1
|
Monedero Cobeta I, Gomez Bris R, Rodríguez-Rodríguez P, Saez A, Quintana-Villamandos B, González Granado JM, Arribas SM. Fetal programming and lactation: modulating gene expression in response to undernutrition during intrauterine life. Pediatr Res 2024; 95:1764-1774. [PMID: 38326476 DOI: 10.1038/s41390-024-03042-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Adverse environmental conditions during intrauterine life, known as fetal programming, significantly contribute to the development of diseases in adulthood. Fetal programming induced by factors like maternal undernutrition leads to low birth weight and increases the risk of cardiometabolic diseases. METHODS We studied a rat model of maternal undernutrition during gestation (MUN) to investigate gene expression changes in cardiac tissue using RNA-sequencing of day 0-1 litters. Moreover, we analyzed the impact of lactation at day 21, in MUN model and cross-fostering experiments, on cardiac structure and function assessed by transthoracic echocardiography, and gene expression changes though qPCR. RESULTS Our analysis identified specific genes with altered expression in MUN rats at birth. Two of them, Agt and Pparg, stand out for being associated with cardiac hypertrophy and fibrosis. At the end of the lactation period, MUN males showed increased expression of Agt and decreased expression of Pparg, correlating with cardiac hypertrophy. Cross-fostering experiments revealed that lactation with control breastmilk mitigated these expression changes reducing cardiac hypertrophy in MUN males. CONCLUSIONS Our findings highlight the interplay between fetal programming, gene expression, and cardiac hypertrophy suggesting that lactation period is a potential intervention window to mitigate the effects of fetal programming. IMPACT Heart remodeling involves the alteration of several groups of genes and lactation period plays a key role in establishing gene expression modification caused by fetal programming. We could identify expression changes of relevant genes in cardiac tissue induced by undernutrition during fetal life. We expose the contribution of the lactation period in modulating the expression of Agt and Pparg, relevant genes associated with cardiac hypertrophy. This evidence reveal lactation as a crucial intervention window for preventing or countering fetal programming.
Collapse
Affiliation(s)
- Ignacio Monedero Cobeta
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Raquel Gomez Bris
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | | | - Angela Saez
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), 28223, Pozuelo de Alarcón, Spain
| | - Begoña Quintana-Villamandos
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Jose Maria González Granado
- LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041, Madrid, Spain.
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain.
| | - Silvia Magdalena Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain.
| |
Collapse
|
2
|
Liu L, Tang Z, Zeng Q, Qi W, Zhou Z, Chen D, Cai D, Chen Y, Sun S, Gong S, He B, Yu S, Zhao L. Transcriptomic Insights into Different Stimulation Intensity of Electroacupuncture in Treating COPD in Rat Models. J Inflamm Res 2024; 17:2873-2887. [PMID: 38741612 PMCID: PMC11090121 DOI: 10.2147/jir.s458580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
Background Electroacupuncture (EA), with varying stimulation intensities, has demonstrated therapeutic potentials in both animal and clinical studies for the treatment of chronic obstructive pulmonary disease (COPD). However, a comprehensive investigation of the intensity-related effects, particularly 1mA and 3mA of EA, and the underlying mechanisms remains lacking. Methods A COPD rat model was established by prolonged exposure to cigarette smoke and intermittent intratracheal instillation of lipopolysaccharide. EA treatment was administered at acupoints BL13 (Feishu) and ST36 (Zusanli), 20 minutes daily for 2 weeks, with intensities of 1mA and 3mA. EA effectiveness was evaluated by pulmonary function, histopathological change, serum level of inflammatory cytokines, and level of oxidative stress markers in serum and lung tissues. Transcriptome profiling and weighted gene co-expression network analysis (WGCNA) were performed to reveal gene expression patterns and identify hub genes. Real-time quantitative PCR (RT-qPCR) and Western blot (WB) were performed to detect the mRNA and protein expression levels, respectively. Results EA at both 1mA and 3mA exerted differing therapeutic effects by improving lung function and reducing inflammation and oxidative stress in COPD rats. Transcriptome analysis revealed distinct expression patterns between the two groups, functionally corresponding to shared and intensity-specific (1mA and 3mA) enriched pathways. Eight candidate genes were identified, including Aqp9, Trem1, Mrc1, and Gpnmb that were downregulated by EA and upregulated in COPD. Notably, Msr1 and Slc26a4 exclusively downregulated in EA-1mA, while Pde3a and Bmp6 upregulated solely in EA-3mA. WGCNA constructed 5 key modules and elucidated the module-trait relationship, with the aforementioned 8 genes being highlighted. Additionally, their mRNA and protein levels were validated by RT-qPCR and WB. Conclusion Our results demonstrated that 1mA and 3mA intensities induce distinct gene expression patterns at the transcriptional level, associated with shared and 1mA vs 3mA-specific enriched pathways. Genes Mrc1, Gpnmb, Trem1, and Aqp9 emerge as promising targets, and further studies are needed to elucidate their functional consequences in COPD.
