-02117-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Acute myocardial infarction (AMI) is a life-threatening disease that often results in heart failure. CRYAB, a small heat shock protein, has been shown to have cardioprotective effects against oxidative stress-induced apoptosis in AMI. Previously, we purified a peptide derived from CRYAB (LEDQFFGEH), which we named PDFC. In this study, we determined the function of PDFC on HL-1 cardiomyocytes and explored the mechanism underlying its function. A hypoxic myocardiocyte cell line was generated by stimulation of HL-1 mouse cardiac muscle cells with different concentrations of CoCl2. Then, the hypoxic HL-1 cells were treated with the synthetic PDFC peptide, and cell proliferation, migration, and apoptosis were assessed to examine the effects of PDFC on HL-1 and hypoxic HL-1 cells. To examine the mechanism underlying the effects of PDFC on hypoxic cells, PDFC-treated hypoxic HL-1 cells were submitted for deep RNA sequencing. Finally, several differentially expressed genes in different pathways were selected for confirmation by RT-qPCR. Hypoxic myocardiocytes were generated by stimulating HL-1 cells with 800 µM CoCl2 for 24 h, which significantly upregulated HIF-1α. PDFC at 200 µg/ml showed the most positive effects on cell viability. Although hypoxic HL-1 cells and PDFC-treated hypoxic HL-1 cells both showed lower viability and migration and higher levels of apoptosis than untreated HL-1 cells, compared to hypoxic HL-1 cells, PDFC-treated hypoxic HL-1 cells showed higher viability and migration and lower apoptosis. The deep sequencing showed that 812 genes were upregulated and 1946 genes were downregulated. Among these differentially expressed genes, 699 of the upregulated genes and 1488 of the downregulated genes were protein-coding genes. Gene ontology and pathway enrichment analysis showed that the downregulated genes were dominant and that the PI3K-Akt pathway was located in the center of the network. A protein-protein interaction network was constructed, and 892 nodes were determined. In PDFC-treated hypoxic HL-1 cells, Fn1, Pik3r5, and Creb5 were downregulated, while Insr, Bcl2, Mapk14, and Pten were upregulated when compared to the levels in hypoxic HL-1 cells. In conclusion, this study reveals the significant bioactive effect of the CRYAB-derived peptide, PDFC on cardiomyocytes and the underlying mechanism.
Collapse
|
32
|
Wang T, Wang H, Chen J, Wang J, Ren D, Hu W, Wang H, Han W, Leng S, Zhang R, Zheng Y. Association between air pollution and lung development in schoolchildren in China. J Epidemiol Community Health 2020; 74:792-798. [PMID: 32527860 PMCID: PMC7577101 DOI: 10.1136/jech-2020-214283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/13/2020] [Accepted: 05/23/2020] [Indexed: 12/24/2022]
Abstract
Background China has been facing nationwide air pollution at unprecedented high levels primarily from fossil–fuel combustion in the past decade. However, few studies have been conducted on the adverse effect of severe air pollution on lung development in school-age children. Methods Using wellness check and air pollution data from 2014 to 2017, we conducted a retrospective analysis of lung development in 21 616 school-age children from Shijiazhuang and Qingdao from North China with severe vs mild air pollution. Linear mixed effects model was performed to assess the effect of air pollution on forced vital capacity (FVC) growth. Results Exposure to severe air pollution was associated with a dramatic reduction in annual FVC growth rate (−71.3 mL, p< 0.001). In addition, every 10 μg/m3 increase in annual PM2.5 level was associated with a reduction of annual FVC growth by 12.2 mL ( p< 0.001). Sex discrepancy (boys vs girls) in FVC growth was greater in Qingdao (35.4 mL/year, 95% CI: 26.0 to 44.7) than in Shijiazhuang (19.8 mL/year, 95% CI: 9.3 to 30.3) (p for interaction=0.063). Exposure to indoor coal- or wood-burning stove heating (−79.4 mL, p< 0.001) and secondhand smoke at home (−59.3 mL, p= 0.003) were inversely associated with FVC growth. Conclusion Our study raised serious alarm over the threat of severe air pollution to lung development in school-age children. Sex discrepancy in lung development was reduced dramatically in heavily polluted area.
