1
|
Daiber A, Rajagopalan S, Kuntic M, Münzel T. Cardiovascular risk posed by the exposome. Atherosclerosis 2025; 405:119222. [PMID: 40339362 DOI: 10.1016/j.atherosclerosis.2025.119222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025]
Abstract
Chronic non-communicable diseases (NCDs) account for 2/3 of global deaths annually, primarily due to an aging population and external risk factors such as air/water/soil pollution, traffic noise, mental stress, and climate change emanating from the environment. These factors contribute to premature deaths and loss of healthy life years, as reflected by disability-adjusted life years. The exposome concept was proposed 16 years ago as a new research field to investigate environment-health associations, also by considering the underlying pathophysiological pathways. The exposome describes lifelong environmental exposures, besides pollutants also socioeconomic and lifestyle factors, aiming to explain the associated diseases and deaths. The exposome can be divided into the specific and general external environment and further subcategories such as organ-specific exposomes as well as spatially and temporally restricted pollutomes. The exposome also shows considerable interaction with genetic predisposition and pre-established chronic diseases, characteristics of the vulnerable groups. The present overview provides background information on the impact of the environment on health and disease by considering recent data of the Global Burden of Disease Study. We also explain the exposome concept with the help of selected studies, briefly describe how the exposome is measured, and discuss biomarkers identified by exposomic research and their impact on the development and progression of atherosclerosis. Major pathophysiological pathways comprise exacerbated stress hormone signaling, oxidative stress, inflammation and circadian rhythm dysregulation promoting impairment of cardiometabolic function. The present overview highlights the relevance of the exposome for future health research and preventive medicine, especially concerning cardiovascular diseases and therapy.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Centre for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany.
| | - Sanjay Rajagopalan
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Marin Kuntic
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Centre for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Centre for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| |
Collapse
|
2
|
Fadadu RP, Bozack AK, Cardenas A. Chemical and climatic environmental exposures and epigenetic aging: A systematic review. ENVIRONMENTAL RESEARCH 2025; 274:121347. [PMID: 40058550 PMCID: PMC12048242 DOI: 10.1016/j.envres.2025.121347] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Epigenetic aging biomarkers are used for evaluating morbidity and mortality, monitoring therapies, and direct-to-consumer testing. However, the influence of environmental exposures on epigenetic age acceleration (EAA), also known as epigenetic age deviation, has not been systematically evaluated. In this systematic review, we synthesized findings from human epidemiologic studies on chemical and climatic environmental exposures, particularly air pollution, chemicals, metals, climate, and cigarette smoke, and EAA. A total of 102 studies analyzing epigenetic data from over 180,000 subjects were evaluated. Overall, studies in each exposure category frequently included adult participants, used a variety of epigenetic clocks, analyzed whole blood samples, and had a low risk of bias. Exposure to air pollution (15/19 of studies; 79%), cigarette smoke (53/66; 80%), and synthetic and occupational chemicals (5/8; 63%) were notably associated with increased EAA. Results for essential and non-essential metal exposure were more equivocal: 7/13 studies (54%) reported increased EAA. One study reported increased EAA with greater temperature exposure. In summary, we identified environmental exposures, such as air pollution and cigarette smoke, that were strongly associated with increased EAA. Further research is needed with larger and more diverse samples and high-quality exposure assessment.
Collapse
Affiliation(s)
- Raj P Fadadu
- Department of Epidemiology and Population Health, Stanford School of Medicine, 1701 Page Mill Rd., Stanford, CA, 94304, USA; Department of Dermatology, University of California San Diego School of Medicine, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Anne K Bozack
- Department of Epidemiology and Population Health, Stanford School of Medicine, 1701 Page Mill Rd., Stanford, CA, 94304, USA
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford School of Medicine, 1701 Page Mill Rd., Stanford, CA, 94304, USA.
| |
Collapse
|
3
|
Davis KA, Coleman JR, Adams M, Breen G, Cai N, Davies HL, Davies K, Dregan A, Eley TC, Fox E, Holliday J, Hübel C, John A, Kassam AS, Kempton MJ, Lee W, Li D, Maina J, McCabe R, McIntosh AM, Oram S, Richards M, Skelton M, Starkey F, ter Kuile AR, Thornton LM, Wang R, Yu Z, Zvrskovec J, Hotopf M. The UK Biobank mental health enhancement 2022: Methods and results. PLoS One 2025; 20:e0324189. [PMID: 40435106 PMCID: PMC12118920 DOI: 10.1371/journal.pone.0324189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/22/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND This paper introduces the UK Biobank (UKB) second mental health questionnaire (MHQ2), describes its design, the respondents and some notable findings. UKB is a large cohort study with over 500,000 volunteer participants aged 40-69 years when recruited in 2006-2010. It is an important resource of extensive health, genetic and biomarker data. Enhancements to UKB enrich the data available. MHQ2 is an enhancement designed to enable and facilitate research with psychosocial and mental health aspects. METHODS UKB sent participants a link to MHQ2 by email in October-November 2022. The MHQ2 was designed by a multi-institutional consortium to build on MHQ1. It characterises lifetime depression further, adds data on panic disorder and eating disorders, repeats 'current' mental health measures and updates information about social circumstances. It includes established measures, such as the PHQ-9 for current depression and CIDI-SF for lifetime panic, as well as bespoke questions. Algorithms and R code were developed to facilitate analysis. RESULTS At the time of analysis, MHQ2 results were available for 169,253 UKB participants, of whom 111,275 had also completed the earlier MHQ1. Characteristics of respondents and the whole UKB cohort are compared. The major phenotypes are lifetime: depression (18%); panic disorder (4.0%); a specific eating disorder (2.8%); and bipolar affective disorder I (0.4%). All mental disorders are found less with older age and also seem to be related to selected social factors. In those participants who answered both MHQ1 (2016) and MHQ2 (2022), current mental health measure showed that fewer respondents have harmful alcohol use than in 2016 (relative risk 0.84), but current depression (RR 1.07) and anxiety (RR 0.98) have not fallen, as might have been expected given the relationship with age. We also compare lifetime concepts for test-retest reliability. CONCLUSIONS There are some drawbacks to UKB due to its lack of population representativeness, but where the research question does not depend on this, it offers exceptional resources that any researcher can apply to access. This paper has just scratched the surface of the results from MHQ2 and how this can be combined with other tranches of UKB data, but we predict it will enable many future discoveries about mental health and health in general.
Collapse
Affiliation(s)
- Katrina A.S. Davis
- King’s College London, London, United Kingdom
- South London and Maudsley NHS Trust, London, United Kingdom
| | - Jonathan R.I. Coleman
- King’s College London, London, United Kingdom
- South London and Maudsley NHS Trust, London, United Kingdom
| | - Mark Adams
- University of Edinburgh, Edinburgh, United Kingdom
| | - Gerome Breen
- King’s College London, London, United Kingdom
- South London and Maudsley NHS Trust, London, United Kingdom
| | - Na Cai
- ETH Zurich, Zurich, Switzerland
- Helmholtz Munich, Neuherberg, Germany
- Technical University of Munich, Munich, Germany
| | - Helena L. Davies
- King’s College London, London, United Kingdom
- Copenhagen University Hospital – Mental Health Services CPH, Copenhagen, Denmark
- Mental Health Center Sct. Hans CPH, Copenhagen, Denmark
| | - Kelly Davies
- University of Oxford, Oxford, United Kingdom
- UK Biobank, Stockport, United Kingdom
| | | | - Thalia C. Eley
- King’s College London, London, United Kingdom
- South London and Maudsley NHS Trust, London, United Kingdom
| | - Elaine Fox
- University of Adelaide, Adelaide, Australia
| | - Jo Holliday
- University of Oxford, Oxford, United Kingdom
- UK Biobank, Stockport, United Kingdom
| | - Christopher Hübel
- King’s College London, London, United Kingdom
- Aarhus University, Aarhus, Denmark
- German Red Cross Clinics, Berlin, Germany
| | - Ann John
- Swansea University, Swansea, United Kingdom
- Public Health Wales NHS Trust, Cardiff, United Kingdom
| | | | | | - William Lee
- Cornwall Partnership NHS Foundation Trust, Bodmin, United Kingdom
| | - Danyang Li
- King’s College London, London, United Kingdom
| | - Jared Maina
- King’s College London, London, United Kingdom
| | - Rose McCabe
- City St George’s University of London, London, United Kingdom
| | | | - Sian Oram
- King’s College London, London, United Kingdom
| | | | | | - Fenella Starkey
- University of Oxford, Oxford, United Kingdom
- UK Biobank, Stockport, United Kingdom
| | - Abigail R. ter Kuile
- King’s College London, London, United Kingdom
- University College London, London, United Kingdom
| | | | - Rujia Wang
- King’s College London, London, United Kingdom
| | - Zhaoying Yu
- King’s College London, London, United Kingdom
| | - Johan Zvrskovec
- King’s College London, London, United Kingdom
- South London and Maudsley NHS Trust, London, United Kingdom
| | - Matthew Hotopf
- King’s College London, London, United Kingdom
- South London and Maudsley NHS Trust, London, United Kingdom
| |
Collapse
|
4
|
Chen Z, Zhou Y, Xue C, Zeng L, Deng S, Xu Z, Li M, Zhao H, He X, Liu S, Liu J, Liu S, Zhao S, Zhang S, Peng X, Wu X, Bai R, Zhuang L, Wu S, Zhang J, Lin D, Huang X, Zheng J. Psychological stress-induced ALKBH5 deficiency promotes tumour innervation and pancreatic cancer via extracellular vesicle transfer of RNA. Nat Cell Biol 2025:10.1038/s41556-025-01667-0. [PMID: 40419796 DOI: 10.1038/s41556-025-01667-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 04/02/2025] [Indexed: 05/28/2025]
Abstract
The pathological role and mechanism of psychological stress in cancer progression are little known. Here we show in a mouse model that psychological stress drives pancreatic ductal adenocarcinoma (PDAC) progression by stimulating tumour nerve innervation. We demonstrate that nociception and other stressors activate sympathetic nerves to release noradrenaline, downregulating RNA demethylase alkB homologue 5 (Alkbh5) in tumour cells. Alkbh5 deficiency in these cancer cells causes aberrant N6-methyladenosine (m6A) modification of RNAs, which are packed into extracellular vesicles and delivered to nerves in the tumour microenvironment, enhancing hyperinnervation and PDAC progression. ALKBH5 levels are inversely correlated with tumour innervation and survival time in patients with PDAC. Animal experiments identify a natural flavonoid, fisetin, that prevents neurons from taking in extracellular vesicles containing m6A-modified RNAs, thus suppressing the excessive innervation and progression of PDAC tumours. Our study sheds light on a molecular mechanism by which crosstalk between the neuroendocrine system and cancer cells links psychological stress and cancer progression and raises a potential strategy for PDAC therapy.
Collapse
Affiliation(s)
- Ziming Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yifan Zhou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Chunling Xue
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Lingxing Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shuang Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Zilan Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Mei Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongzhe Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xiaowei He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaoqiu Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ji Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shuang Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Sihan Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaoping Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xinyi Peng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xiaoyu Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Ruihong Bai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Lisha Zhuang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Shaojia Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Jialiang Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Dongxin Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Xudong Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
| | - Jian Zheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Cory-Slechta DA, Downs CJ, Sobolewski M. Cumulative risk assessment as the pathway to public health protection for behavioral neurotoxicity. Neurotoxicology 2025; 108:400-411. [PMID: 40349850 DOI: 10.1016/j.neuro.2025.04.015] [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: 01/05/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
The formulation of adverse outcome pathways (AOPs) based on high-throughput in vitro new approach methods linking biochemical/mechanistic data with an apical endpoint considered an adverse outcome (AO), is increasingly proposed to accelerate the process of risk assessment for environmental chemical exposures. While a laudable goal, this approach ignores the extensive evidence demonstrating context-dependence of neurotoxicological consequences, including behavioral toxicity of chemical exposures. Such contextual modifiers can include environmental conditions (poverty, psychosocial stress, behavioral experience/history), physiological conditions (sex, period of exposure, nutritional status, brain region, exposure parameters), and genetic background. Context dependence represents a serious omission for AOP formulation because an environmental context can alter a chemical's molecular targets, or potentially enhance toxicity through interactions with other contextual conditions, thus leading to potential underestimation of neurological risks due to such exposures. The integrative physiological basis of AOs requires cumulative risk assessments that model environmental contexts across scales of biology, i.e., integration and testing in whole-animal models. AOPs contribute to the derivation of cumulative risk considerations regarding factors to incorporate into cumulative risk assessments by defining risk factors with shared biological targets. Epidemiological and animal model studies can provide information to prioritize interactive effects of greatest magnitude. Additionally, a focus on how a single risk factor in different physiological contexts may attribute risk across multiple neurologic conditions, rather than to a single unique condition, would provide broader public health protection. Realistic acknowledgement of context-dependence is requisite to understanding both the etiological basis of neurological diseases and disorders and to human health protection.