Collapse
Affiliation(s)
- Lu Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Zili Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Qian Zeng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Wenchuan Qi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Ziyang Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Daohong Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Dingjun Cai
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, Sichuan, People’s Republic of China
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of Traditional Chinese Medicine), Ministry of Education, Chengdu City, Sichuan Province, China
| | - Ying Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Shiqi Sun
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Siyao Gong
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Bin He
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
| | - Shuguang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, Sichuan, People’s Republic of China
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of Traditional Chinese Medicine), Ministry of Education, Chengdu City, Sichuan Province, China
| | - Ling Zhao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu City, Sichuan Province, People’s Republic of China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, Sichuan, People’s Republic of China
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of Traditional Chinese Medicine), Ministry of Education, Chengdu City, Sichuan Province, China
| |
Collapse
|
3
|
Ozekin YH, Saal ML, Pineda RH, Moehn K, Ordonez-Erives MA, Delgado Figueroa MF, Frazier C, Korth KM, Königshoff M, Bates EA, Vladar EK. Intrauterine exposure to nicotine through maternal vaping disrupts embryonic lung and skeletal development via the Kcnj2 potassium channel. Dev Biol 2023; 501:111-123. [PMID: 37353105 PMCID: PMC10445547 DOI: 10.1016/j.ydbio.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023]
Abstract
Smoking cigarettes during pregnancy is associated with adverse effects on infants including low birth weight, defective lung development, and skeletal abnormalities. Pregnant women are increasingly turning to vaping [use of electronic (e)-cigarettes] as a perceived safer alternative to cigarettes. However, nicotine disrupts fetal development, suggesting that like cigarette smoking, nicotine vaping may be detrimental to the fetus. To test the impact of maternal vaping on fetal lung and skeletal development in mice, pregnant dams were exposed to e-cigarette vapor throughout gestation. At embryonic day (E)18.5, vape exposed litter sizes were reduced, and some embryos exhibited growth restriction compared to air exposed controls. Fetal lungs were collected for histology and whole transcriptome sequencing. Maternally nicotine vaped embryos exhibited histological and transcriptional changes consistent with impaired distal lung development. Embryonic lung gene expression changes mimicked transcriptional changes observed in adult mouse lungs exposed to cigarette smoke, suggesting that the developmental defects may be due to direct nicotine exposure. Fetal skeletons were analyzed for craniofacial and long bone lengths. Nicotine directly binds and inhibits the Kcnj2 potassium channel which is important for bone development. The length of the maxilla, palatal shelves, humerus, and femur were reduced in vaped embryos, which was further exacerbated by loss of one copy of the Kcnj2 gene. Nicotine vapor exposed Kcnj2KO/+ embryos also had significantly lower birth weights than unexposed animals of either genotype. Kcnj2 mutants had severely defective lungs with and without vape exposure, suggesting that potassium channels may be broadly involved in mediating the detrimental developmental effects of nicotine vaping. These data indicate that intrauterine nicotine exposure disrupts fetal lung and skeletal development likely through inhibition of Kcnj2.
Collapse
Affiliation(s)
- Yunus H Ozekin
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Maxwell L Saal
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ricardo H Pineda
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kayla Moehn
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Madison A Ordonez-Erives
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Maria F Delgado Figueroa
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Caleb Frazier
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kamryn M Korth
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Melanie Königshoff
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily A Bates
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Eszter K Vladar
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
4
|
Solorio-Rodriguez SA, Williams A, Poulsen SS, Knudsen KB, Jensen KA, Clausen PA, Danielsen PH, Wallin H, Vogel U, Halappanavar S. Single-Walled vs. Multi-Walled Carbon Nanotubes: Influence of Physico-Chemical Properties on Toxicogenomics Responses in Mouse Lungs. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13061059. [PMID: 36985953 PMCID: PMC10057402 DOI: 10.3390/nano13061059] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 05/27/2023]
Abstract
Single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs) are nanomaterials with one or multiple layers of carbon sheets. While it is suggested that various properties influence their toxicity, the specific mechanisms are not completely known. This study was aimed to determine if single or multi-walled structures and surface functionalization influence pulmonary toxicity and to identify the underlying mechanisms of toxicity. Female C57BL/6J BomTac mice were exposed to a single dose of 6, 18, or 54 μg/mouse of twelve SWCNTs or MWCNTs of different properties. Neutrophil influx and DNA damage were assessed on days 1 and 28 post-exposure. Genome microarrays and various bioinformatics and statistical methods were used to identify the biological processes, pathways and functions altered post-exposure to CNTs. All CNTs were ranked for their potency to induce transcriptional perturbation using benchmark dose modelling. All CNTs induced tissue inflammation. MWCNTs were more genotoxic than SWCNTs. Transcriptomics analysis showed similar responses across CNTs at the pathway level at the high dose, which included the perturbation of inflammatory, cellular stress, metabolism, and DNA damage responses. Of all CNTs, one pristine SWCNT was found to be the most potent and potentially fibrogenic, so it should be prioritized for further toxicity testing.