Collapse
Affiliation(s)
- Tao Wang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Haitao Wang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Jian Chen
- Department of School Health, Qingdao Municipal Center for Disease Control and Prevention, Qingdao, China
| | - Jiansheng Wang
- Policy Research Center for Environment and Economy, Ministry of Ecology and Environment of the People's Republic of China, Beijing, China
| | - Dunqiang Ren
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Hu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Hongmei Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Han
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital Group, Qingdao, China
| | - Shuguang Leng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang City, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| |
Collapse
|
33
|
Qin Y, Zheng B, Yang GS, Zhou J, Yang HJ, Nie ZY, Wang TR, Zhang XH, Zhao HY, Shi JH, Wen JK. Tanshinone ⅡA inhibits VSMC inflammation and proliferation in vivo and in vitro by downregulating miR-712-5p expression. Eur J Pharmacol 2020; 880:173140. [PMID: 32387370 DOI: 10.1016/j.ejphar.2020.173140] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
The inflammation and proliferation of vascular smooth muscle cells (VSMCs) are the basic pathological feature of proliferative vascular diseases. Tanshinone ⅡA (Tan ⅡA), which is the most abundant fat-soluble element extracted from Salvia miltiorrhiza, has potent protective effects on the cardiovascular system. However, the underlying mechanism is still not fully understood. Here, we show that Tan ⅡA significantly inhibits neointimal formation and decreases VSMC inflammation by upregulating the expression of KLF4 and inhibiting the activation of NFκB signaling. Using a microRNA array analysis, we found that miR-712-5p expression is significantly upregulated in tumor necrosis factor alpha (TNF-α)-treated VSMCs. Loss- and gain-of-function experiments revealed that transfection of miR-712-5p mimic promotes, whereas depletion of miR-712-5p suppresses TNF-α-induced VSMC inflammation, leading to amelioration of intimal hyperplasia induced by carotid artery ligation. Moreover, depletion of miR-712-5p by its antagomir largely abrogates TNF-α-induced VSMC proliferation. Our findings suggest that miR-712-5p mediates the stimulatory effect of TNF-α on VSMC inflammation, and that Tan ⅡA inhibits VSMC inflammation and proliferation in vivo and in vitro by suppression of miR-712-5p expression. Targeting miR-712-5p may be a novel therapeutic strategy to prevent proliferative vascular diseases.
Collapse
Affiliation(s)
- Yan Qin
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China; Department of Central Laboratory, Affiliated Hospital of Hebei University, Baoding, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Gao-Shan Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China; Department of Biochemistry and Molecular Biology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jing Zhou
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China; Department of Endocrine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hao-Jie Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Zi-Yuan Nie
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tian-Rui Wang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Hong-Ye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Jian-Hong Shi
- Department of Central Laboratory, Affiliated Hospital of Hebei University, Baoding, China
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
34
|
Chen H, Xu Y, Rappold A, Diaz-Sanchez D, Tong H. Effects of ambient ozone exposure on circulating extracellular vehicle microRNA levels in coronary artery disease patients. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2020; 83:351-362. [PMID: 32414303 PMCID: PMC7306136 DOI: 10.1080/15287394.2020.1762814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Exposure to ambient air pollutants such as ozone (O3) and particulate matter (PM) is associated with increased cardiovascular morbidity and rate of mortality, but the underlying biological mechanisms have yet to be described. Emerging evidence shows that extracellular vehicle (EV) microRNAs (miRNAs) may facilitate cell-to-cell and organ-to-organ communications and play a role in the air pollution-induced cardiovascular effects. This study aims to explore the association between air pollutant exposure and miRNA changes related to cardiovascular diseases. Using a panel study design, 14 participants with coronary artery diseases were enrolled in this study. Each participant had up to 10 clinical visits and their plasma samples were collected and measured for expression of miRNA-21 (miR-21), miR-126, miR-146, miR-150, and miR-155. Mixed effects models adjusted for temperature, humidity, and season were used to examine the association between miRNA levels and exposure to 8-hr O3 or 24-hr PM2.5 up to 4 days prior. Results demonstrated that miR-150 expression was positively associated with O3 exposure at 1-4 days lag and 5day moving average while miR-155 expression tracked with O3 exposure at lag 0. No significant association was found between miRNA expression and ambient PM2.5 at any time point. β-blocker and diabetic medication usage significantly modified the correlation between O3 exposure and miR-150 expression where the link was more prominent among non-users. In conclusion, evidence indicated an association between exposure to ambient O3 and circulating levels of EV miR-150 and miR-155 was observed. These findings pointed to a future research direction involving miRNA-mediated mechanisms of O3-induced cardiovascular effects.
Collapse
Affiliation(s)
- Hao Chen
- Oak Ridge Institute of Science and Education, 100 ORAU Way, Oak Ridge, TN 37830, USA
| | - Yunan Xu
- Department of Psychiatry and Behavioral Sciences, Duke University, 905 W. Main Street, Durham, NC 27701, USA
| | - Ana Rappold
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC 27514, USA
| | - David Diaz-Sanchez
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC 27514, USA
| | - Haiyan Tong
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC 27514, USA
| |
Collapse
|
35
|
Kalidhindi RSR, Ambhore NS, Bhallamudi S, Loganathan J, Sathish V. Role of Estrogen Receptors α and β in a Murine Model of Asthma: Exacerbated Airway Hyperresponsiveness and Remodeling in ERβ Knockout Mice. Front Pharmacol 2020; 10:1499. [PMID: 32116656 PMCID: PMC7010956 DOI: 10.3389/fphar.2019.01499] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/19/2019] [Indexed: 01/18/2023] Open
Abstract
Epidemiological data suggests increased prevalence of asthma in females than males, suggesting a plausible role for sex-steroids, especially estrogen in the lungs. Estrogen primarily acts through estrogen-receptors (ERα and ERβ), which play a differential role in asthma. Our previous studies demonstrated increased expression of ERβ in asthmatic human airway smooth muscle (ASM) cells and its activation diminished ASM proliferation in vitro and airway hyperresponsiveness (AHR) in vivo in a mouse (wild-type, WT) model of asthma. In this study, we evaluated the receptor specific effect of circulating endogenous estrogen in regulating AHR and remodeling using ERα and ERβ knockout (KO) mice. C57BL/6J WT, ERα KO, and ERβ KO mice were challenged intranasally with a mixed-allergen (MA) or PBS. Lung function was measured using flexiVent followed by collection of broncho-alveolar lavage fluid for differential leukocyte count (DLC), histology using hematoxylin and eosin (H&E) and Sirius red-fast green (SRFG) and detecting αsmooth muscle actin (α-SMA), fibronectin and vimentin expression using immunofluorescence (IF). Resistance (Rrs), elastance (Ers), tissue-damping (G) and tissue-elasticity (H) were significantly increased, whereas compliance (Crs) was significantly decreased in WT, ERα KO, and ERβ KO mice (males and females) challenged with MA compared to PBS. Interestingly, ERβ KO mice showed declined lung function compared to ERα KO and WT mice at baseline. MA induced AHR, remodeling and immune-cell infiltration was more prominent in females compared to males across all populations, while ERβ KO females showed maximum AHR and DLC, except for neutrophil count. Histology using H&E suggests increased smooth muscle mass in airways with recruitment of inflammatory cells, while SRFG staining showed increased collagen deposition in MA challenged ERβ KO mice compared to ERα KO and WT mice (males and females), with pronounced effects in ERβ KO females. Furthermore, IF studies showed increased expression of α-SMA, fibronectin and vimentin in MA challenged populations compared to PBS, with prominent changes in ERβ KO females. This novel study indicates ERβ plays a pivotal role in airway remodeling and AHR and understanding the mechanisms involved might help to surface it out as a potential target to treat asthma.
