1
|
Lin KA, Su CC, Liu SH, Lee KI, Fang KM, Tang CH, Kuo CY, Chang KC, Ke JA, Huang CF, Chen YW, Yang CY. Antimony induces mitochondria-dependent and ER stress-triggered apoptosis via the oxidative stress-activated JNK signaling pathway in pancreatic islet β-cells. Toxicology 2025; 516:154188. [PMID: 40368022 DOI: 10.1016/j.tox.2025.154188] [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: 02/17/2025] [Revised: 04/30/2025] [Accepted: 05/11/2025] [Indexed: 05/16/2025]
Abstract
Antimony (Sb), a silvery-white metal, is a heavy metal of particular prevalence that has the ability to result in adverse effects in humans through environmental exposure resulting from natural processes and human activities. Epidemiological studies have suggested that Sb has an association with the potential for diabetes mellitus (DM) development. However, the mechanisms by which Sb exerts toxicological effects on pancreatic islet β-cells are still not clear. In this investigation, Sb exposure significantly inhibited rat pancreatic islet β-cell-derived RIN-m5F cell viability and insulin secretion, while inducing mitochondria-dependent apoptotic signals, inclusive of increased apoptotic cell populations, caspase-3 activity, the expression of PARP and caspase-3/-7/-9, and mitochondrial dysfunction. RIN-m5F cells exposure to Sb also led to the triggering of endoplasmic reticulum (ER) stress via the induction of a number of vital molecules, including CHOP, XBP-1s, and caspase-12. In Sb-exposed RIN-m5F cells, 4-PBA pretreatment (an inhibitor of ER stress) significantly suppressed protein expression related to ER stress and events of an apoptotic nature. Furthermore, exposure to Sb resulted in the significant activation of AMPKα, ERK1/2, and JNK signaling, as well as reactive oxygen species (ROS) generation. Pretreatment with SP600125 (an inhibitor of JNK) and antioxidant NAC, but not PD98059 (an inhibitor of ERK) or compound C (an inhibitor of AMPK), effectively abrogated the cytotoxicity, ER stress responses, mitochondrial dysfunction, apoptotic events, insulin secretion inhibition, and JNK activation in Sb-exposed rat pancreatic islet β-cells. However, SP600125 did not prevent ROS generation, which was inhibited by the antioxidant NAC. Collectively, the results demonstrate exposure to Sb to exert β-cell cytotoxicity through oxidative stress-activated JNK signaling downstream-regulated mitochondria-dependent and ER stress-triggered cell apoptotic pathways, eventually resulting in the death of rat pancreatic islet β-cells.
Collapse
Affiliation(s)
- Ken-An Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County, Changhua County 500, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Kai-Min Fang
- Department of Otolaryngology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Chun-Ying Kuo
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County, Changhua County 500, Taiwan
| | - Kai-Chih Chang
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Jun-An Ke
- Department of Medical Education, Changhua Christian Hospital Changhua City 500, Taiwan
| | - Chun-Fa Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan; Department of Nursing, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| | - Ya-Wen Chen
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan.
| | - Ching-Yao Yang
- Department of Surgery, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan.
| |
Collapse
|
2
|
Liu G, Yang J, Li R, Li W, Liu D, Zhang N, Zhao Y, He Z, Gu S. Roles of N 6-methyladenosine in LncRNA changes and oxidative damage in cadmium-induced pancreatic β-cells. Toxicology 2025; 511:154053. [PMID: 39798863 DOI: 10.1016/j.tox.2025.154053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
N6-methyladenosine (m6A) modification and LncRNAs play crucial regulatory roles in various pathophysiological processes, yet roles of m6A modification and the relationship between m6A modification and LncRNAs in cadmium-induced oxidative damage of pancreatic β-cells have not been fully elucidated. In this study, m6A agonist entacapone and inhibitor 3-deazadenosine were used to identify the effects of m6A on cadmium-induced oxidative damage as well as LncRNA changes. Our results indicate that elevated levels of m6A modification by entacapone can rescue the cell viability and attenuate the cell apoptosis, while the inhibition levels of m6A modification can exacerbate the cell death. Furthermore, the elevation of m6A modification can recover cadmium-induced oxidative damage to pancreatic β-cells, which characterized as inhibition the ROS accumulation, MDA contents, protein expressions of Nrf2 and Ho-1, while elevation the expressions of Sod1 and Gclc. On the contrary, the reduction levels of m6A modification can exacerbate the cadmium-induced oxidative damage. More importantly, six significantly differentially expressed LncRNAs were selected according to our preliminary sequencing data (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE253072) and there is a clear correlation between the levels of these LncRNAs and m6A modification after cadmium treatment. Interestingly, the intervention of m6A modification levels can significantly affect the levels of these LncRNAs. In detail, the stimulation of m6A modification reversed the changes of cadmium-induced LncRNAs, while the m6A modification inhibition can significantly exacerbate the changes of cadmium-induced LncRNAs. In conclusion, our data revealed critical roles of m6A modification in cadmium-induced LncRNAs and oxidative damage. Our findings point to a new direction for future studies on the molecular mechanisms of pancreatic β-cell damage induced by cadmium.
