1
|
Shastry A, Wilkinson MS, Miller DM, Kuriakose M, Veeneman JLMH, Smith MR, Hindmarch CCT, Dunham-Snary KJ. Multi-tissue metabolomics reveal mtDNA- and diet-specific metabolite profiles in a mouse model of cardiometabolic disease. Redox Biol 2025; 81:103541. [PMID: 39983345 PMCID: PMC11893332 DOI: 10.1016/j.redox.2025.103541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/08/2025] [Indexed: 02/23/2025] Open
Abstract
RATIONALE Excess consumption of sugar- and fat-rich foods has heightened the prevalence of cardiometabolic disease, which remains a driver of cardiovascular disease- and type II diabetes-related mortality globally. Skeletal muscle insulin resistance is an early feature of cardiometabolic disease and is a precursor to diabetes. Insulin resistance risk varies with self-reported race, whereby African-Americans have a greater risk of diabetes development relative to their White counterparts. Self-reported race is strongly associated with mitochondrial DNA (mtDNA) haplogroups, and previous reports have noted marked differences in bioenergetic and metabolic parameters in cells belonging to distinct mtDNA haplogroups, but the mechanism of these associations remains unknown. Additionally, distinguishing nuclear DNA (nDNA) and mtDNA contributions to cardiometabolic disease remains challenging in humans. The Mitochondrial-Nuclear eXchange (MNX) mouse model enables in vivo preclinical investigation of the role of mtDNA in cardiometabolic disease development, and has been implemented in studies of insulin resistance, fatty liver disease, and obesity in previous reports. METHODS Six-week-old male C57nDNA:C57mtDNA and C3HnDNA:C3HmtDNA wild-type mice, and C57nDNA:C3HmtDNA and C3HnDNA:C57mtDNA MNX mice, were fed sucrose-matched high-fat (45% kcal fat) or control diet (10% kcal fat) until 12 weeks of age (n = 5/group). Mice were weighed weekly and total body fat was collected at euthanasia. Gastrocnemius skeletal muscle and plasma metabolomes were characterized using untargeted dual-chromatography mass spectrometry; both hydrophilic interaction liquid chromatography (HILIC) and C18 columns were used, in positive- and negative-ion modes, respectively. RESULTS Comparative analyses between nDNA-matched wild-type and MNX strains demonstrated significantly increased body fat percentage in mice possessing C57mtDNA regardless of nDNA background. High-fat diet in mice possessing C57mtDNA was associated with differential abundance of phosphatidylcholines, lysophosphatidylcholines, phosphatidylethanolamines, and glucose. Conversely, high-fat diet in mice possessing C3HmtDNA was associated with differential abundance of phosphatidylcholines, cardiolipins, and alanine. Glycerophospholipid metabolism and beta-alanine signaling pathways were enriched in skeletal muscle and plasma, indicating mtDNA-directed priming of mitochondria towards oxidative stress and increased fatty acid oxidation in C57nDNA:C57mtDNA wild-type and C3HnDNA:C57mtDNA MNX mice, relative to their nDNA-matched counterparts. In mtDNA-matched mice, C57mtDNA was associated with metabolite co-expression related to the pentose phosphate pathway and sugar-related metabolism; C3HmtDNA was associated with branched chain amino acid metabolite co-expression. CONCLUSIONS These results reveal novel nDNA-mtDNA interactions that drive significant changes in metabolite levels. Alterations to key metabolites involved in mitochondrial bioenergetic dysfunction and electron transport chain activity are implicated in elevated beta-oxidation during high-fat diet feeding; abnormally elevated rates of beta-oxidation may be a key driver of insulin resistance. The results reported here support the hypothesis that mtDNA influences cardiometabolic disease-susceptibility by modulating mitochondrial function and metabolic pathways.