Collapse
Affiliation(s)
| | | | | | | | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
36
|
Tovar A, Smith GJ, Thomas JM, Crouse WL, Harkema JR, Kelada SNP. Transcriptional Profiling of the Murine Airway Response to Acute Ozone Exposure. Toxicol Sci 2020; 173:114-130. [PMID: 31626304 PMCID: PMC6944221 DOI: 10.1093/toxsci/kfz219] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Ambient ozone (O3) exposure has serious consequences on respiratory health, including airway inflammation and injury. Decades of research have yielded thorough descriptions of these outcomes; however, less is known about the molecular processes that drive them. The aim of this study was to further describe the cellular and molecular responses to O3 exposure in murine airways, with a particular focus on transcriptional responses in 2 critical pulmonary tissue compartments: conducting airways (CA) and airway macrophages (AM). After exposing adult, female C57BL/6J mice to filtered air, 1 or 2 ppm O3, we assessed hallmark responses including airway inflammation (cell counts and cytokine secretion) and injury (epithelial permeability), followed by gene expression profiling of CA and AM by RNA-seq. As expected, we observed concentration-dependent increases in airway inflammation and injury. Conducting airways and AM both exhibited changes in gene expression to both 1 and 2 ppm O3 that were largely compartment-specific. In CA, genes associated with epithelial barrier function, detoxification processes, and cellular proliferation were altered, while O3 affected genes involved in innate immune signaling, cytokine production, and extracellular matrix remodeling in AM. Further, CA and AM also exhibited notable differences in concentration-response expression patterns for large numbers of genes. Overall, our study has described transcriptional responses to acute O3 exposure, revealing both shared and unique gene expression patterns across multiple concentrations of O3 and in 2 important O3-responsive tissues. These profiles provide broad mechanistic insight into pulmonary O3 toxicity, and reveal a variety of targets for focused follow-up studies.
Collapse
Affiliation(s)
- Adelaide Tovar
- Department of Genetics
- Curriculum in Genetics & Molecular Biology
| | - Gregory J Smith
- Department of Genetics
- Curriculum in Toxicology & Environmental Medicine
| | | | - Wesley L Crouse
- Department of Genetics
- Curriculum in Bioinformatics & Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Jack R Harkema
- Department of Pathology & Diagnostic Investigation and Institute for Integrated Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Samir N P Kelada
- Department of Genetics
- Curriculum in Genetics & Molecular Biology
- Curriculum in Toxicology & Environmental Medicine
- Curriculum in Bioinformatics & Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
37
|
Ray JL, Fletcher P, Burmeister R, Holian A. The role of sex in particle-induced inflammation and injury. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1589. [PMID: 31566915 DOI: 10.1002/wnan.1589] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/19/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022]
Abstract
The use of engineered nanomaterials within various applications such as medicine, electronics, and cosmetics has been steadily increasing; therefore, the rate of occupational and environmental exposures has also increased. Inhalation is an important route of exposure to nanomaterials and has been shown to cause various respiratory diseases in animal models. Human lung disease frequently presents with a sex/gender-bias in prevalence or severity, but investigation of potential sex-differences in the adverse health outcomes associated with nanoparticle inhalation is greatly lacking. Only ~20% of basic research in the general sciences use both male and female animals and a substantial percentage of these do not address differences between sexes within their analyses. This has prevented researchers from fully understanding the impact of sex-based variables on health and disease, particularly the pathologies resulting from the inhalation of particles. The mechanisms responsible for sex-differences in respiratory disease remain unclear, but could be related to a number of variables including sex-differences in hormone signaling, lung physiology, or respiratory immune function. By incorporating sex-based analysis into respiratory nanotoxicology and utilizing human data from other relevant particles (e.g., asbestos, silica, particulate matter), we can improve our understanding of sex as a biological variable in nanoparticle exposures. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Jessica L Ray
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Paige Fletcher
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Rachel Burmeister
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| | - Andrij Holian
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana
| |
Collapse
|
38
|
Fuentes N, Nicoleau M, Cabello N, Montes D, Zomorodi N, Chroneos ZC, Silveyra P. 17β-Estradiol affects lung function and inflammation following ozone exposure in a sex-specific manner. Am J Physiol Lung Cell Mol Physiol 2019; 317:L702-L716. [PMID: 31553636 DOI: 10.1152/ajplung.00176.