Collapse
Affiliation(s)
- Guofen Liu
- School of Public Health, Dali University, Dali, Yunnan, China; Yiyang Vocational and Technical College, Yiyang, Hunan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Jie Yang
- College of Engineering, Dali University, Dali, Yunnan, China
| | - Rongxian Li
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Wenhong Li
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - De Liu
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Nan Zhang
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Yuan Zhao
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Zuoshun He
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Shiyan Gu
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China.
| |
Collapse
|
3
|
Mou Y, Sun Y, Liu G, Zhang N, He Z, Gu S. Screening of differentially expressed RNAs and identifying a ceRNA axis during cadmium-induced oxidative damage in pancreatic β cells. Sci Rep 2024; 14:18962. [PMID: 39152192 PMCID: PMC11329516 DOI: 10.1038/s41598-024-69937-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024] Open
Abstract
Cadmium, a common metal pollutant, has been demonstrated to induce type 2 diabetes by disrupting pancreatic β cells function. In this study, transcriptome microarray was utilized to identify differential gene expression in oxidative damage to pancreatic β cells following cadmium exposure. The results indicated that a series of mRNAs, LncRNAs, and miRNAs were altered. Of the differentially expressed miRNAs, miR-29a-3p exhibited the most pronounced alteration, with an 11.62-fold increase relative to the control group. Following this, the target gene of miR-29a-3p was identified as Col3a1 through three databases (miRDB, miRTarbase and Tarbase), which demonstrated a decrease across the transcriptome microarray. The upstream target gene of miR-29a-3p was identified as NONMMUT036805, with decreased expression observed in the microarray. Finally, the expression trend of NONMMUT036805/miR-29a-3p/Col3a1 was reversed following NAC pretreatment. This was accompanied by a reduction in oxidative damage indicators, MDA/ROS/GSH-Px appeared to be negatively affected to varying degrees. In conclusion, this study has demonstrated that multiple RNAs are altered during cadmium exposure-induced oxidative damage in pancreatic β cells. The NONMMUT036805/miR-29a-3p/Col3a1 axis has been shown to be involved in this process, which provides a foundation for the identification of potential targets for cadmium toxicity intervention.
Collapse
Affiliation(s)
- Yahao Mou
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan, 671000, People's Republic of China
- Sichuan Tianfu New Area Public Health Center, Zhengxing Street, Chengdu, Sichuan, 610218, People's Republic of China
| | - Yifei Sun
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan, 671000, People's Republic of China
| | - Guofen Liu
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan, 671000, People's Republic of China
| | - Nan Zhang
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan, 671000, People's Republic of China
| | - Zuoshun He
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan, 671000, People's Republic of China
| | - Shiyan Gu
- Institute of Preventive Medicine, School of Public Health, Dali University, No. 22, Wanhua Road, Dali, Yunnan, 671000, People's Republic of China.
| |
Collapse
|
4
|
Pallagi P, Tóth E, Görög M, Venglovecz V, Madácsy T, Varga Á, Molnár T, Papp N, Szabó V, Kúthy-Sutus E, Molnár R, Ördög A, Borka K, Schnúr A, Kéri A, Kajner G, Csekő K, Ritter E, Csupor D, Helyes Z, Galbács G, Szentesi A, Czakó L, Rakonczay Z, Takács T, Maléth J, Hegyi P. Heavy metals in cigarette smoke strongly inhibit pancreatic ductal function and promote development of chronic pancreatitis. Clin Transl Med 2024; 14:e1733. [PMID: 38877637 PMCID: PMC11178517 DOI: 10.1002/ctm2.1733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND AND AIMS Smoking is recognised as an independent risk factor in the development of chronic pancreatitis (CP). Cystic fibrosis transmembrane conductance regulator (CFTR) function and ductal fluid and bicarbonate secretion are also known to be impaired in CP, so it is crucial to understand the relationships between smoking, pancreatic ductal function and the development of CP. METHODS We measured sweat chloride (Cl-) concentrations in patients with and without CP, both smokers and non-smokers, to assess CFTR activity. Serum heavy metal levels and tissue cadmium concentrations were determined by mass spectrometry in smoking and non-smoking patients. Guinea pigs were exposed to cigarette smoke, and cigarette smoke extract (CSE) was prepared to characterise its effects on pancreatic HCO3 - and fluid secretion and CFTR function. We administered cerulein to both the smoking and non-smoking groups of mice to induce pancreatitis. RESULTS Sweat samples from smokers, both with and without CP, exhibited elevated Cl- concentrations compared to those from non-smokers, indicating a decrease in CFTR activity due to smoking. Pancreatic tissues from smokers, regardless of CP status, displayed lower CFTR expression than those from non-smokers. Serum levels of cadmium and mercury, as well as pancreatic tissue cadmium, were increased in smokers. Smoking, CSE, cadmium, mercury and nicotine all hindered fluid and HCO3 - secretion and CFTR activity in pancreatic ductal cells. These effects were mediated by sustained increases in intracellular calcium ([Ca2+]i), depletion of intracellular ATP (ATPi) and mitochondrial membrane depolarisation. CONCLUSION Smoking impairs pancreatic ductal function and contributes to the development of CP. Heavy metals, notably cadmium, play a significant role in the harmful effects of smoking. KEY POINTS Smoking and cigarette smoke extract diminish pancreatic ductal fluid and HCO3 - secretion as well as the expression and function of CFTR Cd and Hg concentrations are significantly higher in the serum samples of smokers Cd accumulates in the pancreatic tissue of smokers.