Collapse
Affiliation(s)
- Abhishek Shastry
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Mia S Wilkinson
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Dalia M Miller
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Michelle Kuriakose
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | | | - Matthew Ryan Smith
- Atlanta Veterans Affairs Health Care System, Decatur, GA, 30033, USA; Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Charles C T Hindmarch
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada; Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada; Queen's CardioPulmonary Unit, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Kimberly J Dunham-Snary
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada; Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
2
|
Intharuksa A, Arunotayanun W, Takuathung MN, Boongla Y, Chaichit S, Khamnuan S, Prasansuklab A. Therapeutic Potential of Herbal Medicines in Combating Particulate Matter (PM)-Induced Health Effects: Insights from Recent Studies. Antioxidants (Basel) 2024; 14:23. [PMID: 39857357 PMCID: PMC11762796 DOI: 10.3390/antiox14010023] [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: 11/21/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Particulate matter (PM), particularly fine (PM2.5) and ultrafine (PM0.1) particles, originates from both natural and anthropogenic sources, such as biomass burning and vehicle emissions. These particles contain harmful compounds that pose significant health risks. Upon inhalation, ingestion, or dermal contact, PM can penetrate biological systems, inducing oxidative stress, inflammation, and DNA damage, which contribute to a range of health complications. This review comprehensively examines the protective potential of natural products against PM-induced health issues across various physiological systems, including the respiratory, cardiovascular, skin, neurological, gastrointestinal, and ocular systems. It provides valuable insights into the health risks associated with PM exposure and highlights the therapeutic promise of herbal medicines by focusing on the natural products that have demonstrated protective properties in both in vitro and in vivo PM2.5-induced models. Numerous herbal medicines and phytochemicals have shown efficacy in mitigating PM-induced cellular damage through their ability to counteract oxidative stress, suppress pro-inflammatory responses, and enhance cellular defense mechanisms. These combined actions collectively protect tissues from PM-related damage and dysfunction. This review establishes a foundation for future research and the development of effective interventions to combat PM-related health issues. However, further studies, including in vivo and clinical trials, are essential to evaluate the safety, optimal dosages, and long-term effectiveness of herbal treatments for patients under chronic PM exposure.
Collapse
Affiliation(s)
- Aekkhaluck Intharuksa
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (A.I.); (S.C.)
| | - Warunya Arunotayanun
- Kanchanabhishek Institute of Medical and Public Health Technology, Faculty of Public Health and Allied Health Science, Praboromarajchanok Institute, Nonthaburi 11150, Thailand
| | - Mingkwan Na Takuathung
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Yaowatat Boongla
- Department of Sustainable Development Technology, Faculty of Science and Technology, Thammasat University, Pathum Thani 12120, Thailand;
| | - Siripat Chaichit
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (A.I.); (S.C.)
| | - Suthiwat Khamnuan
- Faculty of Pharmacy, Western University, Pathum Thani 12150, Thailand;
| | - Anchalee Prasansuklab
- College of Public Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
3
|
Jang HJ, Min HY, Kang YP, Boo HJ, Kim J, Ahn JH, Oh SH, Jung JH, Park CS, Park JS, Kim SY, Lee HY. Tobacco-induced hyperglycemia promotes lung cancer progression via cancer cell-macrophage interaction through paracrine IGF2/IR/NPM1-driven PD-L1 expression. Nat Commun 2024; 15:4909. [PMID: 38851766 PMCID: PMC11162468 DOI: 10.1038/s41467-024-49199-9] [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: 07/15/2022] [Accepted: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
Tobacco smoking (TS) is implicated in lung cancer (LC) progression through the development of metabolic syndrome. However, direct evidence linking metabolic syndrome to TS-mediated LC progression remains to be established. Our findings demonstrate that 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and benzo[a]pyrene (NNK and BaP; NB), components of tobacco smoke, induce metabolic syndrome characteristics, particularly hyperglycemia, promoting lung cancer progression in male C57BL/6 J mice. NB enhances glucose uptake in tumor-associated macrophages by increasing the expression and surface localization of glucose transporter (GLUT) 1 and 3, thereby leading to transcriptional upregulation of insulin-like growth factor 2 (IGF2), which subsequently activates insulin receptor (IR) in LC cells in a paracrine manner, promoting its nuclear import. Nuclear IR binds to nucleophosmin (NPM1), resulting in IR/NPM1-mediated activation of the CD274 promoter and expression of programmed death ligand-1 (PD-L1). Restricting glycolysis, depleting macrophages, or blocking PD-L1 inhibits NB-mediated LC progression. Analysis of patient tissues and public databases reveals elevated levels of IGF2 and GLUT1 in tumor-associated macrophages, as well as tumoral PD-L1 and phosphorylated insulin-like growth factor 1 receptor/insulin receptor (pIGF-1R/IR) expression, suggesting potential poor prognostic biomarkers for LC patients. Our data indicate that paracrine IGF2/IR/NPM1/PD-L1 signaling, facilitated by NB-induced dysregulation of glucose levels and metabolic reprogramming of macrophages, contributes to TS-mediated LC progression.