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Inflammatory lung diseases affect men and women disproportionately, suggesting that fluctuations of circulating hormone levels mediate inflammatory responses. Studies have shown that ozone exposure contributes to lung injury and impairment of innate immunity with differential effects in men and women. Here, we hypothesized that 17β-estradiol enhances inflammation and airway hyperresponsiveness (AHR), triggered by ozone exposure, in the female lung. We performed gonadectomy and hormone treatment (17β-estradiol, 2 wk) in C57BL/6J female and male mice and exposed animals to 1 ppm of ozone or filtered air for 3 h. Twenty-four hours later, we tested lung function, inflammatory gene expression, and changes in bronchoalveolar lavage fluid (BALF). We found increased AHR and expression of inflammatory genes after ozone exposure. These changes were higher in females and were affected by gonadectomy and 17β-estradiol treatment in a sex-specific manner. Gonadectomized male mice displayed higher AHR and inflammatory gene expression than controls exposed to ozone; 17β-estradiol treatment did not affect this response. In females, ovariectomy reduced ozone-induced AHR, which was restored by 17β-estradiol treatment. Ozone exposure also increased BALF lipocalin-2, which was reduced in both male and female gonadectomized mice. Treatment with 17β-estradiol increased lipocalin-2 levels in females but lowered them in males. Gonadectomy also reduced ozone-induced expression of lung IL-6 and macrophage inflammatory protein-3 in females, which was restored by treatment with 17β-estradiol. Together, these results indicate that 17β-estradiol increases ozone-induced inflammation and AHR in females but not in males. Future studies examining diseases associated with air pollution exposure should consider the patient's sex and hormonal status.
Collapse
Affiliation(s)
- Nathalie Fuentes
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Marvin Nicoleau
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Noe Cabello
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Deborah Montes
- Biobehavioral Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Naseem Zomorodi
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Zissis C Chroneos
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Patricia Silveyra
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.,Biobehavioral Laboratory, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
39
|
Speen AM, Hoffman JR, Kim HYH, Escobar YN, Nipp GE, Rebuli ME, Porter NA, Jaspers I. Small Molecule Antipsychotic Aripiprazole Potentiates Ozone-Induced Inflammation in Airway Epithelium. Chem Res Toxicol 2019; 32:1997-2005. [PMID: 31476115 DOI: 10.1021/acs.chemrestox.9b00149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Inhaled ground level ozone (O3) has well described adverse health effects, which may be augmented in susceptible populations. While conditions, such as pre-existing respiratory disease, have been identified as factors enhancing susceptibility to O3-induced health effects, the potential for chemical interactions in the lung to sensitize populations to pollutant-induced responses has not yet been studied. In the airways, inhaled O3 reacts with lipids, such as cholesterol, to generate reactive and electrophilic oxysterol species, capable of causing cellular dysfunction and inflammation. The enzyme regulating the final step of cholesterol biosynthesis, 7-dehydrocholesterol reductase (DHCR7), converts 7-dehydrocholesterol (7-DHC) to cholesterol. Inhibition of DHCR7 increases the levels of 7-DHC, which is much more susceptible to oxidation than cholesterol. Chemical analysis established the capacity for a variety of small molecule antipsychotic drugs, like Aripiprazole (APZ), to inhibit DHCR7 and elevate circulating 7-DHC. Our results show that APZ and the known DHCR7 inhibitor, AY9944, increase 7-DHC levels in airway epithelial cells and potentiate O3-induced IL-6 and IL-8 expression and cytokine release. Targeted immune-related gene array analysis demonstrates that APZ significantly modified O3-induced expression of 16 genes, causing dysregulation in expression of genes associated with leukocyte recruitment and inflammatory response. Additionally, we find that APZ increases O3-induced IL-6 and IL-8 expression in human nasal epithelial cells from male but not female donors. Overall, the evidence we provide describes a novel molecular mechanism by which chemicals, such as APZ, that perturb cholesterol biosynthesis affect O3-induced biological responses.
Collapse
Affiliation(s)
| | | | - Hye-Young H Kim
- Department of Chemistry and Center for Molecular Toxicology , Vanderbilt University , Nashville , Tennessee 37235 , United States
| | | | | | | | - Ned A Porter
- Department of Chemistry and Center for Molecular Toxicology , Vanderbilt University , Nashville , Tennessee 37235 , United States
| | | |
Collapse
|
40
|
Osgood RS, Kasahara DI, Tashiro H, Cho Y, Shore SA. Androgens augment pulmonary responses to ozone in mice. Physiol Rep 2019; 7:e14214. [PMID: 31544355 PMCID: PMC6755142 DOI: 10.14814/phy2.14214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/28/2022] Open
Abstract
Ozone causes airway hyperresponsiveness, a defining feature of asthma, and is an asthma trigger. In mice, ozone-induced airway hyperresponsiveness is greater in males than in females, suggesting a role for sex hormones in the response to ozone. To examine the role of androgens in these sex differences, we castrated 4-week-old mice. Controls underwent sham surgery. At 8 weeks of age, mice were exposed to ozone (2ppm, 3 h) or room air. Twenty-four hours later, mice were anesthetized and measurements of airway responsiveness to inhaled aerosolized methacholine were made. Mice were then euthanized and bronchoalveolar lavage was performed. Castration attenuated ozone-induced airway hyperresponsiveness and reduced bronchoalveolar lavage cells. In intact males, flutamide, an androgen receptor inhibitor, had similar effects to castration. Bronchoalveolar lavage concentrations of several cytokines were reduced by either castration or flutamide treatment, but only IL-1α was reduced by both castration and flutamide. Furthermore, an anti-IL-1α antibody reduced bronchoalveolar lavage neutrophils in intact males, although it did not alter ozone-induced airway hyperresponsiveness. Our data indicate that androgens augment pulmonary responses to ozone and that IL-1α may contribute to the effects of androgens on ozone-induced cellular inflammation but not airway hyperresponsiveness.