Collapse
Affiliation(s)
- Petra Pallagi
- Department of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Emese Tóth
- Department of Medicine, University of Szeged, Szeged, Hungary
- Department of Theoretical and Integrative Health Sciences, University of Debrecen, Szeged, Hungary
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
| | - Marietta Görög
- Department of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
| | - Viktória Venglovecz
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Tamara Madácsy
- Department of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Árpád Varga
- Department of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Tünde Molnár
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Noémi Papp
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - Viktória Szabó
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Enikő Kúthy-Sutus
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Réka Molnár
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - Attila Ördög
- Department of Plant Biology, University of Szeged, Szeged, Hungary
| | - Katalin Borka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Schnúr
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - Albert Kéri
- Department of Molecular and Analytical Chemistry, University of Szeged, Szeged, Hungary
| | - Gyula Kajner
- Department of Molecular and Analytical Chemistry, University of Szeged, Szeged, Hungary
| | - Kata Csekő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory of Drug Research and Development (Pharmalab), Budapest, Hungary
| | - Emese Ritter
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory of Drug Research and Development (Pharmalab), Budapest, Hungary
| | - Dezső Csupor
- Institute of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute of Clinical Pharmacy, University of Szeged, Szeged, Hungary
- Institute for Translational Medicine, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory of Drug Research and Development (Pharmalab), Budapest, Hungary
- Eötvös Loránd Research Network Chronic Pain Research Group, University of Pécs, Pécs, Hungary
| | - Gábor Galbács
- Department of Molecular and Analytical Chemistry, University of Szeged, Szeged, Hungary
| | - Andrea Szentesi
- Institute for Translational Medicine, University of Pécs, Pécs, Hungary
| | - László Czakó
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - Zoltán Rakonczay
- Department of Pathophysiology, University of Szeged, Szeged, Hungary
| | - Tamás Takács
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - József Maléth
- Department of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
- Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- Center of Translational Medicine and Institute of Pancreatic Disorders, Semmelweis University, Budapest, Hungary
| |
Collapse
|
5
|
He S, Jiang T, Zhang D, Li M, Yu T, Zhai M, He B, Yin T, Wang X, Tao F, Yao Y, Ji D, Yang Y, Liang C. Association of exposure to multiple heavy metals during pregnancy with the risk of gestational diabetes mellitus and insulin secretion phase after glucose stimulation. ENVIRONMENTAL RESEARCH 2024; 248:118237. [PMID: 38244971 DOI: 10.1016/j.envres.2024.118237] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Epidemiological evidence for the association between heavy metals exposure during pregnancy and gestational diabetes mellitus (GDM) is still inconsistent. Additionally, that is poorly understood about the potential cause behind the association, for instance, whether heavy metal exposure is related to the change of insulin secretion phase is unknown. OBJECTIVES We aimed to explore the relationships of blood levels of arsenic (As), lead (Pb), thallium (Tl), nickel (Ni), cadmium (Cd), cobalt (Co), barium (Ba), chromium (Cr), mercury (Hg) and copper (Cu) during early pregnancy with the odds of GDM, either as an individual or a mixture, as well as the association of the metals with insulin secretion phase after glucose stimulation. METHODS We performed a nested case-control study consisting of 302 pregnant women with GDM and 302 controls at the First Affiliated Hospital of Anhui Medical University in Hefei, China. Around the 12th week of pregnancy, blood samples of pregnant women were collected and levels of As, Pb, Tl, Ni, Cd, Co, Ba, Cr, Hg and Cu in blood were measured. An oral glucose tolerance test (OGTT) was done in each pregnant woman during the 24-28th week of pregnancy to diagnose GDM and C-peptide (CP) levels during OGTT were measured simultaneously. The four metals (As, Pb, Tl and Ni) with the highest effect on odds of GDM were selected for the subsequent analyses via the random forest model. Conditional logistic regression models were performed to analyze the relationships of blood As, Pb, Tl and Ni levels with the odds of GDM. The weighted quantile sum (WQS) regression and bayesian kernel machine regression (BKMR) were used to assess the joint effects of levels of As, Pb, Tl and Ni on the odds of GDM as well as to evaluate which metal level contributed most to the association. Latent profile analysis (LPA) was conducted to identify profiles of glycemic and C-peptide levels at different time points. Multiple linear regression models were employed to explore the relationships of metals with glycaemia-related indices (fasting blood glucose (FBG), 1-hour blood glucose (1h BG), 2-hour blood glucose (2h BG), fasting C-peptide (FCP), 1-hour C-peptide (1h CP), 2-hour C-peptide (2h CP), FCP/FBG, 1h CP/1h BG, 2h CP/2h BG, area under the curve of C-peptide (AUCP), area under the curve of glucose (AUCG), AUCP/AUCG and profiles of BGs and CPs, respectively. Mixed-effects models with repeated measures data were used to explore the relationship between As (the ultimately selected metal) level and glucose-stimulated insulin secretion phase. The mediation effects of AUCP and AUCG on the association of As exposure with odds of GDM were investigated using mediation models. RESULTS The odds of GDM in pregnant women increased with every ln unit increase in blood As concentration (odds ratio (OR) = 1.46, 95% confidence interval (CI) = 1.04-2.05). The joint effects of As, Pb, Tl and Ni levels on the odds of GDM was statistically significant when blood levels of four metals were exceeded their 50th percentile, with As level being a major contributor. Blood As level was positively associated with AUCG and the category of glucose latent profile, the values of AUCG were much higher in GDM group than those in non-GDM group, which suggested that As exposure associated with the odds of GDM may be due to that As exposure was related to the impairment of glucose tolerance among pregnant women. The significant and positive relationships of As level with AUCP, CP latent profile category, 2h CP and 2h CP/2h BG were observed, respectively; and the values of 1h CP/1h BG and AUCP/AUCG were much lower in GDM group than those in non-GDM group, which suggested that As exposure may not relate to the impairment of insulin secretion (pancreatic β-cell function) among pregnant women. The relationships between As level and 2h CP as well as 2h CP/2h BG were positive and significant; additionally, the values of 2h CP/2h BG in GDM group were comparable with those in non-GDM group; the peak value of CP occurred at 2h in GDM group, as well as the values of 2h CP/2h BG in high As exposure group were much higher than those in low As exposure group, which suggested that As exposure associated with the increased odds of GDM may be due to that As exposure was related to the change of insulin secretion phase (delayment of the peak of insulin secretion) among pregnant women. In addition, AUCP mediated 11% (p < 0.05) and AUCG mediated 43% (p < 0.05) of the association between As exposure and the odds of GDM. CONCLUSION Our results suggested that joint exposure to As, Pb, Tl and Ni during early pregnancy was positively associated with the odds of GDM, As was a major contributor; and the association of environmental As exposure with the increased odds of GDM may be due to that As exposure was related to the impairment of glucose tolerance and change of insulin secretion phase after glucose stimulation (delayment of the peak of insulin secretion) among pregnant women.