Collapse
Affiliation(s)
- Hyun-Ji Jang
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Young Min
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Jin Boo
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Histology, College of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jisung Kim
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jee Hwan Ahn
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Ho Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin Hwa Jung
- PET core, Convergence Medicine Research Center, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Choon-Sik Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do, 14584, Republic of Korea
| | - Jong-Sook Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do, 14584, Republic of Korea
| | - Seog-Young Kim
- PET core, Convergence Medicine Research Center, Asan Medical Center, Seoul, 05505, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Ho-Young Lee
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
4
|
Batista JM, Neves MJ, Menezes HC, Cardeal ZL. Evaluation of amino acid profile by targeted metabolomics in the eukaryotic model under exposure of benzo[a]pyrene as the exclusive stressor. Talanta 2023; 265:124859. [PMID: 37393711 DOI: 10.1016/j.talanta.2023.124859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/09/2023] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
Amino acids (AAs) are a class of important metabolites in metabolomics methodology that investigates metabolite changes in a cell, tissue, or organism for early diagnosis of diseases. Benzo[a]pyrene (BaP) is considered a priority contaminant by different environmental control agencies because it is a proven carcinogenic compound for humans. Therefore, it is important to evaluate the BaP interference in the metabolism of amino acids. In this work, a new amino acid extraction procedure (derivatized with propyl chloroformate/propanol) using functionalized magnetic carbon nanotubes was developed and optimized. A hybrid nanotube was used followed by desorption without heating, and excellent extraction of analytes was obtained. After exposure of Saccharomyces cerevisiae, the BaP concentration of 25.0 μmol L-1 caused changes in cell viability, indicating metabolic changes. A fast and efficient GC/MS method using a Phenomenex ZB-AAA column was optimized, enabling the determination of 16 AAs in yeasts exposed or not to BaP. A comparison of AA concentrations obtained in the two experimental groups showed that glycine (Gly), serine (Ser), phenylalanine (Phe), proline (Pro), asparagine (Asn), aspartic acid (Asp), glutamic acid (Glu), tyrosine (Tyr), and leucine (Leu) statistically differentiated, after subsequent application of ANOVA with Bonferroni post-hoc test, with a confidence level of 95%. This amino acid pathway analysis confirmed previous studies that revealed the potential of these AAs as toxicity biomarker candidates.
Collapse
Affiliation(s)
- Josimar M Batista
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, 31270901, Belo Horizonte, MG, Brazil
| | - Maria J Neves
- Nuclear Technology Development Center/National Nuclear Energy Commission (CDTN/CNEN), Belo Horizonte, MG, Brazil
| | - Helvécio C Menezes
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, 31270901, Belo Horizonte, MG, Brazil
| | - Zenilda L Cardeal
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627, 31270901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
5
|
Xing Y, Liu Z, Ma X, Zhou C, Wang Y, Yao B, Fu J, Qi Y, Zhao P. Targeted metabolomics analysis identified the role of FOXA1 in the change in glutamate-glutamine metabolic pattern of BaP malignantly transformed 16HBE cells. Toxicol Appl Pharmacol 2023; 461:116402. [PMID: 36702312 DOI: 10.1016/j.taap.2023.116402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023]
Abstract
The carcinogenic mechanism of benzo[a]pyrene (BaP) is far from being elucidated. FOXA1 has been confirmed to play an oncogenic role in BaP-transformed cell THBEc1. To explore the changes in amino acid metabolic patterns, especially glutamate-glutamine (Glu-Gln) metabolic pattern caused by BaP-induced transformation and the possible role FOXA1 might play in it, we compared amino acid metabolic characteristics between THBEc1 cells and control 16HBE cells using a targeted metabolomics method and determined the effects of FOXA1 knockout on the amino acid metabolic pattern using FOXA1 knockout cell THBEc1-ΔFOXA1-c34. The amino acid metabolic patterns of THBEc1 and 16HBE cells were different, which was manifested by the differential consumption of 18 amino acids and the difference in the intracellular content of 21 amino acids. The consumption and intracellular content of Glu and Gln are different between the two types of cells, accompanied by upregulation of FOXA1, GLUL, SLC1A3, SLC1A4, SLC1A5 and SLC6A14, and downregulation of FOXA2 and GPT2 in THBEc1 cells. FOXA1 knockout changed the consumption of 19 amino acids and the intracellular content of 21 amino acids and reversed the metabolic pattern of Glu and the changes in FOXA2, GLUL, SLC1A3 and SLC6A14 in THBEc1 cells. Additionally, FOXA1 knockout inhibited cell proliferation and further increased the dependence of THBEc1 cells on Glu. In conclusion, FOXA1 knockout partially reversed the change in Glu-Gln metabolism caused by BaP-induced transformation by upregulating the expression of GLUL and SLC1A3. Our findings provide a clue for the possible role of FOXA1 in amino acid metabolism regulation.