Collapse
Affiliation(s)
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A0K9, Canada; (S.A.S.-R.); (A.W.)
| | - Sarah Søs Poulsen
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark; (S.S.P.); (K.B.K.); (K.A.J.); (P.A.C.); (P.H.D.); (H.W.); (U.V.)
| | - Kristina Bram Knudsen
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark; (S.S.P.); (K.B.K.); (K.A.J.); (P.A.C.); (P.H.D.); (H.W.); (U.V.)
| | - Keld Alstrup Jensen
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark; (S.S.P.); (K.B.K.); (K.A.J.); (P.A.C.); (P.H.D.); (H.W.); (U.V.)
| | - Per Axel Clausen
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark; (S.S.P.); (K.B.K.); (K.A.J.); (P.A.C.); (P.H.D.); (H.W.); (U.V.)
| | - Pernille Høgh Danielsen
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark; (S.S.P.); (K.B.K.); (K.A.J.); (P.A.C.); (P.H.D.); (H.W.); (U.V.)
| | - Håkan Wallin
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark; (S.S.P.); (K.B.K.); (K.A.J.); (P.A.C.); (P.H.D.); (H.W.); (U.V.)
- Department of Public Health, University of Copenhagen, 1353 Copenhagen, Denmark
- National Institute of Occupational Health, 0304 Oslo, Norway
| | - Ulla Vogel
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark; (S.S.P.); (K.B.K.); (K.A.J.); (P.A.C.); (P.H.D.); (H.W.); (U.V.)
| | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A0K9, Canada; (S.A.S.-R.); (A.W.)
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
5
|
Impact of sub-acute acrolein inhalation on the molecular regulation of mitochondrial metabolism in rat lung. Toxicol Lett 2023; 378:19-30. [PMID: 36806656 DOI: 10.1016/j.toxlet.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Nowadays, mitochondria are recognized as key players in the pathogenesis of a variety of smoking-associated lung diseases. Acrolein, a component of cigarette smoke, is a known driver of biological mechanisms underlying smoking-induced respiratory toxicity. The impact of sub-acute acrolein inhalation in vivo on key processes controlling mitochondrial homeostasis in cells of the airways however is unknown. In this study, we investigated the activity/abundance of a myriad of molecules critically involved in mitochondrial metabolic pathways and mitochondrial quality control processes (mitochondrial biogenesis and mitophagy) in the lungs of Sprague-Dawley rats that were sub-acutely exposed to filtered air or 3 ppm acrolein by whole-body inhalation (5 h/day, 5 days/week for 4 weeks). Acrolein exposure induced a general inflammatory response in the lung as gene expression analysis revealed an increased expression of Icam1 and Cinc1 (p < 0.1; p < 0.05). Acrolein significantly decreased enzyme activity of hydroxyacyl-Coenzyme A dehydrogenase (p < 0.01), and decreased Pdk4 transcript levels (p < 0.05), suggestive of acrolein-induced changes in metabolic processes. Investigation of constituents of the mitochondrial biogenesis pathways and mitophagy machinery revealed no pronounced alterations. In conclusion, sub-acute inhalation of acrolein did not affect the regulation of mitochondrial metabolism and quality control, which is in contrast to more profound changes after acute exposure in other studies.
Collapse
|
6
|
Liu K, Salvati A, Sabirsh A. Physiology, pathology and the biomolecular corona: the confounding factors in nanomedicine design. NANOSCALE 2022; 14:2136-2154. [PMID: 35103268 DOI: 10.1039/d1nr08101b] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The biomolecular corona that forms on nanomedicines in different physiological and pathological environments confers a new biological identity. How the recipient biological system's state can potentially affect nanomedicine corona formation, and how this can be modulated, remains obscure. With this perspective, this review summarizes the current knowledge about the content of biological fluids in various compartments and how they can be affected by pathological states, thus impacting biomolecular corona formation. The content of representative biological fluids is explored, and the urgency of integrating corona formation, as an essential component of nanomedicine designs for effective cargo delivery, is highlighted.