Collapse
Affiliation(s)
- Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States.
| | - Cynthia J Downs
- Department of Environmental Biology, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States.
| |
Collapse
|
6
|
Wang BM, Mills Z, Jones HF, Montgomery JM, Lee KY. Presymptomatic Biological, Structural, and Functional Diagnostic Biomarkers of Autism Spectrum Disorder. J Neurochem 2025; 169:e70088. [PMID: 40390287 PMCID: PMC12089747 DOI: 10.1111/jnc.70088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/21/2025]
Abstract
Autism spectrum disorder (ASD) is a common neurodevelopmental disorder clinically diagnosed by persistent deficits in three areas of social communication and interaction, plus at least two of four types of restricted repetitive behaviors. ASD has been shown to be caused by genetic predisposition and environmental factors; however, the heterogeneity of ASD complicates its diagnosis and treatment. Early behavioral interventions have shown significant benefits, emphasizing the urgent need for reliable diagnostic biomarkers to enhance long-term outcomes. Here we provide a systematic review that outlines current findings on genetic and neurological biomarkers for presymptomatic ASD diagnoses, assessed prior to the observation of behavioral manifestations. Specifically, we offer insights into the mechanisms of presymptomatic neurological, biological, structural, and functional markers for ASD, compare outcomes across studies, and critically assess their limitations and implications. Recent findings highlight genotype-guided therapeutic strategies in animal models, such as dietary zinc supplementation for reversing ASD-associated behaviors by synaptic deficits. However, the differential efficacy based on underlying genotypes, along with challenges in identifying reliable genomic biomarkers prior to symptom onset, indicates the need for further research. Notably, recent advancements in imaging technologies like magnetic resonance imaging, electroencephalography, and pupillometry have shown promising markers in neonates, and at 3 and 9 months old, respectively. Newer developments in magnetoencephalography hardware can facilitate the much-needed infant ASD studies. It is important to note that many of these biomarker findings are preliminary, and further validation for clinical use is required. Continued research is needed to advance the practicality, reliability, and acceptability of these biomarkers to improve ASD diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bonnie M. Wang
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Zoe Mills
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Hannah F. Jones
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Department of NeuroservicesStarship Children's HospitalAucklandNew Zealand
| | - Johanna M. Montgomery
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Kevin Y. Lee
- Department of Physiology, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain Research, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
7
|
Munns S, Brown A, Buckberry S. Type-2 diabetes epigenetic biomarkers: present status and future directions for global and Indigenous health. Front Mol Biosci 2025; 12:1502640. [PMID: 40356723 PMCID: PMC12066322 DOI: 10.3389/fmolb.2025.1502640] [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: 09/27/2024] [Accepted: 02/03/2025] [Indexed: 05/15/2025] Open
Abstract
Type-2 diabetes is a systemic condition with rising global prevalence, disproportionately affecting Indigenous communities worldwide. Recent advances in epigenomics methods, particularly in DNA methylation detection, have enabled the discovery of associations between epigenetic changes and Type-2 diabetes. In this review, we summarise DNA methylation profiling methods, and discuss how these technologies can facilitate the discovery of epigenomic biomarkers for Type-2 diabetes. We critically evaluate previous DNA methylation biomarker studies, particularly those using microarray platforms, and advocate for a shift towards sequencing-based approaches to improve genome-wide coverage. Furthermore, we emphasise the need for biomarker studies that include genetically diverse populations, especially Indigenous communities who are significantly impacted by Type-2 diabetes. We discuss research approaches and ethical considerations that can better facilitate Type-2 diabetes biomarker development to ensure that future genomics-based precision medicine efforts deliver equitable health outcomes. We propose that by addressing these gaps, future research can better capture the genetic and environmental complexities of Type-2 diabetes among populations at disproportionate levels of risk, ultimately leading to more effective diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Sarah Munns
- The Kids Research Institute Australia, Perth, WA, Australia
- National Centre for Indigenous Genomics, Australian National University, Canberra, ACT, Australia
| | - Alex Brown
- The Kids Research Institute Australia, Perth, WA, Australia
- National Centre for Indigenous Genomics, Australian National University, Canberra, ACT, Australia
| | - Sam Buckberry
- The Kids Research Institute Australia, Perth, WA, Australia
- National Centre for Indigenous Genomics, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
8
|
Lin T, He L, Liu S, Li Y, Huang Z, Cai D, Wang Y, Mo Z, Zhu Y, Chen Z, Xu P, Lou X, Sheng J, Chen G, Wang X. Per- and polyfluoroalkyl substances mixture impairs intestinal barrier function through microbiota-derived 21-deoxycortisol and cortisol metabolism dysregulation. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138378. [PMID: 40359747 DOI: 10.1016/j.jhazmat.2025.138378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/28/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are persistent environmental pollutants linked to various health risks, including intestinal disorders. However, the effect of real-world PFASs mixture on intestinal health remains unclear. Therefore, this study aimed to investigate the effects of a PFASs mixture by mimicking the exposure composition in a population on intestinal homeostasis in rats. Although the colon showed no significant morphological alterations, transcriptomic analysis revealed dose-dependent changes in gene expression levels related to cell-cell and tight junctions. Immunofluorescence and immunohistochemistry further confirmed these findings, demonstrating a dose-dependent decrease in key tight junction proteins, occludin and claudin-1, in the colonic epithelium. Integrative analysis revealed that PFASs mixture exposure disrupted the growth and metabolism of gut bacteria, such as Ruminococcus, leading to increased production of 21-deoxycortisol (21-DF). 21-DF inhibited cortisol to cortisone conversion, elevating cortisol levels in intestinal epithelial cells. Consequently, the increased cortisol levels suppressed the expression of tight junction proteins and disrupted the intestinal barrier function. Our findings provide novel insights into the underlying mechanisms by which real-world PFASs mixture disrupt intestinal barrier function through the gut microbiome-metabolome-epithelial cell axis, highlighting the need to consider the complex interplay between environmental pollutants, gut microbiota, and host health in risk assessment and development of intervention strategies.
Collapse
Affiliation(s)
- Tingting Lin
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China; Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Luyang He
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China; School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Sishuo Liu
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China
| | - Yahui Li
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China
| | - Zhi Huang
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China
| | - Delei Cai
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Yiming Wang
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China
| | - Zhe Mo
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Yi Zhu
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China
| | - Zhijian Chen
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Peiwei Xu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Xiaoming Lou
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Jinghao Sheng
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China.
| | - Guangdi Chen
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China.
| | - Xiaofeng Wang
- Institute of Environmental Medicine, Zhejiang University School of Public Health, Hangzhou 310058, China; Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China.
| |
Collapse
|
9
|
Wei S, Tao HY, Duan Z, Wang Y. Environmental Exposure, Epitranscriptomic Perturbations, and Human Diseases. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:6387-6399. [PMID: 40126397 PMCID: PMC11978485 DOI: 10.1021/acs.est.5c00907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Epitranscriptomics is a rapidly evolving field, and it examines how chemical modifications on RNA regulate gene expression. Increasing lines of evidence support that exposure to various environmental agents can change substantially chemical modifications on RNA, thereby perturbing gene expression and contributing to disease development in humans. However, the molecular mechanisms through which environmental exposure impairs RNA modification-associated proteins ("reader", "writer", and "eraser" or RWE proteins) and alters the landscape of RNA modifications remain poorly understood. Here, we provide our perspectives on the current knowledge about how environmental exposure alters the epitranscriptome, where we focus on dynamic changes in RNA modifications and their regulatory proteins elicited by exposure to environmental agents. We discuss how these epitranscriptomic alterations may contribute to the development of human diseases, especially neurodegeneration and cancer. We also discuss the potential and technical challenges of harnessing RNA modifications as biomarkers for monitoring environmental exposure. Finally, we emphasize the need to integrate multiomics approaches to decipher the complex interplay between environmental exposure and the epitranscriptome and offer a forward-looking viewpoint on future research priorities that may inform public health interventions and environmental regulations.
Collapse
Affiliation(s)
- Songbo Wei
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Huan-Yu Tao
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Zheng Duan
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| |
Collapse
|
10
|
Foguet C, Jiang X, Ritchie SC, Persyn E, Xu Y, Ben-Eghan C, Taylor HJ, Di Angelantonio E, Danesh J, Butterworth AS, Lambert SA, Inouye M. Metabolic reaction fluxes as amplifiers and buffers of risk alleles for coronary artery disease. Mol Syst Biol 2025:10.1038/s44320-025-00097-2. [PMID: 40175777 DOI: 10.1038/s44320-025-00097-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
Genome-wide association studies have identified thousands of variants associated with disease risk but the mechanism by which such variants contribute to disease remains largely unknown. Indeed, a major challenge is that variants do not act in isolation but rather in the framework of highly complex biological networks, such as the human metabolic network, which can amplify or buffer the effect of specific risk alleles on disease susceptibility. Here we use genetically predicted reaction fluxes to perform a systematic search for metabolic fluxes acting as buffers or amplifiers of coronary artery disease (CAD) risk alleles. Our analysis identifies 30 risk locus-reaction flux pairs with significant interaction on CAD susceptibility involving 18 individual reaction fluxes and 8 independent risk loci. Notably, many of these reactions are linked to processes with putative roles in the disease such as the metabolism of inflammatory mediators. In summary, this work establishes proof of concept that biochemical reaction fluxes can have non-additive effects with risk alleles and provides novel insights into the interplay between metabolism and genetic variation on disease susceptibility.
Collapse
Affiliation(s)
- Carles Foguet
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
| | - Xilin Jiang
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Elodie Persyn
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Yu Xu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Chief Ben-Eghan
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Henry J Taylor
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Science Research Centre, Fondazione Human Technopole, Milan, Italy
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Department of Human Genetics, the Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
| | - Samuel A Lambert
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Wan M, Simonin EM, Johnson MM, Zhang X, Lin X, Gao P, Patel CJ, Yousuf A, Snyder MP, Hong X, Wang X, Sampath V, Nadeau KC. Exposomics: a review of methodologies, applications, and future directions in molecular medicine. EMBO Mol Med 2025; 17:599-608. [PMID: 39870881 PMCID: PMC11982546 DOI: 10.1038/s44321-025-00191-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/29/2025] Open
Abstract
The exposome is the measure of all the exposures of an individual in a lifetime and how those exposures relate to health. Exposomics is the emerging field of research to measure and study the totality of the exposome. Exposomics can assist with molecular medicine by furthering our understanding of how the exposome influences cellular and molecular processes such as gene expression, epigenetic modifications, metabolic pathways, and immune responses. These molecular alterations can aid as biomarkers for the diagnosis, disease prediction, early detection, and treatment and offering new avenues for personalized medicine. Advances in high throughput omics and other technologies as well as increased computational analytics is enabling comprehensive measurement and sophisticated analysis of the exposome to elucidate their cumulative and combined impacts on health, which can enable individuals, communities, and policymakers to create programs, policies, and protections that promote healthier environments and people. This review provides an overview of the potential role of exposomics in molecular medicine, covering its history, methodologies, current research and applications, and future directions.