Collapse
Affiliation(s)
- Ross S. Osgood
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusetts
| | - David I. Kasahara
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusetts
| | - Hiroki Tashiro
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusetts
| | - Youngji Cho
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusetts
| | - Stephanie A. Shore
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusetts
| |
Collapse
|
41
|
Yang J, Chen Y, Jiang K, Yang Y, Zhao G, Guo S, Deng G. MicroRNA-106a Provides Negative Feedback Regulation in Lipopolysaccharide-Induced Inflammation by targeting TLR4. Int J Biol Sci 2019; 15:2308-2319. [PMID: 31595149 PMCID: PMC6775322 DOI: 10.7150/ijbs.33432] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/03/2019] [Indexed: 12/13/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical disease with high incidence and mortality rate, which is characterized by severe inflammatory response and tissues damage. MicroRNAs (miRNAs) have been regarded as novel regulators of inflammation, and play an important role in various inflammatory diseases. However, it remains unknown whether the regulatory mechanisms mediated by miR-106a is involved in LPS-induced ALI. In this study, we found that expression of miR-106a was significantly decreased in lung tissues of ALI mice and LPS-stimulated macrophages. We also revealed that over-expression of miR-106a significantly decreased the production of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α, whereas this effect was reversed by the inhibition of miR-106a. Moreover, miR-106a inhibits NF-κB activation by targeting TLR4 expression. We further demonstrated that miR-106a inhibited TLR4 expression via binding directly to the 3'-UTR of TLR4. Taken together, the results of the present study illuminated that miR-106a is a negative feedback regulator in LPS-stimulated inflammation through TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| |
Collapse
|
42
|
Thorenoor N, Kawasawa YI, Gandhi CK, Zhang X, Floros J. Differential Impact of Co-expressed SP-A1/SP-A2 Protein on AM miRNome; Sex Differences. Front Immunol 2019; 10:1960. [PMID: 31475015 PMCID: PMC6707024 DOI: 10.3389/fimmu.2019.01960] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/02/2019] [Indexed: 12/04/2022] Open
Abstract
In humans there are two surfactant protein A (SP-A) functional genes SFTPA1 and SFTPA2 encoding innate immune molecules, SP-A1 and SP-A2, respectively, with numerous genetic variants each. SP-A interacts and regulates many of the functions of alveolar macrophages (AM). It is shown that SP-A variants differ in their ability to regulate the AM miRNome in response to oxidative stress (OxS). Because humans have both SP-A gene products, we were interested to determine the combined effect of co-expressed SP-A1/SP-A2 (co-ex) in response to ozone (O3) induced OxS on AM miRNome. Human transgenic (hTG) mice, carrying both SP-A1/SP-A2 (6A2/1A0, co-ex) and SP-A- KO were utilized. The hTG and KO mice were exposed to filtered air (FA) or O3 and miRNA levels were measured after AM isolation with or without normalization to KO. We found: (i) The AM miRNome of co-ex males and females in response to OxS to be largely downregulated after normalization to KO, but after Bonferroni multiple comparison analysis only in females the AM miRNome remained significantly different compared to control (FA); (ii) The targets of the significantly changed miRNAs were downregulated in females and upregulated in males; (iii) Several of the validated mRNA targets were involved in pro-inflammatory response, anti-apoptosis, cell cycle, cellular growth and proliferation; (iv) The AM of SP-A2 male, shown, previously to have major effect on the male AM miRNome in response to OxS, shared similarities with the co-ex, namely in pathways involved in the pro-inflammatory response and anti-apoptosis but also exhibited differences with the cell-cycle, growth, and proliferation pathway being involved in co-ex and ROS homeostasis in SP-A2 male. We speculate that the presence of both gene products vs. single gene products differentially impact the AM responses in males and females in response to OxS.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Departments of Pharmacology and Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chintan K Gandhi
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xuesheng Zhang
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
43
|
Mumby S, Chung KF, Adcock IM. Transcriptional Effects of Ozone and Impact on Airway Inflammation. Front Immunol 2019; 10:1610. [PMID: 31354743 PMCID: PMC6635463 DOI: 10.3389/fimmu.2019.01610] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/27/2019] [Indexed: 12/24/2022] Open
Abstract
Epidemiological and challenge studies in healthy subjects and in individuals with asthma highlight the health impact of environmental ozone even at levels considered safe. Acute ozone exposure in man results in sputum neutrophilia in 30% of subjects particularly young children, females, and those with ongoing cardiopulmonary disease. This may be associated with systemic inflammation although not in all cases. Chronic exposure amplifies these effects and can result in the formation of asthma-like symptoms and immunopathology. Asthmatic patients who respond to ozone (responders) induce a greater number of genes in bronchoalveolar (BAL) macrophages than healthy responders with up-regulation of inflammatory and immune pathways under the control of cytokines and chemokines and the enhanced expression of remodeling and repair programmes including those associated with protease imbalances and cell-cell adhesion. These pathways are under the control of several key transcription regulatory factors including nuclear factor (NF)-κB, anti-oxidant factors such as nuclear factor (erythroid-derived 2)-like 2 NRF2, the p38 mitogen activated protein kinase (MAPK), and priming of the immune system by up-regulating toll-like receptor (TLR) expression. Murine and cellular models of acute and chronic ozone exposure recapitulate the inflammatory effects seen in humans and enable the elucidation of key transcriptional pathways. These studies emphasize the importance of distinct transcriptional networks in driving the detrimental effects of ozone. Studies indicate the critical role of mediators including IL-1, IL-17, and IL-33 in driving ozone effects on airway inflammation, remodeling and hyperresponsiveness. Transcription analysis and proof of mechanisms studies will enable the development of drugs to ameliorate the effects of ozone exposure in susceptible individuals.