Collapse
Affiliation(s)
- Kai Liu
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Anna Salvati
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Groningen 9713AV, The Netherlands
| | - Alan Sabirsh
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
7
|
Zhang H, Liu C, Li L, Feng X, Wang Q, Li J, Xu S, Wang S, Yang Q, Shen Z, Su J, Su X, Sun R, Zhou X, Wang J, Zhou Y, Jiao B, Ding W, Cao X, Wang Y, Huang Y, Ye L. Genomic evidence of lung carcinogenesis associated with coal smoke in Xuanwei area, China. Natl Sci Rev 2021; 8:nwab152. [PMID: 34987843 PMCID: PMC8692936 DOI: 10.1093/nsr/nwab152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/05/2021] [Accepted: 08/13/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Honglei Zhang
- Center for Scientific Research, Yunnan University of Chinese Medicine, China
| | - Chao Liu
- Department of Nuclear Medicine, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| | - Li Li
- Biotherapy Center, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| | - Xu Feng
- Center for Scientific Research, Yunnan University of Chinese Medicine, China
| | - Qing Wang
- Department of Oncology, Qujing First People's Hospital, China
| | - Jihua Li
- Qujing Center for Disease Control and Prevention, China
| | - Shaobin Xu
- Supercomputing Platform of Kunming Institute of Zoology, Kunming Biological Diversity Center of Instruments, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Shuting Wang
- Department of Thoracic Surgery II, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| | - Qianlu Yang
- Department of Thoracic Surgery I, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| | - Zhenghai Shen
- Department of Thoracic Surgery I, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| | - Jinhua Su
- Department of Thoracic Surgery, Xuanwei People's Hospital, China
| | - Xiaosan Su
- Center for Scientific Research, Yunnan University of Chinese Medicine, China
| | - Ruifen Sun
- Center for Scientific Research, Yunnan University of Chinese Medicine, China
| | - Xuhong Zhou
- Center for Scientific Research, Yunnan University of Chinese Medicine, China
| | - Junliang Wang
- Center for Scientific Research, Yunnan University of Chinese Medicine, China
| | - Yongchun Zhou
- Department of Thoracic Surgery I, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| | - Baowei Jiao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Wanbao Ding
- Department of Oncology, Yan'an Affiliated Hospital of Kunming Medical University, China
| | - Xianbao Cao
- Department of Otolaryngology, First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, China
| | - Yue Wang
- The Affiliated Hospital of Guizhou Medical University, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| | - Lianhua Ye
- Department of Thoracic Surgery I, Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, China
| |
Collapse
|
8
|
Novel computational analysis of large transcriptome datasets identifies sets of genes distinguishing chronic obstructive pulmonary disease from healthy lung samples. Sci Rep 2021; 11:10258. [PMID: 33986404 PMCID: PMC8119951 DOI: 10.1038/s41598-021-89762-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/23/2021] [Indexed: 11/08/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) kills over three million people worldwide every year. Despite its high global impact, the knowledge about the underlying molecular mechanisms is still limited. In this study, we aimed to extend the available knowledge by identifying a small set of COPD-associated genes. We analysed different publicly available gene expression datasets containing whole lung tissue (WLT) and airway epithelium (AE) samples from over 400 human subjects for differentially expressed genes (DEGs). We reduced the resulting sets of 436 and 663 DEGs using a novel computational approach that utilises a random depth-first search to identify genes which improve the distinction between COPD patients and controls along the first principle component of the data. Our method identified small sets of 10 and 15 genes in the WLT and AE, respectively. These sets of genes significantly (p < 10–20) distinguish COPD patients from controls with high fidelity. The final sets revealed novel genes like cysteine rich protein 1 (CRIP1) or secretoglobin family 3A member 2 (SCGB3A2) that may underlie fundamental molecular mechanisms of COPD in these tissues.