Collapse
Grants
- UM1 AI109565 NIAID NIH HHS
- R21 AI149277 NIAID NIH HHS
- R01 HL141851 NHLBI NIH HHS
- R01 AI125567 NIAID NIH HHS
- P01 HL152953 NHLBI NIH HHS
- P01 HL152953,R01 HL141851 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 ES032253 NIEHS NIH HHS
- U01 AI140498 NIAID NIH HHS
- R21AI1492771,R21EB030643,U01AI140498,U01 AI147462,R01AI140134,UM1AI109565,UM2AI130836,P01AI15 HHS | NIH | National Institute of Allergy and Infectious Diseases (NIAID)
- R21 EB030643 NIBIB NIH HHS
- P01 AI153559 NIAID NIH HHS
- R01 AI140134 NIAID NIH HHS
- R21ES03304901,R01ES032253 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- U19 AI167903 NIAID NIH HHS
- UM2 AI130836 NIAID NIH HHS
- U01 AI147462 NIAID NIH HHS
Collapse
Affiliation(s)
- Melissa Wan
- Harvard Chan Occupational and Environmental Medicine, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Elisabeth M Simonin
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Mary Margaret Johnson
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Xinyue Zhang
- Cardiovascular Institute Operations, Stanford University, Palo Alto, CA, 94305, USA
| | - Xiangping Lin
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Peng Gao
- School of Public Health, University of Pittsburg, Pittsburgh, PA, 15261, USA
| | | | | | - Michael P Snyder
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Xiumei Hong
- Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health, John Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xiaobin Wang
- Center on Early Life Origins of Disease, Department of Population, Family and Reproductive Health, John Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Vanitha Sampath
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
12
|
Cai Y, Zeng X, Wu M, Chen H, Sun M, Yang H. TET1 mitigates prenatal fluoride-induced cognition impairment by modulating Bcl2 DNA hydroxymethylation level. Mol Med 2025; 31:117. [PMID: 40133886 PMCID: PMC11938627 DOI: 10.1186/s10020-025-01174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Fluoride exposure during pregnancy commonly compromises fetal neurodevelopment and largely results in a broad spectrum of cognitive deficiencies in the adult offspring. However, the precise mechanisms underlying these effects remain to be fully elucidated. Herein, we investigate the impacts of fluoride on neural excitability and apoptosis, synaptic plasticity, and cognitive function, as well as possible underlying mechanisms. Our results indicated that exposure to a high sodium fluoride (100 mg/L) during pregnancy in the mouse can cause the cognitive deficits of their offspring, accompanied by a decrease in the expression of Tet-eleven translocation protein 1 (TET1), an enzyme responsible for DNA hydroxymethylation. Additionally, there is a reduction in the dendritic spine density and the expression of postsynaptic density protein-95 (PSD95) in the hippocampal regions of male offspring. Furthermore, in vitro fluoride treatment significantly exacerbates neuronal apoptosis and reduces the frequency of spikes in spontaneous action potential. More significantly, we also found that TET1 could directly bind to the promotor region of Bcl2, altering its DNA hydroxymethylation and Bcl2 expression. Intriguingly, Tet1 knock-out mice exhibited cognitive deficits similar to those observed in male animals exposed to high levels of fluoride. Furthermore, the down-regulation of TET1 protein, along with the consequent alteration in Bcl2 hydroxymethylation and increased neuronal apoptosis, are likely mechanisms underlying the impact of prenatal fluoride exposure on the neurodevelopment of male offspring. These findings provide novel insights into the molecular mechanisms by which fluoride exposure induces neurodevelopmental impairment of the male offspring.
Collapse
Affiliation(s)
- Yongle Cai
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xingdong Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Mengyan Wu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Haonan Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
13
|
Carvajal V, Jorques Molla JV, Luo Y, Zhao Y, Moncunill G, Gascon M. Air pollution and systemic immune biomarkers in early life: A systematic review. ENVIRONMENTAL RESEARCH 2025; 269:120838. [PMID: 39832545 DOI: 10.1016/j.envres.2025.120838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Children's rapid development and immature immune systems place them at a higher risk of adverse health outcomes associated with air pollution exposure. However, the specific mechanisms in which air pollution mediates immune dysregulation in youth are poorly understood. Thus, we aimed to systematically review the available epidemiological evidence surrounding the effects of indoor and ambient air pollution exposure on systemic immune biomarkers in early life (from birth to 18 years old). METHODS based on PRISMA guidelines, we developed a systematic search strategy and defined inclusion and exclusion criteria to retrieve publications from PubMed, SCOPUS and Web of Science published up to August 10th, 2024. Quality assessment and evidence evaluation were also performed. Five independent reviewers participated in the process. RESULTS In total, 96 studies were included. We found limited evidence of a causal relationship between prenatal ambient PM2.5 and reduced T-cells (CD3+ and CD8+), as well as between postnatal PM exposure and increased IgE levels or allergic sensitization. For the rest of exposure-outcome combinations we classified the evidence as inadequate, mainly due to the limited number of studies available or the lack of consistency in the results obtained among them. This was particularly the case for indoor air pollution research, for which only 12 studies were available. CONCLUSION the present systematic review highlights the need for further research on the impacts of air pollution on youth's immune system. We provided recommendations for future studies in order to better understand the early subclinical and clinical effects of air pollution and the underlying biological pathways, and identify the dynamics of the innate and adaptive immune responses to environmental threats. Considering the significance of childhood immunity on health outcomes within all stages of life, and the globally extensive burden of air pollution exposure, further research on this topic should be prioritized.
Collapse
Affiliation(s)
- Veronica Carvajal
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Juan Vicente Jorques Molla
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain; Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Yana Luo
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Yu Zhao
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Gemma Moncunill
- ISGlobal, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Mireia Gascon
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Spain; Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Manresa, Spain.
| |
Collapse
|
14
|
Li B, Ming H, Qin S, Nice EC, Dong J, Du Z, Huang C. Redox regulation: mechanisms, biology and therapeutic targets in diseases. Signal Transduct Target Ther 2025; 10:72. [PMID: 40050273 PMCID: PMC11885647 DOI: 10.1038/s41392-024-02095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/21/2024] [Indexed: 03/09/2025] Open
Abstract
Redox signaling acts as a critical mediator in the dynamic interactions between organisms and their external environment, profoundly influencing both the onset and progression of various diseases. Under physiological conditions, oxidative free radicals generated by the mitochondrial oxidative respiratory chain, endoplasmic reticulum, and NADPH oxidases can be effectively neutralized by NRF2-mediated antioxidant responses. These responses elevate the synthesis of superoxide dismutase (SOD), catalase, as well as key molecules like nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby maintaining cellular redox homeostasis. Disruption of this finely tuned equilibrium is closely linked to the pathogenesis of a wide range of diseases. Recent advances have broadened our understanding of the molecular mechanisms underpinning this dysregulation, highlighting the pivotal roles of genomic instability, epigenetic modifications, protein degradation, and metabolic reprogramming. These findings provide a foundation for exploring redox regulation as a mechanistic basis for improving therapeutic strategies. While antioxidant-based therapies have shown early promise in conditions where oxidative stress plays a primary pathological role, their efficacy in diseases characterized by complex, multifactorial etiologies remains controversial. A deeper, context-specific understanding of redox signaling, particularly the roles of redox-sensitive proteins, is critical for designing targeted therapies aimed at re-establishing redox balance. Emerging small molecule inhibitors that target specific cysteine residues in redox-sensitive proteins have demonstrated promising preclinical outcomes, setting the stage for forthcoming clinical trials. In this review, we summarize our current understanding of the intricate relationship between oxidative stress and disease pathogenesis and also discuss how these insights can be leveraged to optimize therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Siyuan Qin
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jingsi Dong
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhongyan Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China.
| |
Collapse
|
15
|
Zhang J, Yang Y, Shang M, Guo L, Zhang D, Du L, for the Alzheimer’s Disease Neuroimaging Initiative. Mutual-assistance learning for trustworthy biomarker discovery and disease prediction. Brief Bioinform 2025; 26:bbaf178. [PMID: 40254831 PMCID: PMC12009715 DOI: 10.1093/bib/bbaf178] [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: 11/26/2024] [Revised: 01/23/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Integrating and analyzing multiple omics datasets, such as genomics, environmental influences, and imaging endophenotypes, has yielded an abundance of candidate biomarkers. However, translating such findings into beneficial clinical knowledge for disease prediction remains challenging. This becomes even more challenging when studying interpretable high-order feature interactions such as gene-environment interaction (G$\times $E) to understand the etiology. To fill this gap, we draw on the idea of mutual-assistance (MA) learning and accordingly propose a fresh and powerful scheme, referred to as mutual-assistance causal biomarker discovery and stable disease prediction approach (MA-CBxDP). Specifically, we design an interpretable bi-directional mapping framework, integrated with a causal feature interaction module, to extract co-expression patterns across different modalities and identify trustworthy biomarkers including G$\times $E. A cooperative prediction module is further incorporated to ensure accurate diagnosis and identification of causal effects for pathogenesis. Importantly, biomarker discovery and disease prediction can mutually reinforce each other, helping to provide novel insights into chronic diseases. Furthermore, in light of the large computational burden incurred by the high-dimensional interactions, we devise a rapid strategy and extend it to a more practical but challenging chromosome-wide setting. We conduct extensive experiments on two databases under three tasks, i.e. multimodal correlation, disease diagnosis, and trait prediction. MA-CBxDP establishes new state-of-the-art results in predicting clinical scores and disease status classification, while maintaining exceptional interpretability, verifying its flexibility and versatility in practical applications.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Yan Yang
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Muheng Shang
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Lei Guo
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | - Daoqiang Zhang
- School of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Jiangning District, 210000 Nanjing, China
| | - Lei Du
- Department of Intelligent Science and Technology, Northwestern Polytechnical University, 127 Youyi Road, 710072 Shaanxi, China
| | | |
Collapse
|
16
|
Stepanyan A, Arakelyan A, Schug J. Transcriptome alterations in long-term mining region residents: Insights into immune response and molecular pathways. ENVIRONMENT INTERNATIONAL 2025; 197:109344. [PMID: 40023890 DOI: 10.1016/j.envint.2025.109344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Pollution with metals and metalloids is a global problem that adversely affects human health and environment. Although several studies have reported gene expression changes in response to human exposures to metals, there are a limited number of studies exploring the effect of long-term residence in mining areas. The evidence of increased levels of several essential and non-essential metals in soil, water, and plants in Kapan mining area (Armenia) has been previously demonstrated in several environmental studies. Our study investigated the impact of long-term residence in this mining area on the transcriptome state of human peripheral blood mononuclear cells and the possible association of transcriptome changes with the blood metallome. In total, 58 participants including 27 mining region residents (MRR) and 31 non-mining region residents (NMR) were selected for our study. Transcriptomic analysis of peripheral blood mononuclear cells was performed by mRNA sequencing. Differential expression analyses were conducted using generalized linear modeling, optimized for participant demographics, cell types, and sequencing technical factors, followed by pathway analysis. The study revealed that long-term residence in a mining area is correlated with alterations in the blood transcriptome, with responses varying by sex. The identified transcriptome changes were enriched for pathways related to immune response and RNA translation. These changes correlated with higher blood levels of a mixture of non-essential metals, including arsenic, antimony, nickel, thallium, and beryllium. Additionally, the study identified differences in the transcriptome response between male and female MRR. While females exhibited a stronger immune response, males show dysregulation in ion transport and epigenetic modifications. Our findings contribute to understanding the effects of long-term residence in mining regions and can aid in developing more effective risk assessment and mitigation approaches in target populations.
Collapse
Affiliation(s)
- Ani Stepanyan
- The Institute of Molecular Biology of the National Academy of Sciences of the Republic of Armenia, 7 Ezras Hasratyan St, Yerevan 0014, the Republic of Armenia.
| | - Arsen Arakelyan
- The Institute of Molecular Biology of the National Academy of Sciences of the Republic of Armenia, 7 Ezras Hasratyan St, Yerevan 0014, the Republic of Armenia.
| | - Jonathan Schug
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard Building 421, Philadelphia, PA 19104, the United States of America.
| |
Collapse
|
17
|
Peedicayil J, Santhosh S. Histone monoaminylation is a novel epigenetic mechanism in psychiatric disorders. Front Mol Neurosci 2025; 18:1534569. [PMID: 40041043 PMCID: PMC11876413 DOI: 10.3389/fnmol.2025.1534569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Affiliation(s)
- Jacob Peedicayil
- Department of Pharmacology and Clinical Pharmacology, Christian Medical College, Vellore, India
| | | |
Collapse
|
18
|
Santamarina-Ojeda P, Fernández AF, Fraga MF. Epitranscriptomics in the Glioma Context: A Brief Overview. Cancers (Basel) 2025; 17:578. [PMID: 40002173 PMCID: PMC11853273 DOI: 10.3390/cancers17040578] [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: 12/24/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Epitranscriptomics, the study of chemical modifications in RNA, has emerged as a crucial field in cellular regulation, adding another layer to the established landscape of DNA- and histone-based epigenetics. A wide range of RNA modifications, including N6-methyladenosine, pseudouridine, and inosine, have been identified across nearly all RNA species, influencing essential processes such as transcription, splicing, RNA stability, and translation. In the context of brain tumors, particularly gliomas, specific epitranscriptomic signatures have been reported to play a role in tumorigenesis. Despite growing evidence, the biological implications of various RNA modifications remain poorly understood. This review offers an examination of the main RNA modifications, the interplay between modified and unmodified molecules, how they could contribute to glioma-like phenotypes, and the therapeutic impact of targeting these mechanisms.