Collapse
Affiliation(s)
- Yunkun Xing
- Department of Toxicology, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Zhiyu Liu
- Department of Toxicology, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Xue Ma
- Department of Toxicology, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China; Zhejiang Province Center for Disease Control and Prevention, Hangzhou 310051, People's Republic of China
| | - Chuan Zhou
- Department of Toxicology, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Yu Wang
- National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 10021, People's Republic of China
| | - Biyun Yao
- Department of Toxicology, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Juanling Fu
- Department of Toxicology, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Yanmin Qi
- Civil Aviation Medicine Center, Civil Aviation Administration of China, Beijing 10123, People's Republic of China
| | - Peng Zhao
- Department of Toxicology, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University Health Science Center, Beijing 100191, People's Republic of China.
| |
Collapse
|
6
|
Misra N, Clavaud C, Guinot F, Bourokba N, Nouveau S, Mezzache S, Palazzi P, Appenzeller BMR, Tenenhaus A, Leung MHY, Lee PKH, Bastien P, Aguilar L, Cavusoglu N. Multi-omics analysis to decipher the molecular link between chronic exposure to pollution and human skin dysfunction. Sci Rep 2021; 11:18302. [PMID: 34526566 PMCID: PMC8443591 DOI: 10.1038/s41598-021-97572-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022] Open
Abstract
Environmental pollution is composed of several factors, namely particulate matter (PM2.5, PM10), ozone and Ultra Violet (UV) rays among others and first and the most exposed tissue to these substances is the skin epidermis. It has been established that several skin disorders such as eczema, acne, lentigines and wrinkles are aggravated by exposure to atmospheric pollution. While pollutants can interact with skin surface, contamination of deep skin by ultrafine particles or Polycyclic aromatic hydrocarbons (PAH) might be explained by their presence in blood and hair cortex. Molecular mechanisms leading to skin dysfunction due to pollution exposure have been poorly explored in humans. In addition to various host skin components, cutaneous microbiome is another target of these environment aggressors and can actively contribute to visible clinical manifestation such as wrinkles and aging. The present study aimed to investigate the association between pollution exposure, skin microbiota, metabolites and skin clinical signs in women from two cities with different pollution levels. Untargeted metabolomics and targeted proteins were analyzed from D-Squame samples from healthy women (n = 67 per city), aged 25-45 years and living for at least 15 years in the Chinese cities of Baoding (used as a model of polluted area) and Dalian (control area with lower level of pollution). Additional samples by swabs were collected from the cheeks from the same population and microbiome was analysed using bacterial 16S rRNA as well as fungal ITS1 amplicon sequencing and metagenomics analysis. The level of exposure to pollution was assessed individually by the analysis of polycyclic aromatic hydrocarbons (PAH) and their metabolites in hair samples collected from each participant. All the participants of the study were assessed for the skin clinical parameters (acne, wrinkles, pigmented spots etc.). Women from the two cities (polluted and less polluted) showed distinct metabolic profiles and alterations in skin microbiome. Profiling data from 350 identified metabolites, 143 microbes and 39 PAH served to characterize biochemical events that correlate with pollution exposure. Finally, using multiblock data analysis methods, we obtained a potential molecular map consisting of multi-omics signatures that correlated with the presence of skin pigmentation dysfunction in individuals living in a polluted environment. Overall, these signatures point towards macromolecular alterations by pollution that could manifest as clinical sign of early skin pigmentation and/or other imperfections.