Collapse
Affiliation(s)
- Shitao He
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Tingting Jiang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Dongyang Zhang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Mengzhu Li
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Tao Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Muxin Zhai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Bingxia He
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Tao Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Xin Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Institute of Translational Medicine, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yuyou Yao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Dongmei Ji
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Institute of Translational Medicine, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Yuanyuan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Chunmei Liang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Institute of Translational Medicine, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
6
|
Lee KI, Fang KM, Kuo CY, Huang CF, Liu SH, Liu JM, Lai WC, Chang KC, Su CC, Chen YW. Roles of oxidative stress/JNK/ERK signals in paraquat-triggered hepatic apoptosis. Curr Res Toxicol 2024; 6:100155. [PMID: 38379848 PMCID: PMC10877118 DOI: 10.1016/j.crtox.2024.100155] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/18/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Paraquat (PQ), a toxic and nonselective bipyridyl herbicide, is one of the most extensively used pesticides in agricultural countries. In addition to pneumotoxicity, the liver is an important target organ for PQ poisoning in humans. However, the mechanism of PQ in hepatotoxicity remains unclear. In this study, we found that exposure of rat hepatic H4IIE cells to PQ (0.1-2 mM) induced significant cytotoxicity and apoptosis, which was accompanied by mitochondria-dependent apoptotic signals, including loss of mitochondrial membrane potential (MMP), cytosolic cytochrome c release, and changes in the Bcl-2/Bax mRNA ratio. Moreover, PQ (0.5 mM) exposure markedly induced JNK and ERK1/2 activation, but not p38-MAPK. Blockade of JNK and ERK1/2 signaling by pretreatment with the specific pharmacological inhibitors SP600125 and PD98059, respectively, effectively prevented PQ-induced cytotoxicity, mitochondrial dysfunction, and apoptotic events. Additionally, PQ exposure stimulated significant oxidative stress-related signals, including reactive oxygen species (ROS) generation and intracellular glutathione (GSH) depletion, which could be reversed by the antioxidant N-Acetylcysteine (NAC). Buffering the oxidative stress response with NAC also effectively abrogated PQ-induced hepatotoxicity, MMP loss, apoptosis, and phosphorylation of JNK and ERK1/2 protein, however, the JNK or ERK inhibitors did not suppress ROS generation in PQ-treated cells. Collectively, these results demonstrate that PQ exposure induces hepatic cell toxicity and death via an oxidative stress-dependent JNK/ERK activation-mediated downstream mitochondria-regulated apoptotic pathway.
Collapse
Affiliation(s)
- Kuan-I Lee
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Kai-Min Fang
- Department of Otolaryngology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Chun-Ying Kuo
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
| | - Chun-Fa Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Department of Nursing, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Jui-Ming Liu
- Department of Urology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330, Taiwan
| | - Wei-Cheng Lai
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Kai-Chih Chang
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Ya-Wen Chen
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
7
|
Alruhaimi RS, Hassanein EHM, Abd El-Aziz MK, Siddiq Abduh M, Bin-Ammar A, Kamel EM, Mahmoud AM. The melatonin receptor agonist agomelatine protects against acute pancreatitis induced by cadmium by attenuating inflammation and oxidative stress and modulating Nrf2/HO-1 pathway. Int Immunopharmacol 2023; 124:110833. [PMID: 37634447 DOI: 10.1016/j.intimp.2023.110833] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/29/2023]
Abstract
Pancreatitis is a serious effect of the heavy metal cadmium (Cd) and inflammation and oxidative stress (OS) are implicated in Cd-induced pancreatic injury. This study evaluated the effect of the melatonin receptor agonist agomelatine (AGM) on Cd-induced acute pancreatitis (AP), pointing to its modulatory effect on inflammation, OS, and Nrf2/HO-1 pathway. Rats were supplemented with AGM orally for 14 days and a single injection of cadmium chloride (CdCl2) on day 7. Cd increased serum amylase and lipase and caused pancreatic endocrine and exocrine tissue injury. Malondialdehyde (MDA), nitric oxide (NO) and myeloperoxidase (MPO) were elevated, nuclear factor (NF)-kB p65, inducible NO synthase (iNOS), interleukin (IL)-6, tumor necrosis factor (TNF)-α and CD40 were upregulated, and antioxidants were decreased in the pancreas of Cd-administered rats. AGM ameliorated serum amylase and lipase and pancreatic OS, NF-kB p65, CD40, pro-inflammatory mediators and caspase-3, prevented tissue injury and enhanced antioxidants. AGM downregulated Keap1 and enhanced Nrf2 and HO-1 in the pancreas of Cd-administered rats. In silico findings revealed the binding affinity of AGM with Keap1, HO-1, CD40L and caspase-3. In conclusion, AGM protected against AP induced by Cd by preventing inflammation, OS and apoptosis and modulating Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71562, Egypt
| | | | - Maisa Siddiq Abduh
- Immune Responses in Different Diseases Research Group, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Albandari Bin-Ammar
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Saudi Arabia
| | - Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK; Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
8
|
Lin X, Xu Y, Tong T, Zhang J, He H, Yang L, Deng P, Yu Z, Pi H, Hong H, Zhou Z. Cadmium exposure disturbs myocardial lipid signature and induces inflammation in C57BL/6J mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 265:115517. [PMID: 37776818 DOI: 10.1016/j.ecoenv.2023.115517] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/13/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023]
Abstract
Cadmium is a highly ubiquitous environmental pollutant that poses a serious threat to human health. In this study, we assessed the cardiotoxicity of Cd exposure and explored the possible mechanisms by which Cd exerts its toxic effects. The results demonstrated that exposure to Cd via drinking water containing CdCl2 10 mg/dL for eight consecutive weeks induced cardiac injury in C57BL/6J mice. The histopathological changes of myocardial hemolysis, widening of myocardial space, and fracture of myocardial fiber were observed. Meanwhile, elevated levels of cardiac enzyme markers and up-regulation of pro-apoptotic genes also indicated cardiac injury after Cd exposure. Non-targeted lipidomic analysis demonstrated that Cd exposure altered cardiac lipid metabolism, resulted in an increase in pro-inflammatory lipids, and changed lipid distribution abundance. In addition, Cd exposure affected the secretion of inflammatory cytokines by activating the NF-κB signaling pathway, leading to cardiac inflammation in mice. Taken together, results of our present study expand our understanding of Cd cardiotoxicity at the lipidomic level and provide new experimental evidence for uncovering the association of Cd exposure with cardiovascular diseases.