Collapse
Affiliation(s)
- Pablo Santamarina-Ojeda
- Foundation for Biomedical Research and Innovation in Asturias (FINBA), 33011 Oviedo, Spain; (P.S.-O.); (A.F.F.)
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
- Nanomaterials and Nanotechnology Research Centre (CINN-CSIC), 33940 El Entrego, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| | - Agustín F. Fernández
- Foundation for Biomedical Research and Innovation in Asturias (FINBA), 33011 Oviedo, Spain; (P.S.-O.); (A.F.F.)
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
- Nanomaterials and Nanotechnology Research Centre (CINN-CSIC), 33940 El Entrego, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| | - Mario F. Fraga
- Foundation for Biomedical Research and Innovation in Asturias (FINBA), 33011 Oviedo, Spain; (P.S.-O.); (A.F.F.)
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
- Nanomaterials and Nanotechnology Research Centre (CINN-CSIC), 33940 El Entrego, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| |
Collapse
|
19
|
Choza JI, Virani M, Kuhn NC, Adams M, Kochmanski J, Bernstein AI. Parkinson's disease-associated alterations in DNA methylation and hydroxymethylation in human brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.21.595193. [PMID: 39975085 PMCID: PMC11838189 DOI: 10.1101/2024.05.21.595193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Epigenetic mechanisms are mediators of interactions between aging, genetics, and environmental factors in sporadic Parkinson's disease (PD). Multiple studies have explored the DNA modifications in PD, but few focus on 5-hydroxymethylcytosine (5-hmC), which is important in the central nervous system and sensitive to environmental exposures. To date, studies have not differentiated between 5-methylcytosine (5-mC) and 5-hmC or have analyzed them separately. In this study, we modeled paired 5-mC and 5-hmC data simultaneously. We identified 108 cytosines with significant PD-associated shifts between these marks in an enriched neuronal population from PD postmortem parietal cortex, within 83 genes and 34 enhancers associated with 67 genes. These data potentially link epigenetic regulation of genes related to LRRK2 and endolysosomal sort (RAB32 and AGAP1), and genes involved in neuroinflammation, the inflammasome, and neurodevelopment with early changes in PD and suggest that there are significant shifts between 5mC and 5hmC associated with PD in genes not captured by standard methods.
Collapse
Affiliation(s)
- Juliana I Choza
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
| | - Mahek Virani
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI
| | - Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Alison I Bernstein
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| |
Collapse
|
20
|
Nie T, Li J, You L, Wu Q. Environmental mycotoxins: A potential etiological factor for neurodegenerative diseases? Toxicology 2025; 511:154056. [PMID: 39814257 DOI: 10.1016/j.tox.2025.154056] [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: 12/11/2024] [Revised: 01/06/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Mycotoxins are potential environmental risk factors for neurodegenerative diseases. These toxins penetrate the central nervous system via a compromised blood-brain barrier, which may cause oxidative stress and neuroinflammation, these can also contribute to amyloid-beta (Aβ) plaque accumulation, Tau protein hyperphosphorylation, and neurofibrillary tangle formation. Mycotoxins also activate microglia, cause neuronal apoptosis, and disrupt central nervous system function. This study examines the evidence linking mycotoxin exposure to neurodegenerative disorders like Alzheimer's and Parkinson's diseases. We explore mechanisms such as oxidative stress, mitochondrial dysfunction, blood-brain barrier disruption, neuroinflammation, and direct neurotoxic effects. Epidemiological studies show regional variations in mycotoxin prevalence and corresponding neurodegenerative disease incidences, supporting this association. We also review current approaches to mitigate mycotoxin exposure and discuss the challenges and opportunities in developing strategies to prevent or slow neurodegenerative disease progression. This work highlights the need for increased awareness and research on mycotoxins as modifiable risk factors in neurological health.
Collapse
Affiliation(s)
- Tong Nie
- College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Jiefeng Li
- College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Li You
- College of Physical Education and Health, Chongqing College of International Business and Economics, Chongqing 401520, China
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou 434025, China.
| |
Collapse
|
21
|
Tommasi S, Brocchieri L, Tornaletti S, Besaratinia A. Epigenomic Dysregulation in Youth Vapers: Implications for Disease Risk Assessment. Am J Respir Cell Mol Biol 2025; 72:206-218. [PMID: 39133188 PMCID: PMC11976656 DOI: 10.1165/rcmb.2024-0207oc] [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: 05/01/2024] [Accepted: 08/12/2024] [Indexed: 08/13/2024] Open
Abstract
Despite the ongoing epidemic of youth vaping, the long-term health consequences of electronic cigarette use are largely unknown. We report the effects of vaping versus smoking on the oral cell methylome of healthy young vapers and smokers relative to nonusers. Whereas vapers and smokers differ in the number of differentially methylated regions (DMRs) (831 vs. 2,863), they share striking similarities in the distribution and patterns of DNA methylation, chromatin states, transcription factor binding motifs, and pathways. There is substantial overlap in DMR-associated genes between vapers and smokers, with the shared subset of genes enriched for transcriptional regulation, signaling, tobacco use disorders, and cancer-related pathways. Of significance is the identification of a common hypermethylated DMR at the promoter of HIC1 (hypermethylated in cancer 1), a tumor suppressor gene frequently silenced in smoking-related cancers. Our data support a potential link between epigenomic dysregulation in youth vapers and disease risk. These novel findings have significant implications for public health and tobacco product regulation.
Collapse
Affiliation(s)
- Stella Tommasi
- Department of Population and Public Health Sciences, Keck School of Medicine, and
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California; and
| | | | | | - Ahmad Besaratinia
- Department of Population and Public Health Sciences, Keck School of Medicine, and
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California; and
| |
Collapse
|
22
|
Ainsworth HC, Baker Frost D, Lim SS, Ramos PS. Breaking research silos to achieve equitable precision medicine in rheumatology. Nat Rev Rheumatol 2025; 21:98-110. [PMID: 39794514 PMCID: PMC11910143 DOI: 10.1038/s41584-024-01204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/13/2025]
Abstract
Health disparities in rheumatic disease are well established and urgently need addressing. Obstacles to precision medicine equity span both the clinical and the research domains, with a focus placed on structural barriers limiting equitable health care access and inclusivity in research. Less articulated factors include the use of inaccurate population descriptors and the existence of research silos in rheumatology research, which creates a knowledge gap that precludes addressing the health disparities and fulfilling the goals of precision medicine to understand the 'full patient'. The biopsychosocial model is a research framework that intertwines layers of biological and environmental effects to understand disease. However, very limited rheumatology research bridges across molecular and epidemiological studies of environmental exposures, such as physical and social determinants of health. In this Review, we discuss clinical obstacles to health care equity, including access to health care and the use of inaccurate language when labelling population groups. We explore the goals and data needed for research under the biopsychosocial model. We describe results from a rheumatic disease literature search that highlights the paucity of studies investigating the molecular influences of systemic exposures. We conclude with a list of considerations and recommendations to help achieve equitable precision medicine.
Collapse
Affiliation(s)
- Hannah C Ainsworth
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Wake Forest Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - DeAnna Baker Frost
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| | - S Sam Lim
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, GA, USA
| | - Paula S Ramos
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
23
|
Lauri G, Mills K, Majumder S, Capurso G. The exposome as a target for primary prevention and a tool for early detection of pancreatic cancer. Best Pract Res Clin Gastroenterol 2025; 74:101991. [PMID: 40210335 DOI: 10.1016/j.bpg.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 04/12/2025]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a highly aggressive malignancy with limited survival due to late stage diagnosis and scarce therapeutic options. Emerging evidence highlights the role of the "exposome," which encompasses environmental, lifestyle, and metabolic exposures, as a crucial determinant in PDAC risk and a potential avenue for early detection. This review synthesizes findings on modifiable risk factors, including smoking, obesity, diabetes, diet, and alcohol consumption, and their interplay with genetic and metabolic profiles in PDAC development. Additionally, we explore cutting-edge approaches in exposomic research, such as biobanking, electronic health record analysis, and AI-driven predictive models, to identify early biomarkers and stratify high-risk populations. This integrated framework aims to inform prevention strategies and improve early detection of PDAC.
Collapse
Affiliation(s)
- Gaetano Lauri
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Krystal Mills
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gabriele Capurso
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
24
|
Alemu R, Sharew NT, Arsano YY, Ahmed M, Tekola-Ayele F, Mersha TB, Amare AT. Multi-omics approaches for understanding gene-environment interactions in noncommunicable diseases: techniques, translation, and equity issues. Hum Genomics 2025; 19:8. [PMID: 39891174 PMCID: PMC11786457 DOI: 10.1186/s40246-025-00718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Non-communicable diseases (NCDs) such as cardiovascular diseases, chronic respiratory diseases, cancers, diabetes, and mental health disorders pose a significant global health challenge, accounting for the majority of fatalities and disability-adjusted life years worldwide. These diseases arise from the complex interactions between genetic, behavioral, and environmental factors, necessitating a thorough understanding of these dynamics to identify effective diagnostic strategies and interventions. Although recent advances in multi-omics technologies have greatly enhanced our ability to explore these interactions, several challenges remain. These challenges include the inherent complexity and heterogeneity of multi-omic datasets, limitations in analytical approaches, and severe underrepresentation of non-European genetic ancestries in most omics datasets, which restricts the generalizability of findings and exacerbates health disparities. This scoping review evaluates the global landscape of multi-omics data related to NCDs from 2000 to 2024, focusing on recent advancements in multi-omics data integration, translational applications, and equity considerations. We highlight the need for standardized protocols, harmonized data-sharing policies, and advanced approaches such as artificial intelligence/machine learning to integrate multi-omics data and study gene-environment interactions. We also explore challenges and opportunities in translating insights from gene-environment (GxE) research into precision medicine strategies. We underscore the potential of global multi-omics research in advancing our understanding of NCDs and enhancing patient outcomes across diverse and underserved populations, emphasizing the need for equity and fairness-centered research and strategic investments to build local capacities in underrepresented populations and regions.
Collapse
Affiliation(s)
- Robel Alemu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Anderson School of Management, University of California Los Angeles, Los Angeles, CA, USA.
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| | - Nigussie T Sharew
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Yodit Y Arsano
- Alpert Medical School, Lifespan Health Systems, Brown University, WarrenProvidence, Rhode Island, USA
| | - Muktar Ahmed
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tesfaye B Mersha
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Azmeraw T Amare
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
25
|
Gong FH, Liu L, Wang X, Xiang Q, Yi X, Jiang DS. Ferroptosis induced by environmental pollutants and its health implications. Cell Death Discov 2025; 11:20. [PMID: 39856053 PMCID: PMC11759704 DOI: 10.1038/s41420-025-02305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/19/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Environmental pollution represents a significant public health concern, with the potential health risks associated with environmental pollutants receiving considerable attention over an extended period. In recent years, a substantial body of research has been dedicated to this topic. Since the discovery of ferroptosis, an iron-dependent programmed cell death typically characterized by lipid peroxidation, in 2012, there have been significant advances in the study of its role and mechanism in various diseases. A growing number of recent studies have also demonstrated the involvement of ferroptosis in the damage caused to the organism by environmental pollutants, and the molecular mechanisms involved have been partially elucidated. The targeting of ferroptosis has been demonstrated to be an effective means of ameliorating the health damage caused by PM2.5, organic and inorganic pollutants, and ionizing radiation. This review begins by providing a summary of the most recent and important advances in ferroptosis. It then proceeds to offer a critical analysis of the health effects and molecular mechanisms of ferroptosis induced by various environmental pollutants. Furthermore, as is the case with all rapidly evolving research areas, there are numerous unanswered questions and challenges pertaining to environmental pollutant-induced ferroptosis, which we discuss in this review in an attempt to provide some directions and clues for future research in this field.