Collapse
Affiliation(s)
- Namita Misra
- Research and Innovation, L'Oréal SA, Aulnay Sous Bois, France.
| | - Cécile Clavaud
- Research and Innovation, L'Oréal SA, Aulnay Sous Bois, France
| | - Florent Guinot
- Research and Innovation, L'Oréal SA, Aulnay Sous Bois, France
| | | | | | - Sakina Mezzache
- Research and Innovation, L'Oréal SA, Aulnay Sous Bois, France
| | - Paul Palazzi
- Human Biomonitoring Research Unit, Luxembourg Institute of Health, Strassen, Luxemburg
| | - Brice M R Appenzeller
- Human Biomonitoring Research Unit, Luxembourg Institute of Health, Strassen, Luxemburg
| | - Arthur Tenenhaus
- CentraleSupelec Laboratoire des Signaux et Systemes, Université Paris-Saclay, CNRS, Gif-sur-Yvette, France
- Brain and Spine Institute, Paris, France
| | - Marcus H Y Leung
- School of Energy and Environment, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Patrick K H Lee
- School of Energy and Environment, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | | | - Luc Aguilar
- Research and Innovation, L'Oréal SA, Aulnay Sous Bois, France
| | | |
Collapse
|
7
|
Ran Z, An Y, Zhou J, Yang J, Zhang Y, Yang J, Wang L, Li X, Lu D, Zhong J, Song H, Qin X, Li R. Subchronic exposure to concentrated ambient PM2.5 perturbs gut and lung microbiota as well as metabolic profiles in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 272:115987. [PMID: 33213950 DOI: 10.1016/j.envpol.2020.115987] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/12/2020] [Accepted: 11/01/2020] [Indexed: 05/21/2023]
Abstract
Exposure to ambient fine particular matter (PM2.5) are linked to an increased risk of metabolic disorders, leading to enhanced rate of many diseases, such as inflammatory bowel disease (IBD), cardiovascular diseases, and pulmonary diseases; nevertheless, the underlying mechanisms remain poorly understood. In this study, BALB/c mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CPM) for 2 months using a versatile aerosol concentration enrichment system(VACES). We found subchronic CPM exposure caused significant lung and intestinal damage, as well as systemic inflammatory reactions. In addition, serum and BALFs (bronchoalveolar lavage fluids) metabolites involved in many metabolic pathways in the CPM exposed mice were markedly disrupted upon PM2.5 exposure. Five metabolites (glutamate, glutamine, formate, pyruvate and lactate) with excellent discriminatory power (AUC = 1, p < 0.001) were identified to predict PM2.5 exposure related toxicities. Furthermore, subchronic exposure to CPM not only significantly decreased the richness and composition of the gut microbiota, but also the lung microbiota. Strong associations were found between several gut and lung bacterial flora changes and systemic metabolic abnormalities. Our study showed exposure to ambient PM2.5 not only caused dysbiosis in the gut and lung, but also significant systemic and local metabolic alterations. Alterations in gut and lung microbiota were strongly correlated with metabolic abnormalities. Our study suggests potential roles of gut and lung microbiota in PM2.5 caused metabolic disorders.