Collapse
|
9
|
Modeling Cardiovascular Risks of E-Cigarettes With Human-Induced Pluripotent Stem Cell-Derived Endothelial Cells. J Am Coll Cardiol 2020; 73:2722-2737. [PMID: 31146818 DOI: 10.1016/j.jacc.2019.03.476] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Electronic cigarettes (e-cigarettes) have experienced a tremendous increase in use. Unlike cigarette smoking, the effects of e-cigarettes and their constituents on mediating vascular health remain understudied. However, given their increasing popularity, it is imperative to evaluate the health risks of e-cigarettes, including the effects of their ingredients, especially nicotine and flavorings. OBJECTIVES The purpose of this study was to investigate the effects of flavored e-cigarette liquids (e-liquids) and serum isolated from e-cigarette users on endothelial health and endothelial cell-dependent macrophage activation. METHODS Human-induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) and a high-throughput screening approach were used to assess endothelial integrity following exposure to 6 different e-liquids with varying nicotine concentrations and to serum from e-cigarette users. RESULTS The cytotoxicity of the e-liquids varied considerably, with the cinnamon-flavored product being most potent and leading to significantly decreased cell viability, increased reactive oxygen species (ROS) levels, caspase 3/7 activity, and low-density lipoprotein uptake, activation of oxidative stress-related pathway, and impaired tube formation and migration, confirming endothelial dysfunction. Upon exposure of ECs to e-liquid, conditioned media induced macrophage polarization into a pro-inflammatory state, eliciting the production of interleukin-1β and -6, leading to increased ROS. After exposure of human iPSC-ECs to serum of e-cigarette users, increased ROS linked to endothelial dysfunction was observed, as indicated by impaired pro-angiogenic properties. There was also an observed increase in inflammatory cytokine expression in the serum of e-cigarette users. CONCLUSIONS Acute exposure to flavored e-liquids or e-cigarette use exacerbates endothelial dysfunction, which often precedes cardiovascular diseases.
Collapse
|
10
|
Sarlak S, Lalou C, Amoedo ND, Rossignol R. Metabolic reprogramming by tobacco-specific nitrosamines (TSNAs) in cancer. Semin Cell Dev Biol 2020; 98:154-166. [PMID: 31699542 DOI: 10.1016/j.semcdb.2019.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 08/25/2019] [Accepted: 09/02/2019] [Indexed: 12/21/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer and the link between oncogenes activation, tumor supressors inactivation and bioenergetics modulation is well established. However, numerous carcinogenic environmental factors are responsible for early cancer initiation and their impact on metabolic reprogramming just starts to be deciphered. For instance, it was recently shown that UVB irradiation triggers metabolic reprogramming at the pre-cancer stage with implication for skin cancer detection and therapy. These observations foster the need to study the early changes in tissue metabolism following exposure to other carcinogenic events. According to the International Agency for Research on Cancer (IARC), tobacco smoke is a major class I-carcinogenic environmental factor that contains different carcinogens, but little is known on the impact of tobacco smoke on tissue metabolism and its participation to cancer initiation. In particular, tobacco-specific nitrosamines (TSNAs) play a central role in tobacco-smoke mediated cancer initiation. Here we describe the recent advances that have led to a new hypothesis regarding the link between nitrosamines signaling and metabolic reprogramming in cancer.
Collapse
Affiliation(s)
- Saharnaz Sarlak
- INSERM U1211, 33000 Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Claude Lalou
- INSERM U1211, 33000 Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Nivea Dias Amoedo
- CELLOMET, Functional Genomics Center (CGFB), 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Rodrigue Rossignol
- INSERM U1211, 33000 Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33000 Bordeaux, France; CELLOMET, Functional Genomics Center (CGFB), 146 rue Léo Saignat, 33000 Bordeaux, France.
| |
Collapse
|
11
|
Chu SH, Huang M, Kelly RS, Benedetti E, Siddiqui JK, Zeleznik OA, Pereira A, Herrington D, Wheelock CE, Krumsiek J, McGeachie M, Moore SC, Kraft P, Mathé E, Lasky-Su J. Integration of Metabolomic and Other Omics Data in Population-Based Study Designs: An Epidemiological Perspective. Metabolites 2019; 9:E117. [PMID: 31216675 PMCID: PMC6630728 DOI: 10.3390/metabo9060117] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 12/30/2022] Open
Abstract
It is not controversial that study design considerations and challenges must be addressed when investigating the linkage between single omic measurements and human phenotypes. It follows that such considerations are just as critical, if not more so, in the context of multi-omic studies. In this review, we discuss (1) epidemiologic principles of study design, including selection of biospecimen source(s) and the implications of the timing of sample collection, in the context of a multi-omic investigation, and (2) the strengths and limitations of various techniques of data integration across multi-omic data types that may arise in population-based studies utilizing metabolomic data.
Collapse
Affiliation(s)
- Su H Chu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Mengna Huang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Elisa Benedetti
- Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Jalal K Siddiqui
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Oana A Zeleznik
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Alexandre Pereira
- Department of Genetics and Molecular Medicine, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil.
| | - David Herrington
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden.
| | - Jan Krumsiek
- Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Michael McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA.