Collapse
Affiliation(s)
- Fu-Han Gong
- Department of Cardiology, Tongren People's Hospital, Tongren, Guizhou, China
| | - Liyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuesheng Wang
- Department of Cardiology, Tongren People's Hospital, Tongren, Guizhou, China
| | - Qi Xiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
26
|
Liu X, Liu H, Nie H, Tian L, Shi Y, Lai W, Xi Z, Lin B. Oil mist particulate matter induces myocardial tissue injury by impairing fatty acid metabolism and mitochondrial bioenergetics function via inhibiting the PPAR alpha signaling pathway in rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 365:125340. [PMID: 39581367 DOI: 10.1016/j.envpol.2024.125340] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/10/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
Air pollution is a significant concern for human health, particularly in relation to cardiovascular damage. Currently, the precise mechanisms underlying myocardial tissue injury induced by air pollution remain to be fully elucidated. Oil mist particulate matter (OMPM) is a key environmental factor that has been linked to increased mortality from cardiovascular diseases. The research aims to explore the detrimental effects and underlying molecular mechanisms of OMPM exposure on myocardial tissue. In this study, we established exposure models with different concentrations of OMPM both in vivo and in vitro to assess their deleterious effects on myocardial tissue. The results indicated that OMPM exposure induced alterations in myocardial enzymes and large accumulation of lipid droplets in rat myocardial tissue, with a dose-dependent increase in cell apoptosis, oxidative stress, and inflammatory responses, accompanied by mitochondrial structural damage and dysfunction. Proteomic analysis suggested that OMPM induced myocardial tissue damage is closely associated with changes in mitochondrial biological functions and fatty acid metabolism, possibly through inhibition of the PPAR signaling pathway. Further experiments using a PPARα agonist (WY-14643) and PPARα siRNA transfection cell model demonstrated that WY-14643 could mitigate abnormal fatty acid metabolism, mitochondrial dysfunction, and cell apoptosis caused by OMPM exposure. Overall, the study suggests that OMPM exposure disrupts myocardial fatty acid metabolism, contributes to mitochondrial damage and dysfunction through targeted inhibition of the PPAR signaling pathway, and ultimately results in cardiomyocyte apoptosis and myocardial tissue injury.
Collapse
Affiliation(s)
- Xuan Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Huanliang Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Huipeng Nie
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Lei Tian
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yue Shi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Wenqing Lai
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Zhuge Xi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| | - Bencheng Lin
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| |
Collapse
|
27
|
Shen X, Zhang Y, Li J, Zhou Y, Butensky S, Zhang Y, Cai Z, DeWan AT, Khan SA, Yan H, Johnson CH, Zhu F. OncoSexome: the landscape of sex-based differences in oncologic diseases. Nucleic Acids Res 2025; 53:D1443-D1459. [PMID: 39535034 PMCID: PMC11701605 DOI: 10.1093/nar/gkae1003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/28/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The NIH policy on sex as biological variable (SABV) emphasized the importance of sex-based differences in precision oncology. Over 50% of clinically actionable oncology genes are sex-biased, indicating differences in drug efficacy. Research has identified sex differences in non-reproductive cancers, highlighting the need for comprehensive sex-based cancer data. We therefore developed OncoSexome, a multidimensional knowledge base describing sex-based differences in cancer (https://idrblab.org/OncoSexome/) across four key topics: antineoplastic drugs and responses (SDR), oncology-related biomarkers (SBM), risk factors (SRF) and microbial landscape (SML). SDR covers sex-based differences in 2051 anticancer drugs; SBM describes 12 551 sex-differential biomarkers; SRF illustrates 350 sex-dependent risk factors; SML demonstrates 1386 microbes with sex-differential abundances associated with cancer development. OncoSexome is unique in illuminating multifaceted influences of biological sex on cancer, providing both external and endogenous contributors to cancer development and describing sex-based differences for the broadest oncological classes. Given the increasing global research interest in sex-based differences, OncoSexome is expected to impact future precision oncology practices significantly.
Collapse
Affiliation(s)
- Xinyi Shen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Jiamin Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | | | - Yechi Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
- School of Public Health, Zhejiang University, Hangzhou 310058, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Sajid A Khan
- Yale School of Medicine, Yale University, New Haven 06510, USA
- Division of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven 06510, USA
| | - Hong Yan
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
28
|
Wu Y, Xu R, Li S, Wen B, Southey MC, Dugue PA, Hopper JL, Abramson MJ, Li S, Guo Y. Association between wildfire-related PM 2.5 and epigenetic aging: A twin and family study in Australia. JOURNAL OF HAZARDOUS MATERIALS 2025; 481:136486. [PMID: 39566450 DOI: 10.1016/j.jhazmat.2024.136486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/30/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Wildfire-related PM2.5 has been associated with various adverse health outcomes, but its association with epigenetic aging remains unclear. This study examined the association between wildfire-related PM2.5 exposure and epigenetic aging using DNA methylation data from a twin and family study. With a within-sibship analysis, we found that each 1 µg/m3 increase in annual wildfire-related PM2.5 was associated with a 0.25-year (95 % CI: 0.04, 0.47) increase in GrimAge1 acceleration and a 0.36-year (95 % CI: 0.12, 0.59) increase in GrimAge2 acceleration. Subgroup analyses found that participants aged ≥ 60 years, those with a history of current or former smoking and alcohol consumption, and those living in rural regions exhibited more pronounced epigenetic age acceleration. These findings suggest that wildfire smoke could accelerate biological aging, particularly in vulnerable populations, posing a significant challenge to healthy aging.
Collapse
Affiliation(s)
- Yao Wu
- Climate, Air Quality Research (CARE) Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, VIC, Australia
| | - Rongbin Xu
- Climate, Air Quality Research (CARE) Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, VIC, Australia
| | - Shanshan Li
- Climate, Air Quality Research (CARE) Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, VIC, Australia
| | - Bo Wen
- Climate, Air Quality Research (CARE) Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, VIC, Australia
| | - Melissa C Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton 3800, VIC, Australia; Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne 3010, VIC, Australia; Cancer Epidemiology Division, Cancer Council Victoria, 3004 VIC, Australia
| | - Pierre-Antoine Dugue
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton 3800, VIC, Australia; Cancer Epidemiology Division, Cancer Council Victoria, 3004 VIC, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne 3010, VIC, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne 3010, VIC, Australia
| | - Michael J Abramson
- School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, VIC, Australia
| | - Shuai Li
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton 3800, VIC, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne 3010, VIC, Australia; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, United Kingdom; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3052, VIC, Australia.
| | - Yuming Guo
- Climate, Air Quality Research (CARE) Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, VIC, Australia.
| |
Collapse
|
29
|
Mao X, Ahmad B, Hussain S, Azeem F, Waseem M, Alhaj Hamoud Y, Shaghaleh H, Abeed AHA, Rizwan M, Yong JWH. Microbial assisted alleviation of nickel toxicity in plants: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117669. [PMID: 39788037 DOI: 10.1016/j.ecoenv.2025.117669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/16/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025]
Abstract
Nickel (Ni) is required in trace amounts (less than 500 µg kg-1) in plants to regulate metabolic processes, the immune system, and to act as an enzymatic catalytic cofactor. Conversely, when nickel is present in high concentration, it is considered as a toxic substance. Excessive human nickel exposure occurs through ingestion, inhalation, and skin contact, ultimately leading to respiratory, cardiovascular, and chronic kidney diseases. Due to anthropogenic activities, the nickel concentrations in various environmental scenarios have progressively risen to levels as high as 26,000 ppm in soil and 0.2 mg L-1 in water; surpassing the established safety threshold limits of 100 ppm for soil and 0.005 ppm for surface water. Nickel is required by various plant species for facilitating biological processes; in the range of 0.01-5 µg g-1 (dry weight). When present in excess, nickel toxicity in plants (10-1000 mg kg-1 dry weight mass) causes many disrupted metabolic processes; leading to lower growth, altered development, hindered seed germination, chlorosis, and necrosis. To tackle any metal-linked pollution issues, various remediation approaches are employed to remove heavy metals (especially nickel) and metalloids including physicochemical, and biological methods. Based on literature, the physicochemical methods are not commonly used due to their costly nature and the potential for producing secondary pollutants. Interestingly, bioremediation is considered by many practitioners as an easy-to-handle, efficient, and cost-effective approach, encompassing techniques such as phytoremediation, bioleaching, bioreactors, green landforming, and bio-augmentation. Operationally, phytoremediation is widely utilized for cleaning up contaminated sites. To support the phytoremediative processes, numerous nickel hyperaccumulating plants have been identified; these species can absorb from their surroundings and store high concentrations of nickel (through various mechanisms) in their biomass, thereby helping to detoxify nickel-contaminated soils via phytoextraction. The microbe-assisted phytoremediation further optimizes the nickel detoxification processes by fostering beneficial interactions between microbes and the nickel-hyperaccumulators; promoting enhanced metal uptake, transformation, and sequestration. Microbe-assisted phytoremediation can be categorized into four subtypes: bacterial-assisted phytoremediation, cyanoremediation, mycorrhizal-assisted remediation, and rhizoremediation. These diverse approaches are likely to offer more effective and sustainable remediative strategy to ecologically restore the nickel-contaminated environments.
Collapse
Affiliation(s)
- Xinyu Mao
- College of Agricultural Science and Engineering, Hohai University, Nanjing 210098, China
| | - Bilal Ahmad
- Molecular, Cellular, and Developmental Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Sabir Hussain
- Department of Environmental Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Farrukh Azeem
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Waseem
- Department of Environmental Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Yousef Alhaj Hamoud
- College of Hydrology and Water Resources, Hohai University, Nanjing 210098, China
| | - Hiba Shaghaleh
- College of Environment, Hohai University, Nanjing 210098, China
| | - Amany H A Abeed
- Department of Botany and Microbiology, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Muhammad Rizwan
- Department of Environmental Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan.
| | - Jean Wan Hong Yong
- Department of Biosystems and Technology, Swedish University of Agricultural Sciences, Alnarp 23456, Sweden.
| |
Collapse
|
30
|
Ortega-García JA. Unraveling the exposome: towards a model of planetary health. An Pediatr (Barc) 2025; 102:503695. [PMID: 39741046 DOI: 10.1016/j.anpede.2024.503695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 01/02/2025] Open
Affiliation(s)
- Juan Antonio Ortega-García
- Comité de Salud Medioambiental, Global Alliance for Rewilding Child and Adolescent Health, Asociación Española de Pediatría, Madrid, Spain; Pediatric Environmental Health Speciality Unit (PEHSU), Hospital Clínico Universitario Virgen de la Arrixaca, Universidad de Murcia. Murcia, Spain.
| |
Collapse
|
31
|
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
|
32
|
Jo Y, Lim E, Park J, Kang K, Shin MY, Choi JW, Kim S, Lee J. Epigenetic dysregulation of H19/IGF2 in hepatic cells exposed to toxic metal mixtures in vitro. Sci Rep 2024; 14:29413. [PMID: 39592715 PMCID: PMC11599747 DOI: 10.1038/s41598-024-80142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Exposure to mixtures of toxic metals is known to cause adverse health effects through epigenetic alterations. Here we aimed to examine the unexplored area of aberrant DNA methylation in the H19/IGF2 domain following combined toxic metal exposure. An in vitro epigenotoxicity assay using the human normal liver epithelial cell line THLE-3 was conducted. When THLE-3 cells were exposed to specific concentrations of either organic arsenic or MeHgCl, an increase in the H19 lncRNA levels and a marked reduction in the IGF2 mRNA levels were observed. In contrast, combined exposures coupled with CdCl2 resulted in the transcriptional repression of H19 and transcriptional activation of IGF2. It should be noted that the correlation between the dysregulated expression of H19/IGF2 and the hypermethylated CpG sites within the H19 differentially methylated region (DMR) was statistically significant. Furthermore, we performed transcriptomic analysis of the hepatocytes exposed to toxic metal combinations indicating enrichment of pro-inflammatory and anti-proliferative pathways compared to the unexposed cells. Our results suggest that hazardous metal mixtures may trigger epigenetic aberrations at the H19/IGF2 locus. We propose that altered CpG methylation in the H19 DMR could be a candidate biomarker for hepatic epigenotoxicity, in part, due to environmental exposure.