Collapse
Affiliation(s)
- Sharon Mumby
- Respiratory Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kian Fan Chung
- Respiratory Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- Respiratory Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
44
|
Kobets T, Čepičková M, Volkova V, Sohrabi Y, Havelková H, Svobodová M, Demant P, Lipoldová M. Novel Loci Controlling Parasite Load in Organs of Mice Infected With Leishmania major, Their Interactions and Sex Influence. Front Immunol 2019; 10:1083. [PMID: 31231359 PMCID: PMC6566641 DOI: 10.3389/fimmu.2019.01083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/29/2019] [Indexed: 12/24/2022] Open
Abstract
Leishmaniasis is a serious health problem in many countries, and continues expanding to new geographic areas including Europe and USA. This disease, caused by parasites of Leishmania spp. and transmitted by phlebotomine sand flies, causes up to 1.3 million new cases each year and despite efforts toward its functional dissection and treatment it causes 20-50 thousands deaths annually. Dependence of susceptibility to leishmaniasis on sex and host's genes was observed in humans and in mouse models. Several laboratories defined in mice a number of Lmr (Leishmania major response) genetic loci that control functional and pathological components of the response to and outcome of L. major infection. However, the development of its most aggressive form, visceral leishmaniasis, which is lethal if untreated, is not yet understood. Visceral leishmaniasis is caused by infection and inflammation of internal organs. Therefore, we analyzed the genetics of parasite load, spread to internal organs, and ensuing visceral pathology. Using a new PCR-based method of quantification of parasites in tissues we describe a network-like set of interacting genetic loci that control parasite load in different organs. Quantification of Leishmania parasites in lymph nodes, spleen and liver from infected F2 hybrids between BALB/c and recombinant congenic strains CcS-9 and CcS-16 allowed us to map two novel parasite load controlling Leishmania major response loci, Lmr24 and Lmr27. We also detected parasite-controlling role of the previously described loci Lmr4, Lmr11, Lmr13, Lmr14, Lmr15, and Lmr25, and describe 8 genetic interactions between them. Lmr14, Lmr15, Lmr25, and Lmr27 controlled parasite load in liver and lymph nodes. In addition, Leishmania burden in lymph nodes but not liver was influenced by Lmr4 and Lmr24. In spleen, parasite load was controlled by Lmr11 and Lmr13. We detected a strong effect of sex on some of these genes. We also mapped additional genes controlling splenomegaly and hepatomegaly. This resulted in a systematized insight into genetic control of spread and load of Leishmania parasites and visceral pathology in the mammalian organism.
Collapse
Affiliation(s)
- Tatyana Kobets
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Marie Čepičková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Valeriya Volkova
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Yahya Sohrabi
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Helena Havelková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czechia
| | | | - Peter Demant
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czechia
| |
Collapse
|
45
|
Birukova A, Cyphert-Daly J, Cumming RI, Yu YR, Gowdy KM, Que LG, Tighe RM. Sex Modifies Acute Ozone-Mediated Airway Physiologic Responses. Toxicol Sci 2019; 169:499-510. [PMID: 30825310 PMCID: PMC6542336 DOI: 10.1093/toxsci/kfz056] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sex differences clearly exist in incidence, susceptibility, and severity of airway disease and in pulmonary responses to air pollutants such as ozone (O3). Prior rodent O3 exposure studies demonstrate sex-related differences in the expression of lung inflammatory mediators and signaling. However, whether or not sex modifies O3-induced airway physiologic responses remains less explored. To address this, we exposed 8- to 10-week-old male and female C57BL/6 mice to either 1 or 2 ppm O3 or filtered air (FA) for 3 h. At 12, 24, 48, and 72 h following exposure, we assessed airway hyperresponsiveness to methacholine (MCh), bronchoalveolar lavage fluid cellularity, cytokines and total protein/albumin, serum progesterone, and whole lung immune cells by flow cytometry. Male mice generated consistent airway hyperresponsiveness to MCh at all time points following exposure. Alternatively, females had less consistent airway physiologic responses to MCh, which were more variable between individual experiments and did not correlate with serum progesterone levels. Bronchoalveolar lavage fluid total cells peaked at 12 h and were persistently elevated through 72 h. At 48 h, bronchoalveolar lavage cells were greater in females versus males. Bronchoalveolar lavage fluid cytokines and total protein/albumin increased following O3 exposure without sex differences. Flow cytometry of whole lung tissue identified dynamic O3-induced immune cell changes also independent of sex. Our results indicate sex differences in acute O3-induced airway physiology responses and airspace influx without significant difference in other injury and inflammation measures. This study highlights the importance of considering sex as a biological variable in acute O3-induced airway physiology responses.