Collapse
Affiliation(s)
- Xiqin Lin
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yudong Xu
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Tong
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Zhang
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Haotian He
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingling Yang
- Department of Occupational Health, Army Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Army Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Army Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Army Medical University, Chongqing, China
| | - Huihui Hong
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China.
| | - Zhou Zhou
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China; Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
9
|
Martins AC, Ferrer B, Tinkov AA, Caito S, Deza-Ponzio R, Skalny AV, Bowman AB, Aschner M. Association between Heavy Metals, Metalloids and Metabolic Syndrome: New Insights and Approaches. TOXICS 2023; 11:670. [PMID: 37624175 PMCID: PMC10459190 DOI: 10.3390/toxics11080670] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023]
Abstract
Metabolic syndrome (MetS) is an important public health issue that affects millions of people around the world and is growing to pandemic-like proportions. This syndrome is defined by the World Health Organization (WHO) as a pathologic condition characterized by abdominal obesity, insulin resistance, hypertension, and hyperlipidemia. Moreover, the etiology of MetS is multifactorial, involving many environmental factors, including toxicant exposures. Several studies have associated MetS with heavy metals exposure, which is the focus of this review. Environmental and/or occupational exposure to heavy metals are a major risk, contributing to the development of chronic diseases. Of particular note, toxic metals such as mercury, lead, and cadmium may contribute to the development of MetS by altering oxidative stress, IL-6 signaling, apoptosis, altered lipoprotein metabolism, fluid shear stress and atherosclerosis, and other mechanisms. In this review, we discuss the known and potential roles of heavy metals in MetS etiology as well as potential targeted pathways that are associated with MetS. Furthermore, we describe how new approaches involving proteomic and transcriptome analysis, as well as bioinformatic tools, may help bring about an understanding of the involvement of heavy metals and metalloids in MetS.
Collapse
Affiliation(s)
- Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA; (A.C.M.)
| | - Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA; (A.C.M.)
| | - Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia; (A.A.T.)
- IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Samuel Caito
- School of Pharmacy, Husson University, Bangor, ME 04401, USA
| | - Romina Deza-Ponzio
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA; (A.C.M.)
| | - Anatoly V. Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia; (A.A.T.)
- IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA; (A.C.M.)
| |
Collapse
|
10
|
Saedi S, Watson SE, Young JL, Tan Y, Wintergerst KA, Cai L. Does maternal low-dose cadmium exposure increase the risk of offspring to develop metabolic syndrome and/or type 2 diabetes? Life Sci 2023; 315:121385. [PMID: 36634865 PMCID: PMC9912173 DOI: 10.1016/j.lfs.2023.121385] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/31/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
Cadmium is a hazardous metal with multiple organ toxicity that causes great harm to human health. Cadmium enters the human body through occupational exposure, diet, drinking water, breathing, and smoking. Cadmium accumulation in the human body is associated with increased risk of developing obesity, cardiovascular disease, diabetes, and metabolic syndrome (MetS). Cadmium uptake is enhanced during pregnancy and can cross the placenta affecting placental development and function. Subsequently, cadmium can pass to fetus, gathering in multiple organs such as the liver and pancreas. Early-life cadmium exposure can induce hepatic oxidative stress and pancreatic β-cell dysfunction, resulting in insulin resistance and glucose metabolic dyshomeostasis in the offspring. Prenatal exposure to cadmium is also associated with increasing epigenetic effects on the offspring's multi-organ functions. However, whether and how maternal exposure to low-dose cadmium impacts the risks of developing type 2 diabetes (T2D) in the young and/or adult offspring remains unclear. This review collected available data to address the current evidence for the potential role of cadmium exposure, leading to insulin resistance and the development of T2D in offspring. However, this review reveals that underlying mechanisms linking prenatal cadmium exposure during pregnancy with T2D in offspring remain to be adequately investigated.
Collapse
Affiliation(s)
- Saman Saedi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Sara E Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children's Hospital, Louisville, KY, USA
| | - Jamie L Young
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children's Hospital, Louisville, KY, USA; The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Wendy Novak Diabetes Institute, Norton Children's Hospital, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA; Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
11
|
Zhou M, Peng L, Wang J, Cao R, Ou Z, Fang Y. Cadmium exposure and the risk of GDM: evidence emerging from the systematic review and meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:77253-77274. [PMID: 35672642 DOI: 10.1007/s11356-022-21171-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Gestational diabetes mellitus (GDM) has become a global concern for its severe adverse effects on both mother and fetus. Recent epidemiological studies reported inconsistent results of the association between cadmium (Cd) exposure and GDM. Therefore, a systematic review and meta- analysis were performed. PubMed, Web of Science, Scopus, Embase, and SpringerLink were searched up to July 2021. Observational studies containing the adjusted relative risks between Cd exposure and GDM were included in the quantitative synthesis. The retrieval comprised 218 articles out of which 11 met our criteria and 9 were included in the meta-analysis, representing a total of 32,392 subjects (2881 GDM). In total, Cd exposure might increase the risk of GDM in some extent (OR = 1.21, 95% CI [0.89, 1.64]), even without statistical significance in high heterogeneity (Q = 28.45, p < 0.05, I2 = 71.9%). Filtering two outliers indicated by Galbraith plot yielded a similar risk (OR = 1.19, 95% CI [1.02, 1.39]) with statistical significance. However, the heterogeneity among studies was obviously reduced (Q = 11.75, p = 0.068, I2 = 48.9%). Additionally, biological specimen, study design, and diagnostic criteria contributed to the high heterogeneity according to the subgroup analysis. Since some important results do not deny that Cd exposure increases the risk of GDM, high-quality multi-centered large cohort studies are required in the future.