Collapse
Affiliation(s)
- Yehoon Jo
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Eugene Lim
- Institute of Health and Environment, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Jihye Park
- Department of Microbiology, College of Science & Technology, Dankook University, Cheonan, Republic of Korea
| | - Keunsoo Kang
- Department of Microbiology, College of Science & Technology, Dankook University, Cheonan, Republic of Korea
| | - Mi-Yeon Shin
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Office of Dental Education, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Jeong Weon Choi
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Department of Environmental Science, Baylor University, Waco, TX, USA
| | - Sungkyoon Kim
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea.
- Institute of Health and Environment, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea.
| | - Jaehyouk Lee
- Institute of Health and Environment, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Yi Q, Ouyang X, Zhong K, Chen Z, Zhu W, Zhu G, Zhong J. circFOXP1: a potential diagnostic and therapeutic target in human diseases. Front Immunol 2024; 15:1489378. [PMID: 39606233 PMCID: PMC11599189 DOI: 10.3389/fimmu.2024.1489378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Circular RNA (circRNA) are a unique class of non-coding RNAs characterized by their covalently closed loop structures, which grant them properties such as stability and conservation. Among these, circFOXP1 has been implicated in various diseases, including cancers, respiratory, skeletal, and cardiovascular disorders. This review systematically examines circFOXP1's role in disease progression, highlighting its involvement in critical biological processes, including cell proliferation, invasion, apoptosis, and autophagy. Mechanistically, circFOXP1 functions through miRNA sponging, protein interactions, and modulation of key signaling pathways such as Wnt and PI3K/AKT. We discuss its potential as a diagnostic and therapeutic target. Our analysis also identifies key unresolved questions, such as the precise regulatory networks involving circFOXP1 and its translation potential, offering pathways for future research.
Collapse
Affiliation(s)
- Qiang Yi
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinting Ouyang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Kui Zhong
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zheng Chen
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Weijian Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Gangfeng Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jinghua Zhong
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
34
|
Zhao C, Jin H, Lei Y, Li Q, Zhang Y, Lu Q. The dual effects of Benzo(a)pyrene/Benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide on DNA Methylation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175042. [PMID: 39084379 DOI: 10.1016/j.scitotenv.2024.175042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Benzo(a)pyrene (BaP) is one of the most thoroughly studied polycyclic aromatic hydrocarbons(PAHs) and a widespread organic pollutant in various areas of human life. Its teratogenic, immunotoxic and carcinogenic effects on organisms are well documented and widely recognized by researchers. In the body, BaP is enzymatically converted to form a more active benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE). BaP/BPDE has the potential to trigger gene mutations, influence epigenetic modifications and cause damage to cellular structures, ultimately contributing to disease onset and progression. However, there are different points of view when studying epigenetics using BaP/BPDE. On the one hand, it is claimed in cancer research that BaP/BPDE contributes to gene hypermethylation and, in particular, induces the hypermethylation of tumor's suppressor gene promoters, leading to gene silencing and subsequent cancer development. Conversely, studies in human and animal populations suggest that exposure to BaP results in genome-wide DNA hypomethylation, potentially leading to adverse outcomes in inflammatory diseases. This apparent contradiction has not been summarized in research for almost four decades. This article presents a comprehensive review of the current literature on the influence of BaP/BPDE on DNA methylation regulation. It demonstrates that BaP/BPDE exerts a dual-phase regulatory effect on methylation, which is influenced by factors such as the concentration and duration of BaP/BPDE exposure, experimental models and detection methods used in various studies. Acute/high concentration exposure to BaP/BPDE often results in global demethylation of DNA, which is associated with inhibition of DNA methyltransferase 1 (DNMT1) after exposure. At certain specific gene loci (e.g., RAR-β), BPDE can form DNA adducts, recruiting DNMT3 and leading to hypermethylation at specific sites. By integrating these different mechanisms, our goal is to unravel the patterns and regulations of BaP/BPDE-induced DNA methylation changes and provide insights into future precision therapies targeting epigenetics.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Jin
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
| | - Yu Lei
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qilin Li
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Central South University Hunan Key Laboratory of Medical Epigenomics Changsha, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
| |
Collapse
|
35
|
Moormann E. Normative implications of postgenomic deterministic narratives: the case study of epigenetic harm. HISTORY AND PHILOSOPHY OF THE LIFE SCIENCES 2024; 46:38. [PMID: 39503914 DOI: 10.1007/s40656-024-00636-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 09/23/2024] [Indexed: 12/20/2024]
Abstract
What do we mean when we talk about epigenetic harm? This paper presents a multidimensional view of epigenetic harm. It is a plea to take a step back from discussions of epigenetic responsibility distributions prevalent in ELSA literature on epigenetics. Instead, it urges researchers to take a closer look at the normative role played by the concept of epigenetic harm. It starts out by showing that the ways in which the object of epigenetic responsibility has already been conceptualized are all related to 'epigenetic harm': something negative that happens in which epigenetic mechanisms play a role, or rather something that needs to be avoided. Epigenetic harm is then characterized as a bridging concept between relatively neutral findings on epigenetics on the one side, and potential ethical and societal implications of those findings, primarily in terms of responsibility ascriptions and distributions, on the other. The paper proposes that a sufficiently nuanced account of epigenetic harm should include at least three dimensions. The dimension of causation alone leads to an overly narrow understanding of harm, and a wrong understanding of this dimension might prompt researchers to support an excessively simplistic epigenetic determinism. It is argued that a multidimensional analysis of epigenetic harm is less vulnerable to this threat and more reflective of the various kinds of harm that may be experienced by the subjects of epigenetic alterations. The paper applies insights from disability studies and feminist philosophy to draw attention to two other dimensions of epigenetic harm, namely lived experiences and relationality. The paper concludes by exploring what a shift towards a multidimensional approach to epigenetic harm might mean for epigenetic research and responsibility ascriptions by formulating some concrete implications.
Collapse
Affiliation(s)
- Emma Moormann
- Department of Philosophy, University of Antwerp, Rodestraat 14, 2000, Antwerp, Belgium.
| |
Collapse
|
36
|
Herrera-Luis E, Benke K, Volk H, Ladd-Acosta C, Wojcik GL. Gene-environment interactions in human health. Nat Rev Genet 2024; 25:768-784. [PMID: 38806721 DOI: 10.1038/s41576-024-00731-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 05/30/2024]
Abstract
Gene-environment interactions (G × E), the interplay of genetic variation with environmental factors, have a pivotal impact on human complex traits and diseases. Statistically, G × E can be assessed by determining the deviation from expectation of predictive models based solely on the phenotypic effects of genetics or environmental exposures. Despite the unprecedented, widespread and diverse use of G × E analytical frameworks, heterogeneity in their application and reporting hinders their applicability in public health. In this Review, we discuss study design considerations as well as G × E analytical frameworks to assess polygenic liability dependent on the environment, to identify specific genetic variants exhibiting G × E, and to characterize environmental context for these dynamics. We conclude with recommendations to address the most common challenges and pitfalls in the conceptualization, methodology and reporting of G × E studies, as well as future directions.
Collapse
Affiliation(s)
- Esther Herrera-Luis
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kelly Benke
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Heather Volk
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Ladd-Acosta
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Genevieve L Wojcik
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
37
|
Tu Q, Liu G, Liu X, Zhang J, Xiao W, Lv L, Zhao B. Perspective on using non-human primates in Exposome research. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117199. [PMID: 39426107 DOI: 10.1016/j.ecoenv.2024.117199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/02/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
The physiological and pathological changes in the human body caused by environmental pressures are collectively referred to as the Exposome. Human society is facing escalating environmental pollution, leading to a rising prevalence of associated diseases, including respiratory diseases, cardiovascular diseases, neurological disorders, reproductive development disorders, among others. Vulnerable populations to the pathogenic effects of environmental pollution include those in the prenatal, infancy, and elderly stages of life. Conducting Exposome mechanistic research and proposing effective health interventions are urgent in addressing the current severe environmental pollution. In this review, we address the core issues and bottlenecks faced by current Exposome research, specifically focusing on the most toxic ultrafine nanoparticles. We summarize multiple research models being used in Exposome research. Especially, we discuss the limitations of rodent animal models in mimicking human physiopathological phenotypes, and prospect advantages and necessity of non-human primates in Exposome research based on their evolutionary relatedness, anatomical and physiological similarities to human. Finally, we declare the initiation of NHPE (Non-Human Primate Exposome) project for conducting Exposome research using non-human primates and provide insights into its feasibility and key areas of focus. SYNOPSIS: Non-human primate models hold unique advantages in human Exposome research.
Collapse
Affiliation(s)
- Qiu Tu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China
| | - Gaojing Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiuyun Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jiao Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China
| | - Wenxian Xiao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Primate Facility, National Research Facility for Phenotypic & Genetic Analysis of Model Animals, and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Longbao Lv
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Primate Facility, National Research Facility for Phenotypic & Genetic Analysis of Model Animals, and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China.
| | - Bo Zhao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan 650223, China; Primate Facility, National Research Facility for Phenotypic & Genetic Analysis of Model Animals, and National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| |
Collapse
|
38
|
Bordoni L, Agostinho de Sousa J, Zhuo J, von Meyenn F. Evaluating the connection between diet quality, EpiNutrient intake and epigenetic age: an observational study. Am J Clin Nutr 2024; 120:1143-1155. [PMID: 39510725 DOI: 10.1016/j.ajcnut.2024.08.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/25/2024] [Accepted: 08/30/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND DNA methylation (DNAm) has unique properties which makes it a potential biomarker for lifestyle-related exposures. Epigenetic clocks, particularly DNAm-based biological age predictors [epigenetic age (EA)], represent an exciting new area of clinical research and deviations of EA from chronological age [epigenetic age acceleration (EAA)] have been linked to overall health, age-related diseases, and environmental exposures. OBJECTIVES This observational study investigates the relationships between biological aging and various dietary factors within the LifeLines-DEEP Cohort. These factors include diet quality, processed food consumption, dietary glycemic load, and intake of vitamins involved in maintaining the epigenetic homeostasis (vitamins B-9, B-12, B-6, B-2, and C). METHODS Dietary records collected using food-frequency questionnaires were used to estimate diet quality [LifeLines Diet Score (LLDS)], measure the intake of unprocessed/ultraprocessed food according to the NOVA food classification system, and the adequacy of the dietary intake of vitamins B-9, B-12, B-2, B-6, and C. EA using Horvath, Hannum, Levine, and Horvath2 epigenetic clock models and DNAm-predicted telomere length (DNAm-TL) were calculated from DNAm data in 760 subjects. Associations between dietary factors and EAA were tested, adjusting for sex, energy intake, and body composition. RESULTS LLDS was associated with EAA (EAA_Horvath: β: -0.148; P = 1 × 10-4; EAA_Hannum: β: -0.148; P = 9 × 10-5; EAA_Levine: β: -0.174; P = 1 × 10-5; and EAA_Horvath2: β: -0.176; P = 4 × 10-6) and DNAm-TL (β: 0.116; P = 0.003). Particularly, EAA was associated with dietary glycemic load (EAA_Horvath: β: 0.476; P = 9 × 10-10; EAA_Hannum: β: 0.565; P = 1 × 10-13; EAA_Levine: β: 0.469; P = 5 × 10-9; EAA_Horvath2: β: 0.569; P = 1 × 10-13; and DNAmTL adjusted for age: β: -0.340; P = 2 × 10-5) and different measures of food processing (NOVA classes 1 and 4). Positive EAA was also associated with inadequate intake of vitamin B-12 (EAA_Horvath: β: -0.167; P = 0.002; EAA_Hannum: β: -0.144; P = 0.007; and EAA_Horvath2: β: -0.126; P = 0.019) and C (EAA_Hannum: β: -0.136; P = 0.010 and EAA_Horvath2: β: -0.151; P = 0.005). CONCLUSIONS Our findings corroborate the hypothesis that nutrition plays a pivotal role in influencing epigenetic homeostasis, especially DNAm, thereby contributing to individual health trajectories and the pace of aging.