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA.
| | - Ewy Mathé
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Engle ML, Monk JN, Jania CM, Martin JR, Gomez JC, Dang H, Parker JS, Doerschuk CM. Dynamic changes in lung responses after single and repeated exposures to cigarette smoke in mice. PLoS One 2019; 14:e0212866. [PMID: 30818335 PMCID: PMC6395068 DOI: 10.1371/journal.pone.0212866] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/11/2019] [Indexed: 12/18/2022] Open
Abstract
Cigarette smoke is well recognized to cause injury to the airways and the alveolar walls over time. This injury usually requires many years of exposure, suggesting that the lungs may rapidly develop responses that initially protect it from this repetitive injury. Our studies tested the hypotheses that smoke induces an inflammatory response and changes in mRNA profiles that are dependent on sex and the health status of the lung, and that the response of the lungs to smoke differs after 1 day compared to 5 days of exposure. Male and female wildtype (WT) and Scnn1b-transgenic (βENaC) mice, which have chronic bronchitis and emphysematous changes due to dehydrated mucus, were exposed to cigarette smoke or sham air conditions for 1 or 5 days. The inflammatory response and gene expression profiles were analyzed in lung tissue. Overall, the inflammatory response to cigarette smoke was mild, and changes in mediators were more numerous after 1 than 5 days. βENaC mice had more airspace leukocytes than WT mice, and smoke exposure resulted in additional significant alterations. Many genes and gene sets responded similarly at 1 and 5 days: genes involved in oxidative stress responses were upregulated while immune response genes were downregulated. However, certain genes and biological processes were regulated differently after 1 compared to 5 days. Extracellular matrix biology genes and gene sets were upregulated after 1 day but downregulated by 5 days of smoke compared to sham exposure. There was no difference in the transcriptional response to smoke between WT and βENaC mice or between male and female mice at either 1 or 5 days. Taken together, these studies suggest that the lungs rapidly alter gene expression after only one exposure to cigarette smoke, with few additional changes after four additional days of repeated exposure. These changes may contribute to preventing lung damage.
Collapse
Affiliation(s)
- Michelle L. Engle
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Justine N. Monk
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Pathobiology and Translational Science Graduate Program, University of North Carolina, Chapel Hill, NC, United States of America
| | - Corey M. Jania
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| | - Jessica R. Martin
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - John C. Gomez
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - Joel S. Parker
- Department of Genetics, University of North Carolina, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America
| | - Claire M. Doerschuk
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| |
Collapse
|
13
|
Metabolomics and transcriptomics pathway approach reveals outcome-specific perturbations in COPD. Sci Rep 2018; 8:17132. [PMID: 30459441 PMCID: PMC6244246 DOI: 10.1038/s41598-018-35372-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) comprises multiple phenotypes such as airflow obstruction, emphysema, and frequent episodes of acute worsening of respiratory symptoms, known as exacerbations. The goal of this pilot study was to test the usefulness of unbiased metabolomics and transcriptomics approaches to delineate biological pathways associated with COPD phenotypes and outcomes. Blood was collected from 149 current or former smokers with or without COPD and separated into peripheral blood mononuclear cells (PBMC) and plasma. PBMCs and plasma were analyzed using microarray and liquid chromatography mass spectrometry, respectively. Statistically significant transcripts and compounds were mapped to pathways using IMPaLA. Results showed that glycerophospholipid metabolism was associated with worse airflow obstruction and more COPD exacerbations. Sphingolipid metabolism was associated with worse lung function outcomes and exacerbation severity requiring hospitalizations. The strongest associations between a pathway and a certain COPD outcome were: fat digestion and absorption and T cell receptor signaling with lung function outcomes; antigen processing with exacerbation frequency; arginine and proline metabolism with exacerbation severity; and oxidative phosphorylation with emphysema. Overlaying transcriptomic and metabolomics datasets across pathways enabled outcome and phenotypic differences to be determined. Findings are relevant for identifying molecular targets for animal intervention studies and early intervention markers in human cohorts.
Collapse
|
14
|
Liu TC, Kern JT, VanDussen KL, Xiong S, Kaiko GE, Wilen CB, Rajala MW, Caruso R, Holtzman MJ, Gao F, McGovern DP, Nunez G, Head RD, Stappenbeck TS. Interaction between smoking and ATG16L1T300A triggers Paneth cell defects in Crohn's disease. J Clin Invest 2018; 128:5110-5122. [PMID: 30137026 DOI: 10.1172/jci120453] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022] Open
Abstract
It is suggested that subtyping of complex inflammatory diseases can be based on genetic susceptibility and relevant environmental exposure (G+E). We propose that using matched cellular phenotypes in human subjects and corresponding preclinical models with the same G+E combinations is useful to this end. As an example, defective Paneth cells can subtype Crohn's disease (CD) subjects; Paneth cell defects have been linked to multiple CD susceptibility genes and are associated with poor outcome. We hypothesized that CD susceptibility genes interact with cigarette smoking, a major CD environmental risk factor, to trigger Paneth cell defects. We found that both CD subjects and mice with ATG16L1T300A (T300A; a prevalent CD susceptibility allele) developed Paneth cell defects triggered by tobacco smoke. Transcriptional analysis of full-thickness ileum and Paneth cell-enriched crypt base cells showed the T300A-smoking combination altered distinct pathways, including proapoptosis, metabolic dysregulation, and selective downregulation of the PPARγ pathway. Pharmacologic intervention by either apoptosis inhibitor or PPARγ agonist rosiglitazone prevented smoking-induced crypt apoptosis and Paneth cell defects in T300A mice and mice with conditional Paneth cell-specific knockout of Atg16l1. This study demonstrates how explicit G+E can drive disease-relevant phenotype and provides rational strategies for identifying actionable targets.