Collapse
Affiliation(s)
- Minqi Zhou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lianqi Peng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingming Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rong Cao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zixuan Ou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yiwei Fang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
12
|
Ma P, Sun C, Liu M, You H, Shen Y, Kang Y, Sun Y, Yang Z, Ma P, Yang L, Xue F. Metagenomic insights into the rumen epithelial integrity responses to the vitamin B1 supplement under high-concentrate diets treatments. Front Microbiol 2022; 13:1008373. [PMID: 36386689 PMCID: PMC9642323 DOI: 10.3389/fmicb.2022.1008373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
Subacute ruminal acidosis (SARA) becomes the most common nutritional metabolic disease in high-yielding dairy cows and later fatting beef cattle because of the increasing consumption of high-concentrate diets in modern feeding patterns. Our previous research found a certain piece of evidence that adding 180 mg thiamine/kg DMI could increase the rumen pH and regulate the structure of the rumen microbial community in vivo. However, there is still limited experimental data on the effects of SARA on thiamine status, the damage to the structure of rumen epithelial cells, and the underlying mechanism of the epithelium alterations. For this purpose, a total of 18 Angus bulls (average 22.0-months-old) with an average live weight of 567.6 ± 27.4 kg were randomly allocated into a control treatment (CON), a high-concentrate diet treatment (HC), and a high-concentrate diet with the vitamin B1 supplement treatment (HCB). All bulls were conducted with a 7-day adjustment period followed by a 60-day-long main feeding procedure. Results indicated that ADFI and ADG significantly decreased in the HC treatment compared with CON (P < 0.05), while significantly increased after the VB1 supplement (P < 0.05). Besides, ruminal acetate content was significantly downregulated while propionate was significantly upregulated under the HC treatment compared with CON (P < 0.05); however, these alterations showed a completely inverse regulatory effect on the VB1 supplement compared with HC (P < 0.05). These changes causatively induced a significant decrease in the A/P ratio in the HC treatment compared with CON and HCB treatments (P < 0.05). Bacterial communities in the HC treatment could be separated from those in CON through PCoA axes 1 and 2. Meanwhile, the VB1 supplement significantly altered the bacterial communities compared with the HC treatment, except for HCB-3. Furthermore, the HC treatment significantly upregulated the expression of JNK, Bax, Caspase-8, Caspase-3, Caspase-9, and Cyt-C compared with CON, while significantly downregulated the expression of Bcl-2. The VB1 supplement showed a complete converse gene expression compared with HC. In conclusion, the VB1 supplement could effectively attenuate the alterations that occurred when exposed to high-concentrate diets, and help promote production performance through increased fermentability.
Collapse
Affiliation(s)
- Peng Ma
- Nanchang Key Laboratory of Animal Health and Safety Production, Jiangxi Agricultural University, Nanchang, Jiangxi, China
- Anyou Biotechnology Group Co., Ltd., Taicang, Jiangsu, China
| | - Chaoqun Sun
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Minze Liu
- Yangxin Yiliyuan Halal Meat Co., Ltd., Yangxin, Shandong, China
| | - Hongnan You
- School of Foreign Language, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yao Shen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yajie Kang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuqin Sun
- Yangxin Yiliyuan Halal Meat Co., Ltd., Yangxin, Shandong, China
| | - Zhengang Yang
- Yangxin Yiliyuan Halal Meat Co., Ltd., Yangxin, Shandong, China
| | - Pengyun Ma
- Nanchang Key Laboratory of Animal Health and Safety Production, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Liang Yang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Fuguang Xue
- Nanchang Key Laboratory of Animal Health and Safety Production, Jiangxi Agricultural University, Nanchang, Jiangxi, China
- Yangxin Yiliyuan Halal Meat Co., Ltd., Yangxin, Shandong, China
- *Correspondence: Fuguang Xue,
| |
Collapse
|
13
|
The Molecular Mechanisms of Panax ginseng in Treating Type 2 Diabetes Mellitus: Network Pharmacology Analysis and Molecular Docking Validation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3082109. [PMID: 36159557 PMCID: PMC9507733 DOI: 10.1155/2022/3082109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/07/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022]
Abstract
Background Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder with a high global incidence. Panax ginseng has been used to treat T2DM in traditional medicine, with previous in vitro, in vivo, and clinical trial studies demonstrating its efficacy. This study aimed to determine the mechanism of P. ginseng in treating T2DM by network pharmacology. Methods The bioactive compounds of P. ginseng and corresponding targets of P. ginseng-T2DM were retrieved across multiple databases. The protein-protein interaction network was established using the STRING database and topological analysis helped identify the core target. Using the DAVID tool, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Finally, we checked the binding of core targets and bioactive compounds using molecular docking. Results The P. ginseng-T2DM networks mainly contained 22 bioactive compounds and 314 overlapping targets. The five most significant core targets were SRC, STAT3, MAPK1, AKT1, and PIK3R1. There were 244 GO terms and 95 KEGG pathways (adjusted p < 0.01) that were strongly correlated with diabetes-related signaling pathways, such as insulin resistance, the HIF-1 signaling pathway, the PI3K/Akt signaling pathway, the prolactin signaling pathway, the Rap1 signaling pathway, the Ras signaling pathway, the calcium signaling pathway, and the FoxO signaling pathway. Molecular docking results revealed that the top five core targets had a high binding affinity with the bioactive compounds of P. ginseng. Conclusion The bioactive compounds and targets in P. ginseng ameliorate T2DM by regulating insulin resistance and multiple signaling pathways.