Collapse
Affiliation(s)
- Zihan Ran
- Department of Research, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhouyuan Road, 201318, Shanghai, China; Inspection and Quarantine Department, The College of Medical Technology, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Road, 201318, Shanghai, China
| | - Yanpeng An
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Fudan University, Shanghai, 200438, China
| | - Ji Zhou
- Department of Atmospheric and Oceanic Sciences & Institute of Atmospheric Sciences, Fudan University, Shanghai, China
| | - Jingmin Yang
- Key Laboratory of Birth Defects and Reproductive Health of National Health and Family Planning Commission (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning, Science and Technology Research Institute), Chongqing, 400020, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Youyi Zhang
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
| | - Jingcheng Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Lei Wang
- Department of Oral & Maxillofacial - Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Xin Li
- Department of Oral & Maxillofacial - Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China; Key Laboratory of Birth Defects and Reproductive Health of National Health and Family Planning Commission (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning, Science and Technology Research Institute), Chongqing, 400020, China
| | - Jiang Zhong
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438, China
| | - Huaidong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200011, China
| | - Xingjun Qin
- Department of Oral & Maxillofacial - Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Rui Li
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostic & Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
8
|
Smith MR, Jarrell ZR, Orr M, Liu KH, Go YM, Jones DP. Metabolome-wide association study of flavorant vanillin exposure in bronchial epithelial cells reveals disease-related perturbations in metabolism. ENVIRONMENT INTERNATIONAL 2021; 147:106323. [PMID: 33360165 PMCID: PMC7856097 DOI: 10.1016/j.envint.2020.106323] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 06/12/2023]
Abstract
Electronic cigarettes (e-cig) are an increasingly popular alternative to traditional smoking but have been in use for too short of a period of time to fully understand health risks. Furthermore, associated health risks are difficult to evaluate because of a large range of flavoring agents and their combinations for use with e-cig. Many flavoring agents are generally regarded as safe but have limited studies for effects on lung. Vanillin, an aromatic aldehyde, is one of the most commonly used flavoring agents in e-cig. Vanillin is electrophilic and can be redox active, with chemical properties expected to interact within biologic systems. Because accumulating lung metabolomics studies have identified metabolic disruptions associated with idiopathic pulmonary fibrosis, asthma and acute respiratory distress syndrome, we used human bronchial epithelial cells (BEAS-2B) with high-resolution metabolomics analysis to determine whether these disease-associated pathways are impacted by vanillin over the range used in e-cig. A metabolome-wide association study showed that vanillin perturbed specific energy, amino acid, antioxidant and sphingolipid pathways previously associated with human disease. Analysis of a small publicly available human dataset showed associations with several of the same pathways. Because vanillin is a common and high-abundance flavorant in e-cig, these results show that vanillin has potential to be mechanistically important in lung diseases and warrants in vivo toxicity testing in the context of e-cig use.
Collapse
Affiliation(s)
- Matthew Ryan Smith
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA
| | - Zachery R Jarrell
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA
| | - Michael Orr
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ken H Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA
| | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
9
|
Advances in Comprehensive Exposure Assessment: Opportunities for the US Military. J Occup Environ Med 2020; 61 Suppl 12:S5-S14. [PMID: 31800446 DOI: 10.1097/jom.0000000000001677] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Review advances in exposure assessment offered by the exposome concept and new -omics and sensor technologies. METHODS Narrative review of advances, including current efforts and potential future applications by the US military. RESULTS Exposure assessment methods from both bottom-up and top-down exposomics approaches are advancing at a rapid pace, and the US military is engaged in developing both approaches. Top-down approaches employ various -omics technologies to identify biomarkers of internal exposure and biological effect. Bottom-up approaches use new sensor technology to better measure external dose. Key challenges of both approaches are largely centered around how to integrate, analyze, and interpret large datasets that are multidimensional and disparate. CONCLUSIONS Advances in -omics and sensor technologies may dramatically enhance exposure assessment and improve our ability to characterize health risks related to occupational and environmental exposures, including for the US military.
Collapse
|
10
|
Use of Biomarkers to Assess Environmental Exposures and Health Outcomes in Deployed Troops. J Occup Environ Med 2020; 61 Suppl 12:S1-S4. [PMID: 31800445 DOI: 10.1097/jom.0000000000001752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This paper provides an overview of our Military Biomarkers Research Study (MBRS) designed to assess whether biomarkers can be used to retrospectively assess deployment exposures and health impacts related to deployment environmental exposures. METHODS The MBRS consists of four phases. Phase I was a feasibility study of stored sera. Phase II looks at associations between exposures and biomarkers. Phase III examines relationships of biomarkers and health outcomes, and Phase IV investigates in vitro biomarker changes associated with exposures to chemicals of interest. This paper briefly summarizes work already published and introduces the new reports contained in this supplement. RESULTS Novel biomarkers were identified. These were associated with deployment exposures. CONCLUSIONS Significant associations were noted between deployment exposures, microRNA biomarkers and metabolomic biomarkers, and deployment health outcomes.