Collapse
Affiliation(s)
- Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Justin T Kern
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Kelli L VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Shanshan Xiong
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Gerard E Kaiko
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Craig B Wilen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Michael W Rajala
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Roberta Caruso
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | | | - Feng Gao
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Dermot Pb McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gabriel Nunez
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Richard D Head
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
15
|
Sears CR, Zhou H, Justice MJ, Fisher AJ, Saliba J, Lamb I, Wicker J, Schweitzer KS, Petrache I. Xeroderma Pigmentosum Group C Deficiency Alters Cigarette Smoke DNA Damage Cell Fate and Accelerates Emphysema Development. Am J Respir Cell Mol Biol 2018; 58:402-411. [PMID: 29111769 DOI: 10.1165/rcmb.2017-0251oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cigarette smoke (CS) exposure is a major risk factor for the development of emphysema, a common disease characterized by loss of cells comprising the lung parenchyma. The mechanisms of cell injury leading to emphysema are not completely understood but are thought to involve persistent cytotoxic or mutagenic DNA damage induced by CS. Using complementary cell culture and mouse models of CS exposure, we investigated the role of the DNA repair protein, xeroderma pigmentosum group C (XPC), on CS-induced DNA damage repair and emphysema. Expression of XPC was decreased in mouse lungs after chronic CS exposure and XPC knockdown in cultured human lung epithelial cells decreased their survival after CS exposure due to activation of the intrinsic apoptosis pathway. Similarly, cell autophagy and apoptosis were increased in XPC-deficient mouse lungs and were further increased by CS exposure. XPC deficiency was associated with structural and functional changes characteristic of emphysema, which were worsened by age, similar to levels observed with chronic CS exposure. Taken together, these findings suggest that repair of DNA damage by XPC plays an important and previously unrecognized role in the maintenance of alveolar structures. These findings support that loss of XPC, possibly due to chronic CS exposure, promotes emphysema development and further supports a link between DNA damage, impaired DNA repair, and development of emphysema.
Collapse
Affiliation(s)
| | | | - Matthew J Justice
- 1 Department of Medicine and.,2 Department of Medicine, National Jewish Health and University of Colorado Denver, Denver, Colorado
| | - Amanda J Fisher
- 3 Department of Anesthesia, Indiana University School of Medicine, Indianapolis, Indiana; and
| | | | | | | | - Kelly S Schweitzer
- 1 Department of Medicine and.,2 Department of Medicine, National Jewish Health and University of Colorado Denver, Denver, Colorado
| | - Irina Petrache
- 1 Department of Medicine and.,2 Department of Medicine, National Jewish Health and University of Colorado Denver, Denver, Colorado
| |
Collapse
|
16
|
Vishweswaraiah S, George L, Purushothaman N, Ganguly K. A candidate gene identification strategy utilizing mouse to human big-data mining: "3R-tenet" in COPD genetic research. Respir Res 2018; 19:92. [PMID: 29871630 PMCID: PMC5989378 DOI: 10.1186/s12931-018-0795-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/27/2018] [Indexed: 12/13/2022] Open
Abstract
Background Early life impairments leading to lower lung function by adulthood are considered as risk factors for chronic obstructive pulmonary disease (COPD). Recently, we compared the lung transcriptomic profile between two mouse strains with extreme total lung capacities to identify plausible pulmonary function determining genes using microarray analysis (GSE80078). Advancement of high-throughput techniques like deep sequencing (eg. RNA-seq) and microarray have resulted in an explosion of genomic data in the online public repositories which however remains under-exploited. Strategic curation of publicly available genomic data with a mouse-human translational approach can effectively implement “3R- Tenet” by reducing screening experiments with animals and performing mechanistic studies using physiologically relevant in vitro model systems. Therefore, we sought to analyze the association of functional variations within human orthologs of mouse lung function candidate genes in a publicly available COPD lung RNA-seq data-set. Methods Association of missense single nucleotide polymorphisms, insertions, deletions, and splice junction variants were analyzed for susceptibility to COPD using RNA-seq data of a Korean population (GSE57148). Expression of the associated genes were studied using the Gene Paint (mouse embryo) and Human Protein Atlas (normal adult human lung) databases. The genes were also assessed for replication of the associations and expression in COPD−/mouse cigarette smoke exposed lung tissues using other datasets. Results Significant association (p < 0.05) of variations in 20 genes to higher COPD susceptibility have been detected within the investigated cohort. Association