Collapse
|
14
|
Wong WPS, Wang JC, Meyers MS, Wang NJ, Sponenburg RA, Allen NB, Edwards JR, El Muayed M. A novel chronic in vivo oral cadmium exposure-washout mouse model for studying cadmium toxicity and complex diabetogenic effects. Toxicol Appl Pharmacol 2022; 447:116057. [PMID: 35550884 PMCID: PMC9854171 DOI: 10.1016/j.taap.2022.116057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 01/25/2023]
Abstract
Type II diabetes mellitus (T2DM) is characterized by insulin resistance, β-cell dysfunction and hyperglycemia. In addition to well known risk factors such as lifestyle and genetic risk score, accumulation of environmental toxicants in organs relevant to glucose metabolism is increasingly recognized as additional risk factors for T2DM. Here, we describe the development of an in vivo oral cadmium (Cd) exposure model. It was shown that oral Cd exposure in drinking water followed by washout and high fat diet (HFD) in C57BL/6N mice results in islet Cd bioaccumulation comparable to that found in native human islets while mitigating the anorexic effects of Cd to achieve the same weight gain required to induce insulin resistance as in Cd naïve control mice. Inter individual variation in plasma glucose and insulin levels as well as islet Cd bioaccumulation was observed in both female and male mice. Regression analysis showed an inverse correlation between islet Cd level and plasma insulin following a glucose challenge in males but not in females. This finding highlights the need to account for inter individual target tissue Cd concentrations when interpreting results from in vivo Cd exposure models. No effect of Cd on insulin secretion was observed in islets ex vivo, highlighting differences between in vivo and ex vivo cadmium exposure models. In summary, our oral in vivo Cd exposure-washout with HFD model resulted in islet Cd bioaccumulation that is relevant in the context of environmental cadmium exposure in humans. Here, we showed that islet Cd bioaccumulation is associated with complex cadmium-mediated changes in glucose clearance and β-cell function. The model described here will serve as a useful tool to further examine the relationship between Cd exposure, islet Cd bioaccumulation, dysglycemia and their underlying mechanisms.
Collapse
Affiliation(s)
- Winifred P S Wong
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Janice C Wang
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Matthew S Meyers
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nathan J Wang
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rebecca A Sponenburg
- Quantitative Bio-element Imaging Centre, Chemistry of Life Processes, Northwestern University, Evanston, IL 60208, USA
| | - Norrina B Allen
- Institute for Public Health and Medicine, Center for Epidemiology and Population Health, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Joshua R Edwards
- College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Malek El Muayed
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
15
|
Hong H, He H, Lin X, Hayuehashi T, Xu J, Zhang J, Xu Y, Tong T, Lu Y, Zhou Z. Cadmium exposure suppresses insulin secretion through mtROS-mediated mitochondrial dysfunction and inflammatory response in pancreatic beta cells. J Trace Elem Med Biol 2022; 71:126952. [PMID: 35183883 DOI: 10.1016/j.jtemb.2022.126952] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cadmium (Cd) exposure is a worldwide environmental threat to the public health and participates in the pathogenesis of multiple diseases. Epidemiologic research have established a direct relation between Cd exposure and diabetes development in humans. Although pancreatic β-cell dysfunction has been considered as the major culprit in the pathogenesis of diabetes, there is a paucity of studies to elucidate the molecular mechanism of Cd toxicity on β-cells. METHODS To unveil the toxic effect and its underlying mechanism of Cd exposure on β-cells, we used an in vitro MIN6 cell model of environment-relevant Cd exposure to elucidate the crucial role of mtROS-mediated mitochondrial dysfunction and inflammatory response in suppression of pancreatic β-cell insulin secretion. RESULTS We uncovered that Cd treatment suppresses cell viability and induces insulin secretion dysfunction in a dose-dependent manner. Moreover, Cd exposure elicits the inflammatory response, as indicated by increased IL-1β, IL-6 and TNF-α expressions. Significant elevations of intracellular ROS and mitochondrial ROS levels were detected as early as 3 h after Cd treatment. In mitochondrial function analysis, we demonstrated that Cd treatment induced mitochondrial dysfunction and disorder of mitochondrial fission indicated by the significant decline in ATP production, the marked depolarization of mitochondrial membrane potential, the decrease in mtDNA copy numbers, the suppressions of mitochondrial transcription factor A (Tfam) and mitochondrial fission-related gene Drp1 expressions. Pretreatment with TEMPO, a specific mitochondrial ROS (mtROS) scavenger, efficiently antagonizes Cd cytotoxicity, which is indicated by attenuating Cd-induced mitochondrial dysfunction, suppressing IL-1β, IL-6 and TNF-α expressions, ameliorating insulin production dysfunction and preserving cell viability in MIN6 cells. CONCLUSION Our study demonstrates that Cd exposure induces an inflammatory response through mtROS-mediated mitochondrial dysfunction. Antagonism of mtROS production might be an effective strategy to prevent pancreatic toxicity from environment-relevant Cd exposure.
Collapse
Affiliation(s)
- Huihui Hong
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haotian He
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiqin Lin
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tali Hayuehashi
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Xu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou, China
| | - Jingjing Zhang
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yudong Xu
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Tong
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanqiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou, China.