Collapse
Affiliation(s)
- Laura Bordoni
- Unit of Molecular Biology and Nutrigenomics, School of Pharmacy, University of Camerino, Camerino, Italy.
| | - João Agostinho de Sousa
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Jingran Zhuo
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Switzerland.
| |
Collapse
|
39
|
Irizar H, Chun Y, Hsu HHL, Li YC, Zhang L, Arditi Z, Grishina G, Grishin A, Vicencio A, Pandey G, Bunyavanich S. Multi-omic integration reveals alterations in nasal mucosal biology that mediate air pollutant effects on allergic rhinitis. Allergy 2024; 79:3047-3061. [PMID: 38796780 PMCID: PMC11560721 DOI: 10.1111/all.16174] [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: 11/02/2023] [Revised: 03/12/2024] [Accepted: 03/30/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Allergic rhinitis is a common inflammatory condition of the nasal mucosa that imposes a considerable health burden. Air pollution has been observed to increase the risk of developing allergic rhinitis. We addressed the hypotheses that early life exposure to air toxics is associated with developing allergic rhinitis, and that these effects are mediated by DNA methylation and gene expression in the nasal mucosa. METHODS In a case-control cohort of 505 participants, we geocoded participants' early life exposure to air toxics using data from the US Environmental Protection Agency, assessed physician diagnosis of allergic rhinitis by questionnaire, and collected nasal brushings for whole-genome DNA methylation and transcriptome profiling. We then performed a series of analyses including differential expression, Mendelian randomization, and causal mediation analyses to characterize relationships between early life air toxics, nasal DNA methylation, nasal gene expression, and allergic rhinitis. RESULTS Among the 505 participants, 275 had allergic rhinitis. The mean age of the participants was 16.4 years (standard deviation = 9.5 years). Early life exposure to air toxics such as acrylic acid, phosphine, antimony compounds, and benzyl chloride was associated with developing allergic rhinitis. These air toxics exerted their effects by altering the nasal DNA methylation and nasal gene expression levels of genes involved in respiratory ciliary function, mast cell activation, pro-inflammatory TGF-β1 signaling, and the regulation of myeloid immune cell function. CONCLUSIONS Our results expand the range of air pollutants implicated in allergic rhinitis and shed light on their underlying biological mechanisms in nasal mucosa.
Collapse
Affiliation(s)
- Haritz Irizar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Yoojin Chun
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Hsiao-Hsien Leon Hsu
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Yan-Chak Li
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Lingdi Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Zoe Arditi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Galina Grishina
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Alexander Grishin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Alfin Vicencio
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gaurav Pandey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
40
|
Molfino A, Ambrosani F, Udali S, Imbimbo G, Moruzzi S, Castagna A, Pattini P, Tambaro F, Ramaccini C, Muscaritoli M, Friso S. DNA Methylation Signatures Characterize Gene Expression Modulation in Lung Cancer Patients Affected by Anorexia. Nutrients 2024; 16:3721. [PMID: 39519555 PMCID: PMC11547925 DOI: 10.3390/nu16213721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES The pathophysiology of cancer anorexia is multifactorial and unclear. Transcriptomic analysis from PBMCs RNA showed diverse patterns of gene expression pathways in anorexic cancer patients. We assessed whether the different transcriptomic signatures are modulated by DNA methylation in lung cancer patients presenting with poor appetite. METHODS Lung cancer patients and controls were enrolled, and anorexia was assessed by the FAACT-score questionnaire. Genome-wide DNA methylation was determined by Human Infinium MethylationEPIC BeadChip Kit. Data from genome-wide methylation analysis were merged with those from gene expression analysis, previously obtained by RNA sequencing (NGS). Four groups of genes were identified for each comparison: hypermethylated repressed, hypermethylated induced, hypomethylated repressed, and hypomethylated induced. RESULTS Cancer patients (n = 16) showed 382 differentially methylated genes when compared with controls (n = 8). Anorexic patients (n = 8) presented 586 hypomethylated and 174 hypermethylated genes compared with controls. In anorexic patients vs. non-anorexic (n = 8), 211 genes were identified as hypomethylated and 90 hypermethylated. When microarray methylation data were merged with transcriptomic data by RNA sequencing, we observed significant differences in anorexic patients vs. controls; a total of 42 genes resulted as hypomethylated and induced, 5 hypermethylated repressed, 10 hypermethylated induced, and 15 hypomethylated repressed. The CG sites analyzed by targeted bisulfite NGS in four genes of interest (FLNA, PGRMC1, GNL3L, and FHL1) resulting as hypomethylated in anorexic vs. controls allowed the validation of the data obtained from DNA methylation. Interestingly, the four genes resulted as hypomethylated in anorexic patients vs. non-anorexic patients and vs. controls (p < 0.0001). CONCLUSIONS Our data support that methylation is implicated in cancer-associated anorexia and nutritional derangements among lung cancer patients.
Collapse
Affiliation(s)
- Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Francesca Ambrosani
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Silvia Udali
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Giovanni Imbimbo
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Sara Moruzzi
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Annalisa Castagna
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Patrizia Pattini
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| | - Federica Tambaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Cesarina Ramaccini
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.I.); (F.T.); (C.R.); (M.M.)
| | - Simonetta Friso
- Unit of Internal Medicine B, Department of Medicine, School of Medicine, University of Verona, 37129 Verona, Italy; (F.A.); (S.U.); (S.M.); (A.C.); (P.P.)
| |
Collapse
|
41
|
Ruehlmann AK, Cecil KM, Lippert F, Yolton K, Ryan PH, Brunst KJ. Epigenome-wide association study of fluoride exposure during early adolescence and DNA methylation among U.S. children. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 948:174916. [PMID: 39038671 PMCID: PMC11514227 DOI: 10.1016/j.scitotenv.2024.174916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Exposure to fluoride in early childhood has been associated with altered cognition, intelligence, attention, and neurobehavior. Fluoride-related neurodevelopment effects have been shown to vary by sex and very little is known about the mechanistic processes involved. There is limited research on how fluoride exposure impacts the epigenome, potentially leading to changes in DNA methylation of specific genes regulating key developmental processes. In the Cincinnati Childhood Allergy and Air Pollution Study (CCAAPS), urine samples were analyzed using a microdiffusion method to determine childhood urinary fluoride adjusted for specific gravity (CUFsg) concentrations. Whole blood DNA methylation was assessed using the Infinium MethylationEPIC BeadChip 850 k Array. In a cross-sectional analysis, we interrogated epigenome-wide DNA methylation at 775,141 CpG loci across the methylome in relation to CUFsg concentrations in 272 early adolescents at age 12 years. Among all participants, higher concentrations of CUF were associated with differential methylation of one CpG (p < 6 × 10-8) located in the gene body of GBF1 (cg25435255). Among females, higher concentrations of CUFsg were associated with differential methylation of 7 CpGs; only three CpGs were differentially methylated among males with no overlap of significant CpGs observed among females. Secondary analyses revealed several differentially methylated regions (DMRs) and CpG loci mapping to genes with key roles in psychiatric outcomes, social interaction, and cognition, as well as immunologic and metabolic phenotypes. While fluoride exposure may impact the epigenome during early adolescence, the functional consequences of these changes are unclear warranting further investigation.
Collapse
Affiliation(s)
- Anna K Ruehlmann
- University of Cincinnati, College of Medicine, Department of Environmental and Public Health Sciences, Cincinnati, OH, USA
| | - Kim M Cecil
- University of Cincinnati, College of Medicine, Department of Environmental and Public Health Sciences, Cincinnati, OH, USA; Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Frank Lippert
- Department of Cariology, Operative Dentistry, and Dental Public Health, Oral Health Research Institute, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Kimberly Yolton
- University of Cincinnati, College of Medicine, Department of Environmental and Public Health Sciences, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Patrick H Ryan
- University of Cincinnati, College of Medicine, Department of Environmental and Public Health Sciences, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kelly J Brunst
- University of Cincinnati, College of Medicine, Department of Environmental and Public Health Sciences, Cincinnati, OH, USA.
| |
Collapse
|
42
|
Kim D, Gil J, Bae ON. PM2.5 potentiates oxygen glucose deprivation-induced neurovascular unit damage via inhibition of the Akt/β-catenin pathway and autophagy dysregulation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124728. [PMID: 39147226 DOI: 10.1016/j.envpol.2024.124728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/25/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Air pollution has recently emerged as a significant risk factor for ischemic stroke. Although there is a robust association between higher concentrations of ambient particulate matter (PM2.5) and increased incidence and mortality rates of ischemic stroke, the precise mechanisms underlying PM2.5-induced ischemic stroke remain to be fully elucidated. The purpose of this study was to examine the synergistic effect of PM2.5 and hypoxic stress using in vivo and in vitro ischemic stroke models. Intravenously administered PM2.5 exacerbated the ischemic brain damage induced by middle cerebral artery occlusion (MCAo) in Sprague Dawley rats. Alterations in autophagy flux and decreased levels of tight junction proteins were observed in the brain of PM2.5-administered rats after MCAo. The underlying mechanism of PM2.5-induced potentiation of ischemic brain damage was investigated in neurons, perivascular macrophages, and brain endothelial cells, which are the major components of the integrated neurovascular unit. Co-treatment with PM2.5 and oxygen-glucose deprivation (OGD) amplified the effects of OGD on the reduction of viability in primary neurons, immortalized murine hippocampal neuron (HT-22), and brain endothelial cells (bEND.3). After co-treatment with PM2.5 and OGD, the Akt/β-catenin and autophagy flux were significantly inhibited in HT-22 cells. Notably, the protein levels of metalloproteinase-9 and cystatin C were elevated in the conditioned media of murine macrophages (RAW264.7) exposed to PM2.5, and tight junction protein expression was significantly decreased after OGD exposure in bEND.3 cells pretreated with the conditioned media. Our findings suggest that perivascular macrophages may mediate PM2.5-induced brain endothelial dysfunction following ischemia and that PM2.5 can exacerbate ischemia-induced neurovascular damage.
Collapse
Affiliation(s)
- Donghyun Kim
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, South Korea
| | - Junkyung Gil
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, South Korea
| | - Ok-Nam Bae
- College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, South Korea.
| |
Collapse
|
43
|
Zhao J, Qiu YK, Xie YX, Li XY, Li YB, Wu B, Wang YW, Tian XY, Lv YL, Zhang LH, Li WL, Yang HF. Imbalance of mitochondrial quality control regulated by STING and PINK1 affects cyfluthrin-induced neuroinflammation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174313. [PMID: 38964406 DOI: 10.1016/j.scitotenv.2024.174313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Nervous system diseases are a global health problem, and with the increase in the elderly population around the world, their incidence will also increase. Harmful substances in the environment are closely related to the occurrence of nervous system diseases. China is a large agricultural country, and thus the insecticide cyfluthrin has been widely used. Cyfluthrin is neurotoxic, but the mechanism of this injury is not clear. Inflammation is an important mechanism for the occurrence of nervous system diseases. Mitochondria are the main regulators of the inflammatory response, and various cellular responses, including autophagy, directly affect the regulation of inflammatory processes. Mitochondrial damage is related to mitochondrial quality control (MQC) and PTEN-induced kinase 1 (PINK1). As an anti-inflammatory factor, stimulator of interferon genes (STING) participates in the regulation of inflammation. However, the relationship between STING and mitochondria in the process of cyfluthrin-induced nerve injury is unclear. This study established in vivo and in vitro models of cyfluthrin exposure to explore the role of MQC and to clarify the mechanism of action of STING and PINK1. Our results showed that cyfluthrin can increase the reactive oxygen species (ROS) level, resulting in mitochondrial damage and inflammation. In this process, an imbalance in MQC leads to the aggravation of mitochondrial damage, and high STING expression drives the occurrence of inflammation. We established a differential expression model of STING and PINK1 to further determine the underlying mechanism and found that the interaction between STING and PINK1 regulates MQC to affect the levels of mitochondrial damage and inflammation. When STING and PINK1 expression are downregulated, mitochondrial damage and STING-induced inflammation are significantly alleviated. In summary, a synergistic effect between STING and PINK1 on cyfluthrin-induced neuroinflammation may exist, which leads to an imbalance in MQC by inhibiting mitochondrial biogenesis and division/fusion, and PINK1 can reduce STING-driven inflammation.