of HJURP, MCRS1 and TLR8 are novel in relation to COPD. The associated ADAM19 and KIT loci have been reported earlier. The remaining 15 genes have also been previously associated to COPD. Differential transcript expression levels of the associated genes in COPD- and/ or mouse emphysematous lung tissues have been detected. Conclusion Our findings suggest strategic mouse-human datamining approaches can identify novel COPD candidate genes using existing datasets in the online repositories. The candidates can be further evaluated for mechanistic role through in vitro studies using appropriate primary cells/cell lines. Functional studies can be limited to transgenic animal models of only well supported candidate genes. This approach will lead to a significant reduction of animal experimentation in respiratory research. Electronic supplementary material The online version of this article (10.1186/s12931-018-0795-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Leema George
- SRM Research Institute, SRM University, Chennai, 603203, India
| | - Natarajan Purushothaman
- Department of Genetic Engineering, School of Bioengineering, Faculty of Engineering and Technology, SRM University, Chennai, 603203, India
| | - Koustav Ganguly
- SRM Research Institute, SRM University, Chennai, 603203, India. .,Work Environment Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 287, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
17
|
Kopa PN, Pawliczak R. Effect of smoking on gene expression profile – overall mechanism, impact on respiratory system function, and reference to electronic cigarettes. Toxicol Mech Methods 2018; 28:397-409. [DOI: 10.1080/15376516.2018.1461289] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Paulina Natalia Kopa
- Department of Immunopathology, Division of Allergology, Immunology and Dermatology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Division of Allergology, Immunology and Dermatology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
18
|
López-Hernández Y, Rivas-Santiago CE, López JA, Mendoza-Almanza G, Hernandez-Pando R. Tuberculosis and cigarette smoke exposure: An update of in vitro and in vivo studies. Exp Lung Res 2018; 44:113-126. [PMID: 29565741 DOI: 10.1080/01902148.2018.1444824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tuberculosis (TB) has been declared the first cause of death by an infectious agent. Annually, 10.4 million people suffer active TB. Most infected individuals live in low-income countries, where social and economic conditions enhance the dissemination and progression of the disease. These countries have a high percentage of smokers. Thousands of studies have linked cigarette smoke (CS) with increased risk of many diseases, such as cancer and lung diseases. Numerous in vitro studies have been conducted to evaluate the general and specific toxic effects of CS in lung immune function. Smoke exposure increases the risk of TB development three-fold. However, until now, only few animal studies have been performed to analyze the association between smoke and TB. In the present work, we review in vitro and in vivo studies whose aim was to analyze the molecular basis of TB susceptibility caused by exposure to CS.
Collapse
Affiliation(s)
- Y López-Hernández
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - C E Rivas-Santiago
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - J A López
- b Laboratorio de MicroRNAs, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - G Mendoza-Almanza
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - R Hernandez-Pando
- c Departamento de Patologia, Unidad de Patologia Experimental , Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran , Mexico
| |
Collapse
|
19
|
Metabolomic similarities between bronchoalveolar lavage fluid and plasma in humans and mice. Sci Rep 2017; 7:5108. [PMID: 28698669 PMCID: PMC5505974 DOI: 10.1038/s41598-017-05374-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
This observational study catalogues the overlap in metabolites between matched bronchoalveolar lavage fluid (BALF) and plasma, identifies the degree of congruence between these metabolomes in human and mouse, and determines how molecules may change in response to cigarette smoke (CS) exposure. Matched BALF and plasma was collected from mice (ambient air or CS-exposed) and humans (current or former smokers), and analyzed using mass spectrometry. There were 1155 compounds in common in all 4 sample types; fatty acyls and glycerophospholipids strongly overlapped between groups. In humans and mice, more than half of the metabolites present in BALF were also present in plasma. Mouse BALF and human BALF had a strong positive correlation with 2040 metabolites in common, suggesting that mouse models can be used to interrogate human lung metabolome changes. While power was affected by small sample size in the mouse study, the BALF metabolome appeared to be more affected by CS than plasma. CS-exposed mice showed increased plasma and BALF glycerolipids and glycerophospholipids. This is the first report cataloguing the metabolites present across mouse and human, BALF and plasma. Findings are relevant to translational studies where mouse models are used to examine human disease, and where plasma may be interrogated in lieu of BALF or lung tissue.
Collapse
|