Collapse
|
11
|
Jones DP, Cohn BA. A vision for exposome epidemiology: The pregnancy exposome in relation to breast cancer in the Child Health and Development Studies. Reprod Toxicol 2020; 92:4-10. [PMID: 32197999 PMCID: PMC7306421 DOI: 10.1016/j.reprotox.2020.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Etiology of complex diseases, such as breast cancer, involves multiple genetic, behavioral and environmental factors. Gene sequencing enabled detection of genetic risks with relatively small effect size, and high-resolution metabolomics (HRM) to provide omics level data for exposures is poised to do the same for environmental epidemiology. Coupling HRM to the Child Health and Development Studies (CHDS) cohort combines two unique resources to create a prototype for exposome epidemiology, in which omics scale measures of exposure are used for study of distribution and determinants of health and disease. Using this approach, exposures and biologic responses during pregnancy have been linked to breast cancer in the CHDS. With improved chemical coverage and extension to larger populations and other disease processes, development of exposome epidemiology portends discovery of new disease-associated environment factors with small effect size as well as new capabilities to disentangle these from behavioral and other risk factors.
Collapse
Affiliation(s)
- Dean P Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, 1683 Shattuck Avenue, Suite B, Berkeley, CA 94709, USA
| |
Collapse
|
12
|
Smith MR, Woeller CF, Uppal K, Thatcher TH, Walker DI, Hopke PK, Rohrbeck P, Mallon TM, Krahl PL, Utell MJ, Go YM, Jones DP. Associations of Benzo(ghi)perylene and Heptachlorodibenzo-p-dioxin in Serum of Service Personnel Deployed to Balad, Iraq, and Bagram, Afghanistan Correlates With Perturbed Amino Acid Metabolism in Human Lung Fibroblasts. J Occup Environ Med 2019; 61 Suppl 12:S35-S44. [PMID: 31800449 PMCID: PMC7861127 DOI: 10.1097/jom.0000000000001669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE A study was conducted to identify metabolic-related effects of benzo(ghi)perylene (BghiP) and 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin (HpCDD), on primary human fibroblasts to verify biological associations previously found in occupational health research. METHODS Human lung fibroblasts were exposed to BghiP or HpCDD and extracts were analyzed with a metabolome-wide association study to test for pathways and metabolites altered relative to controls. Gene expression was measured by quantitative-real time polymerase chain reaction. RESULTS Metabolic perturbations in amino-acid, oxidative stress, and fatty-acid pathways were observed for BghiP and HpCDD. HpCDD but not BghiP exposure increased gene expression of the amino acid transporters SLC7A5 and SLC7A11. CONCLUSIONS Exposure to polycyclic aromatic hydrocarbons (PAH) or dioxins perturbs amino acid pathways at physiologically relevant concentrations with different mechanisms. These findings imply an effect on central homeostatic systems by environmental exposures which could have implications on disease susceptibility.
Collapse
Affiliation(s)
- Matthew Ryan Smith
- Clinical Biomarkers Laboratory, Division of Pulmonary Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Collynn F. Woeller
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester NY
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Thomas H. Thatcher
- Department of Medicine, Pulmonary Division, University of Rochester Medical Center, Rochester, NY
| | - Douglas I. Walker
- Clinical Biomarkers Laboratory, Division of Pulmonary Medicine, Department of Medicine, Emory University, Atlanta, GA
- Current address: Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Philip K. Hopke
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY
| | | | - Timothy M. Mallon
- Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Department of Preventive Medicine & Biostatistics, Bethesda, MD
| | - Pamela L. Krahl
- Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Department of Preventive Medicine & Biostatistics, Bethesda, MD
| | - Mark J. Utell
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester NY
- Department of Medicine, Pulmonary Division, University of Rochester Medical Center, Rochester, NY
| | - Young-Mi Go
- Clinical Biomarkers Laboratory, Division of Pulmonary Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Dean P. Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary Medicine, Department of Medicine, Emory University, Atlanta, GA
| |
Collapse
|