Collapse
Affiliation(s)
- Ji Zhao
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China; Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, PR China
| | - Yi-Kai Qiu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Eduction, Yinchuan 750004, PR China
| | - Yong-Xing Xie
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Xiao-Yu Li
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Yu-Bin Li
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Bing Wu
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Yu-Wen Wang
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Xue-Yan Tian
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Yan-Ling Lv
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Ling-He Zhang
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China
| | - Wen-Li Li
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, PR China.
| | - Hui-Fang Yang
- College of Public Health, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, PR China.
| |
Collapse
|
44
|
Navarro-Gallinad A, Orlandi F, Scott J, Havyarimana E, Basu N, Little MA, O'Sullivan D. Enabling data linkages for rare diseases in a resilient environment with the SERDIF framework. NPJ Digit Med 2024; 7:274. [PMID: 39367112 PMCID: PMC11452697 DOI: 10.1038/s41746-024-01267-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/16/2024] [Indexed: 10/06/2024] Open
Abstract
Environmental factors amplified by climate change contribute significantly to the global burden of disease, disproportionately impacting vulnerable populations, such as individuals with rare diseases. Researchers require innovative, dynamic data linkage methods to enable the development of risk prediction models, particularly for diseases like vasculitis with unknown aetiology but potential environmental triggers. In response, we present the Semantic Environmental and Rare Disease Data Integration Framework (SERDIF). SERDIF was evaluated with researchers studying climate-related health hazards of vasculitis disease activity across European countries (NP1 = 10, NP2 = 17, NP3 = 23). Usability metrics consistently improved, indicating SERDIF's effectiveness in linking complex environmental and health datasets. Furthermore, SERDIF-enabled epidemiologists to study environmental factors in a pregnancy cohort in Lombardy, showcasing its versatility beyond rare diseases. This framework offers for the first time a user-friendly, FAIR-compliant design for environment-health data linkage with export capabilities enabling data analysis to mitigate health risks posed by climate change.
Collapse
Affiliation(s)
- Albert Navarro-Gallinad
- ADAPT Centre for Digital Content, School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland.
- Health Data Science Centre, Fondazione Human Technopole, Milan, Italy.
| | - Fabrizio Orlandi
- ADAPT Centre for Digital Content, School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
| | - Jennifer Scott
- Trinity Kidney Centre, Trinity College Dublin, The University of Dublin, Trinity Translational Medicine Institute, Dublin, Ireland
| | - Enock Havyarimana
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Neil Basu
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Mark A Little
- ADAPT Centre for Digital Content, School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
- Trinity Kidney Centre, Trinity College Dublin, The University of Dublin, Trinity Translational Medicine Institute, Dublin, Ireland
| | - Declan O'Sullivan
- ADAPT Centre for Digital Content, School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
45
|
Jia D, Chen S. Environmental pollutants and bad bugs work hand in glove. Trends Cancer 2024; 10:873-875. [PMID: 39256091 DOI: 10.1016/j.trecan.2024.08.007] [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: 08/23/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024]
Abstract
'Bad bacteria' could alter the toxicokinetics of environmental pollutants, thereby exacerbating chemically induced tumorigenesis. Recently, Roje et al. reported that specific gut microbiota can metabolize nitrosamine compounds to a toxic oxidation product, aggravating bladder cancer development and progression. These findings have important implications for tumor intervention through the gut microbiota.
Collapse
Affiliation(s)
- Dingjiacheng Jia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Shujie Chen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China; Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| |
Collapse
|
46
|
Yao L, Yang C, Graff JC, Wang G, Wang G, Gu W. From Reactive to Proactive - The Future Life Design to Promote Health and Extend the Human Lifespan. Adv Biol (Weinh) 2024; 8:e2400148. [PMID: 39037380 DOI: 10.1002/adbi.202400148] [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: 03/15/2024] [Revised: 06/11/2024] [Indexed: 07/23/2024]
Abstract
Disease treatment and prevention have improved the human lifespan. Current studies on aging, such as the biological clock and senolytic drugs have focused on the medical treatments of various disorders and health maintenance. However, to efficiently extend the human lifespan to its theoretical maximum, medicine can take a further proactive approach and identify the inapparent disorders that affect the gestation, body growth, and reproductive stages of the so-called "healthy" population. The goal is to upgrade the standard health status to a new level by targeting the inapparent disorders. Thus, future research can shift from reaction, response, and prevention to proactive, quality promotion and vigor prolonging; from single disease-oriented to multiple dimension protocol for a healthy body; from treatment of symptom onset to keep away from disorders; and from the healthy aging management to a healthy promotion design beginning at the birth.
Collapse
Affiliation(s)
- Lan Yao
- College of Health management, Harbin Medical University, 157 Baojian Road, Harbin, Heilongjiang, 150081, China
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Chengyuan Yang
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - J Carolyn Graff
- Department of Health Promotion and Disease Prevention, College of Nursing, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Guiying Wang
- Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150007, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150007, China
| | - Weikuan Gu
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Research Service, Memphis VA Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN, 38163, USA
| |
Collapse
|
47
|
Joyce BT. Epigenomics of Cardio-Oncology. JACC CardioOncol 2024; 6:743-745. [PMID: 39479332 PMCID: PMC11520199 DOI: 10.1016/j.jaccao.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Affiliation(s)
- Brian T. Joyce
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
48
|
Yu N, Su M, Wang J, Liu Y, Yang J, Zhang J, Wang M. Long-Term Exposure of Fresh and Aged Nano Zinc Oxide Promotes Hepatocellular Carcinoma Malignancy by Up-Regulating Claudin-2. Int J Nanomedicine 2024; 19:9989-10008. [PMID: 39371475 PMCID: PMC11453161 DOI: 10.2147/ijn.s478279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024] Open
Abstract
Background Tumor development and progression is a long and complex process influenced by a combination of intrinsic (eg, gene mutation) and extrinsic (eg, environmental pollution) factors. As a detoxification organ, the liver plays an important role in human exposure and response to various environmental pollutants including nanomaterials (NMs). Hepatocellular carcinoma (HCC) is one of the most common malignant tumors and remains a serious threat to human health. Whether NMs promote liver cancer progression remains elusive and assessing long-term exposure to subtoxic doses of nanoparticles (NPs) remains a challenge. In this study, we focused on the promotional effects of nano zinc oxide (nZnO) on the malignant progression of human HCC cells HepG2, especially aged nZnO that has undergone physicochemical transformation. Methods In in vitro experiments, we performed colony forming efficiency, soft agar colony formation, and cell migration/invasion assays on HepG2 cells that had been exposed to a low dose of nZnO (1.5 μg/mL) for 3 or 4 months. In in vivo experiments, we subcutaneously inoculated HepG2 cells that had undergone long-term exposure to nZnO for 4 months into BALB/c athymic nude mice and observed tumor formation. ZnCl2 was administered to determine the role of zinc ions. Results Chronic low-dose exposure to nZnO significantly intensified the malignant progression of HCC cells, whereas aged nZnO may exacerbate the severity of malignant progression. Furthermore, through transcriptome sequencing analysis and in vitro cellular rescue experiments, we demonstrated that the mechanism of nZnO-induced malignant progression of HCC could be linked to the activation of Claudin-2 (CLDN2), one of the components of cellular tight junctions, and the dysregulation of its downstream signaling pathways. Conclusion Long-term exposure of fresh and aged nZnO promotes hepatocellular carcinoma malignancy by up-regulating CLDN2. The implications of this work can be profound for cancer patients, as the use of various nanoproducts and unintentional exposure to environmentally transformed NMs may unknowingly hasten the progression of their cancers.
Collapse
Affiliation(s)
- Na Yu
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Mingqin Su
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Juan Wang
- Department of Public Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Yakun Liu
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Jingya Yang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Jingyi Zhang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Meimei Wang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People’s Republic of China
| |
Collapse
|
49
|
Zeng Y, Bai X, Zhu G, Zhu M, Peng W, Song J, Cai H, Ye L, Chen C, Song Y, Jin M, Zhang XQ, Wang J. m 6A-mediated HDAC9 upregulation promotes particulate matter-induced airway inflammation via epigenetic control of DUSP9-MAPK axis and acts as an inhaled nanotherapeutic target. JOURNAL OF HAZARDOUS MATERIALS 2024; 477:135093. [PMID: 39088948 DOI: 10.1016/j.jhazmat.2024.135093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/15/2024] [Accepted: 07/01/2024] [Indexed: 08/03/2024]
Abstract
Exposure to particulate matter (PM) can cause airway inflammation and worsen various airway diseases. However, the underlying molecular mechanism by which PM triggers airway inflammation has not been completely elucidated, and effective interventions are lacking. Our study revealed that PM exposure increased the expression of histone deacetylase 9 (HDAC9) in human bronchial epithelial cells and mouse airway epithelium through the METTL3/m6A methylation/IGF2BP3 pathway. Functional assays showed that HDAC9 upregulation promoted PM-induced airway inflammation and activation of MAPK signaling pathway in vitro and in vivo. Mechanistically, HDAC9 modulated the deacetylation of histone 4 acetylation at K12 (H4K12) in the promoter region of dual specificity phosphatase 9 (DUSP9) to repress the expression of DUSP9 and resulting in the activation of MAPK signaling pathway, thereby promoting PM-induced airway inflammation. Additionally, HDAC9 bound to MEF2A to weaken its anti-inflammatory effect on PM-induced airway inflammation. Then, we developed a novel inhaled lipid nanoparticle system for delivering HDAC9 siRNA to the airway, offering an effective treatment for PM-induced airway inflammation. Collectively, we elucidated the crucial regulatory mechanism of HDAC9 in PM-induced airway inflammation and introduced an inhaled therapeutic approach targeting HDAC9. These findings contribute to alleviating the burden of various airway diseases caused by PM exposure.
Collapse
Affiliation(s)
- Yingying Zeng
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Bai
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guiping Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mengchan Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenjun Peng
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Juan Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hui Cai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ling Ye
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cuicui Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai 200032, China; Shanghai Respiratory Research Institute, Shanghai 200032, China
| | - Meiling Jin
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xue-Qing Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jian Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
50
|
Potter C, Hill C, Smyth LJ, Neville C, Scott A, Kee F, McGuinness B, McKnight A. Cohort profile: DNA methylation in the Northern Ireland Cohort for the Longitudinal Study of Ageing (NICOLA) - recruitment and participant characteristics. BMJ Open 2024; 14:e085652. [PMID: 39277204 PMCID: PMC11404182 DOI: 10.1136/bmjopen-2024-085652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024] Open
Abstract
PURPOSE Epigenetic modifications including DNA methylation (DNAm) are proposed mechanisms by which social or environmental exposures may influence health and behaviours as we age. The Northern Ireland Cohort for the Longitudinal Study of Ageing (NICOLA) DNAm cohort, established in 2013, is one of several worldwide, nationally representative prospective studies of ageing with biological samples from participants who consented to multiomic analysis. PARTICIPANTS NICOLA recruited 8478 participants (8283 aged 50 years or older and 195 spouses or partners at the same address aged under 50 years). Computer-Assisted Personal Interviews, Self-Completion Questionnaires and detailed Health Assessments (HA) were completed. Of the 3471 (44.1%) participants who attended the HA in wave 1, which included venous blood sampling, 2000 were identified for the DNAm cohort. Following technical and data quality control checks, DNAm data are currently available for n=1870. FINDINGS TO DATE There was no significant difference based on age, self-reported gender, education, employment, smoking or alcohol status and subjective health reports between the DNAm cohort and other HA attendees. Participants were more likely to be in the DNAm group if they lived with one other person (OR 1.26, 95% CI 1.07 to 1.49). The DNAm group had a lower proportion of depressed participants and those meeting criteria for post-traumatic stress disorder (11.7% and 4.4% vs 13.5% and 4.5%, respectively) categorised by objective assessment tools but this was not significant (OR 0.84, 95% CI 0.69 to 1.02 and OR 0.87, 95% CI 0.64 to 1.19). FUTURE PLANS The deeply phenotyped DNAm cohort in NICOLA with planned prospective follow-up and additional multiomic data releases will increase the cohort's utility for research into ageing. The genomic and epigenetic data for the DNAm cohort has been deposited on the European Genome-Phenome Archive, increasing the profile of this cohort and data availability to researchers.
Collapse
Affiliation(s)
- Claire Potter
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Claire Hill
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Laura J Smyth
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | | | - Angela Scott
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Frank Kee
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | | | - Amy McKnight
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| |
Collapse
|