Collapse
Affiliation(s)
| | | | | | - Yen-Rei Yu
- Department of Medicine, Duke University, Durham, North Carolina 27710
| | - Kymberly M Gowdy
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, North Carolina 27858
| | - Loretta G Que
- Department of Medicine, Duke University, Durham, North Carolina 27710
| | - Robert M Tighe
- Department of Medicine, Duke University, Durham, North Carolina 27710
| |
Collapse
|
46
|
Fesen K, Silveyra P, Fuentes N, Nicoleau M, Rivera L, Kitch D, Graff GR, Siddaiah R. The role of microRNAs in chronic pseudomonas lung infection in Cystic fibrosis. Respir Med 2019; 151:133-138. [PMID: 31047110 DOI: 10.1016/j.rmed.2019.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Cystic Fibrosis (CF) is the most common life limiting genetic disorder, characterized by chronic respiratory failure secondary to inflammation and chronic bacterial lung infection. Pseudomonas aeruginosa lung infection is associated with more severe lung disease and rapid progression of respiratory failure when compared to Staphylococcus aureus infection. We hypothesized that a specific signature of epigenetic factors targeting specific gene transcripts contributes to the increased morbidity seen in CF patients with chronic Pseudomonas infection. METHODS We collected exhaled breath condensate (EBC) from 27 subjects and evaluated miRNA signatures in these samples using commercial PCR array. We identified predicted mRNA targets and associated signaling pathways using Ingenuity Pathway Analysis. RESULTS We found 11 differentially expressed miRNAs in EBC of patients infected with Pseudomonas aeruginosa compared to EBC from CF patients who were not chronically infected with Pseudomonas aeruginosa (p < 0.05). Six of these miRNAs (hsa-miRNA-1247, hsa-miRNA-1276, hsa-miRNA-449c, hsa-miRNA-3170, hsa-miRNA-432-5p and hsa-miR-548) were significantly different in the CF Pseudomonas positive group when compared to both the CF Pseudomonas negative group and healthy control group. Ingenuity pathway analysis (IPA) revealed organismal injury and abnormalities, reproductive system disease and cancer as the top diseases and bio functions associated with these miRNAs. IPA also detected RELA, JUN, TNF, IL-10, CTNNB1, IL-13, SERPINB8, CALM1, STARD3NL, SFI1, CD55, RPS6KA4, TTC36 and HIST1H3D as the top target genes for these miRNAs. CONCLUSION Our study identified 6 miRNAs as epigenetic factors specifically associated with chronic Pseudomonas infection in patients with CF.
Collapse
Affiliation(s)
- Katherine Fesen
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Patricia Silveyra
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Biobehavioral Laboratory, The University of North Carolina at Chapel Hill, School of Nursing, Chapel Hill, NC, 27599, USA
| | - Nathalie Fuentes
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Marvin Nicoleau
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Lidys Rivera
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Diane Kitch
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Gavin R Graff
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Roopa Siddaiah
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
47
|
Fuentes N, Cabello N, Nicoleau M, Chroneos ZC, Silveyra P. Modulation of the lung inflammatory response to ozone by the estrous cycle. Physiol Rep 2019; 7:e14026. [PMID: 30848106 PMCID: PMC6405886 DOI: 10.14814/phy2.14026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence suggests that sex differences exist in the control of lung innate immunity; however, the specific roles of sex hormones in the inflammatory response, and the mechanisms involved are unclear. Here, we investigated whether fluctuations in circulating hormone levels occurring in the mouse estrous cycle could affect the inflammatory response to air pollution exposure. For this, we exposed female mice (C57BL/6J, 8 weeks old) at different phases of the estrous cycle to 2 ppm of ozone or filtered air (FA) for 3 h. Following exposure, we collected lung tissue and bronchoalveolar lavage fluid (BAL), and performed lung function measurements to evaluate inflammatory responses and respiratory mechanics. We found a differential inflammatory response to ozone in females exposed in the luteal phase (metestrus, diestrus) versus the follicular phase (proestrus, estrus). Females exposed to ozone in the follicular phase had significantly higher expression of inflammatory genes, including Ccl2, Cxcl2, Ccl20, and Il6, compared to females exposed in the luteal phase (P < 0.05), and displayed differential activation of regulatory pathways. Exposure to ozone in the follicular phase also resulted in higher BAL neutrophilia, lipocalin levels, and airway resistance than exposure in the luteal phase (P < 0.05). Together, these results show that the effects of ozone exposure in the female lung are affected by the estrous cycle phase, and potentially hormonal status. Future studies investigating air pollution effects and inflammation in women should consider the menstrual cycle phase and/or circulating hormone levels.
Collapse
Affiliation(s)
- Nathalie Fuentes
- Department of PediatricsThe Pennsylvania State University College of MedicineHersheyPennsylvania
| | - Noe Cabello
- Department of PediatricsThe Pennsylvania State University College of MedicineHersheyPennsylvania
| | - Marvin Nicoleau
- Department of PediatricsThe Pennsylvania State University College of MedicineHersheyPennsylvania
| | - Zissis C. Chroneos
- Department of PediatricsThe Pennsylvania State University College of MedicineHersheyPennsylvania
| | - Patricia Silveyra
- Department of PediatricsThe Pennsylvania State University College of MedicineHersheyPennsylvania
- Biobehavioral LaboratoryThe University of North Carolina at Chapel HillChapel HillNorth Carolina
| |
Collapse
|
48
|
Fuentes N, Silveyra P. Lung microRNA Profiling Across the Estrous Cycle in Ozone-exposed Mice. J Vis Exp 2019. [PMID: 30663642 DOI: 10.3791/58664] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MicroRNA (miRNA) profiling has become of interest to researchers working in various research areas of biology and medicine. Current studies show a promising future of using miRNAs in the diagnosis and care of lung diseases. Here, we define a protocol for miRNA profiling to measure the relative abundance of a group of miRNAs predicted to regulate inflammatory genes in the lung tissue from of an ozone-induced airway inflammation mouse model. Because it has been shown that circulating sex hormone levels can affect the regulation of lung innate immunity in females, the purpose of this method is to describe an inflammatory miRNA profiling protocol in female mice, taking into consideration the estrous cycle stage of each animal at the time of ozone exposure. We also address applicable bioinformatics approaches to miRNA discovery and target identification methods using limma, an R/Bioconductor software, and functional analysis software to understand the biological context and pathways associated with differential miRNA expression.
Collapse
Affiliation(s)
- Nathalie Fuentes
- Pulmonary, Immunology and Physiology Laboratory, Department of Pediatrics, Pennsylvania State University College of Medicine
| | - Patricia Silveyra
- Pulmonary, Immunology and Physiology Laboratory, Department of Pediatrics, Pennsylvania State University College of Medicine; Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine;
| |
Collapse
|
49
|
Fuentes N, Silveyra P. Endocrine regulation of lung disease and inflammation. Exp Biol Med (Maywood) 2018; 243:1313-1322. [PMID: 30509139 DOI: 10.1177/1535370218816653] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
IMPACT STATEMENT Sex-differences in the incidence and severity of inflammatory lung diseases have been recognized for years. Women of reproductive age are more likely to suffer from chronic lung disease, with higher mortality rates than men. Physiological changes in hormone levels such as those occurring during the menstrual cycle, pregnancy, and menopause have been associated with lung function changes and asthma symptoms. Despite this, the roles of sex hormones in the mechanisms associated with lung diseases have not been fully elucidated. This review summarizes basic and clinical studies of sex hormones as potential modulators of lung function and inflammation. The information obtained from sex-specific research on lung physiology and pathology will potentially help in the development of sex-specific therapeutics for inflammatory lung disease that may account for the hormonal status of the patient.
Collapse
Affiliation(s)
- Nathalie Fuentes
- 1 Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Patricia Silveyra
- 1 Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.,2 Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
50
|
Lang CH. Lack of sexual dimorphism on the inhibitory effect of alcohol on muscle protein synthesis in rats under basal conditions and after anabolic stimulation. Physiol Rep 2018; 6:e13929. [PMID: 30512248 PMCID: PMC6278815 DOI: 10.14814/phy2.13929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 11/24/2022] Open
Abstract
Previous studies indicate women have a higher blood alcohol (i.e., ethanol) and acetaldehyde concentration after consuming an equivalent amount of alcohol, and that women are more susceptible to the long-term negative health effects of alcohol. However, there is a paucity of data pertaining to whether there is a sexual dimorphic response in skeletal muscle to alcohol. Adult male and female Sprague-Dawley rats were used and the primary endpoint was in vivo determined muscle (gastrocnemius) protein synthesis (MPS). The initial study indicated MPS did not differ in female rats during proestrus, estrus, metestrus, or diestrus; hence, subsequent studies used female rats irrespective of estrus cycle phase. There was no difference in MPS between male and female rats under basal fasted conditions, and the time- and dose-responsiveness of both groups to the inhibitory effect of acute alcohol did not differ. The ability of alcohol to suppress MPS was comparable in male and female rats pretreated with alcohol dehydrogenase inhibitor 4-methylpyrazol. Chronic alcohol feeding for 6 weeks decreased MPS in male but not in female rats; however, MPS was reduced in both sexes at 14 weeks. Finally, oral gavage of leucine increased MPS similarly in male and female rats and chronic alcohol feeding for 14 weeks prevented the anabolic effect in both sexes. These data suggest normal fluctuations in ovarian hormones do not significantly alter MPS in female rats, and that there is no sexual dimorphic response to the effects of acute alcohol intoxication on MPS. While chronic alcohol consumption appeared to decrease MPS at an early time point in male compared to female rats, there was no sex difference in the suppressive effect of alcohol at a later time point. Overall, these data do not support the prevailing belief that females are more susceptible than males to alcohol's catabolic effect on MPS.
Collapse
Affiliation(s)
- Charles H. Lang
- Department of Cellular and Molecular PhysiologyPenn State College of MedicineHersheyPennsylvania
| |
Collapse
|