| | - Zhou Zhou
- Department of Environmental Medicine and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
16
|
Norketamine, the Main Metabolite of Ketamine, Induces Mitochondria-Dependent and ER Stress-Triggered Apoptotic Death in Urothelial Cells via a Ca2+-Regulated ERK1/2-Activating Pathway. Int J Mol Sci 2022; 23:ijms23094666. [PMID: 35563057 PMCID: PMC9102902 DOI: 10.3390/ijms23094666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 02/01/2023] Open
Abstract
Ketamine-associated cystitis is characterized by suburothelial inflammation and urothelial cell death. Norketamine (NK), the main metabolite of ketamine, is abundant in urine following ketamine exposure. NK has been speculated to exert toxic effects in urothelial cells, similarly to ketamine. However, the molecular mechanisms contributing to NK-induced urothelial cytotoxicity are almost unclear. Here, we aimed to investigate the toxic effects of NK and the potential mechanisms underlying NK-induced urothelial cell injury. In this study, NK exposure significantly reduced cell viability and induced apoptosis in human urinary bladder epithelial-derived RT4 cells that NK (0.01–0.5 mM) exhibited greater cytotoxicity than ketamine (0.1–3 mM). Signals of mitochondrial dysfunction, including mitochondrial membrane potential (MMP) loss and cytosolic cytochrome c release, were found to be involved in NK-induced cell apoptosis and death. NK exposure of cells also triggered the expression of endoplasmic reticulum (ER) stress-related proteins including GRP78, CHOP, XBP-1, ATF-4 and -6, caspase-12, PERK, eIF-2α, and IRE-1. Pretreatment with 4-phenylbutyric acid (an ER stress inhibitor) markedly prevented the expression of ER stress-related proteins and apoptotic events in NK-exposed cells. Additionally, NK exposure significantly activated JNK, ERK1/2, and p38 signaling and increased intracellular calcium concentrations ([Ca2+]i). Pretreatment of cells with both PD98059 (an ERK1/2 inhibitor) and BAPTA/AM (a cell-permeable Ca2+ chelator), but not SP600125 (a JNK inhibitor) and SB203580 (a p38 inhibitor), effectively suppressed NK-induced mitochondrial dysfunction, ER stress-related signals, and apoptotic events. The elevation of [Ca2+]i in NK-exposed cells could be obviously inhibited by BAPTA/AM, but not PD98059. Taken together, these findings suggest that NK exposure exerts urothelial cytotoxicity via a [Ca2+]i-regulated ERK1/2 activation, which is involved in downstream mediation of the mitochondria-dependent and ER stress-triggered apoptotic pathway, consequently resulting in urothelial cell death. Our findings suggest that regulating [Ca2+]i/ERK signaling pathways may be a promising strategy for treatment of NK-induced urothelial cystitis.
Collapse
|
17
|
Li M, Wang S, Liu X, Sheng Z, Li B, Li J, Zhang J, Zhang Z. Cadmium exposure decreases fasting blood glucose levels and exacerbates type-2 diabetes in a mouse model. Endocrine 2022; 76:53-61. [PMID: 35041127 DOI: 10.1007/s12020-021-02974-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/22/2021] [Indexed: 01/12/2023]
Abstract
PURPOSE Although the effects of cadmium (Cd) on the development of diabetes have been extensively investigated, the relationship between Cd exposure and the severity of established diabetes is unclear. Herein, we investigate the effects of long-term exposure to Cd in a streptozotocin-induced mouse model of type-2 diabetes mellitus (T2DM) and the underlying mechanism. METHODS C57BL/6 Mice were divided into the following four groups: (1) control group; (2) Cd-exposed group; (3) diabetic group; (4) Cd-exposed diabetic group. Cd exposure was established by the administration of 155 ppm CdCl2 in drinking water. After 25 weeks of treatment, serum fasting glucose and insulin were measured. Meanwhile, the liver and pancreas specimens were sectioned and stained with Hematoxylin and eosin. Gluconeogenesis, glycolysis, lactate concentration, and fibrosis in liver were evaluated. RESULTS Clinical signs attributable to diabetes were more apparent in Cd-exposed diabetic mice, while no effects of Cd exposure were found on non-diabetic mice. Cd exposure significantly decreased fasting blood glucose (FBG) levels in diabetic group. We further demonstrated that the glycolysis related hepatic enzymes, pyruvate kinase M2 (PKM-2) and lactic dehydrogenase A (LDHA) were both increased, while the gluconeogenesis related hepatic enzymes, phosphoenolpyruvate-1 (PCK-1) and glucose-6-phosphatase (G6Pase) were both decreased in Cd exposed diabetic mice, indicating that Cd increased glycolysis and inhibited gluconeogenesis in diabetic model. Moreover, lactate accumulation was noted accompanied by the increased inflammation and fibrosis in the livers of diabetic mice following Cd exposure. CONCLUSIONS Cd exposure disturbed glucose metabolism and exacerbated diabetes, providing a biological relevance that DM patients are at greater risk when exposed to Cd.
Collapse
Affiliation(s)
- Mengyang Li
- School of Public Health, Soochow University, 215123, Suzhou, China
| | - Shuai Wang
- The Shishan Community Hospital of SND in Suzhou, 215011, Suzhou, China
| | - Xiuxiu Liu
- School of Public Health, Soochow University, 215123, Suzhou, China
| | - Zhijie Sheng
- The Fifth People's Hospital in Suzhou, 215007, Suzhou, China
| | - Bingyan Li
- School of Public Health, Soochow University, 215123, Suzhou, China
| | - Jiafu Li
- School of Public Health, Soochow University, 215123, Suzhou, China
| | - Jie Zhang
- School of Public Health, Soochow University, 215123, Suzhou, China.
| | - Zengli Zhang
- School of Public Health, Soochow University, 215123, Suzhou, China.
| |
Collapse
|
18
|
Yang CY, Liu SH, Su CC, Fang KM, Yang TY, Liu JM, Chen YW, Chang KC, Chuang HL, Wu CT, Lee KI, Huang CF. Methylmercury Induces Mitochondria- and Endoplasmic Reticulum Stress-Dependent Pancreatic β-Cell Apoptosis via an Oxidative Stress-Mediated JNK Signaling Pathway. Int J Mol Sci 2022; 23:2858. [PMID: 35270009 PMCID: PMC8910963 DOI: 10.3390/ijms23052858] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Methylmercury (MeHg), a long-lasting organic pollutant, is known to induce cytotoxic effects in mammalian cells. Epidemiological studies have suggested that environmental exposure to MeHg is linked to the development of diabetes mellitus (DM). The exact molecular mechanism of MeHg-induced pancreatic β-cell cytotoxicity is still unclear. Here, we found that MeHg (1-4 μM) significantly decreased insulin secretion and cell viability in pancreatic β-cell-derived RIN-m5F cells. A concomitant elevation of mitochondrial-dependent apoptotic events was observed, including decreased mitochondrial membrane potential and increased proapoptotic (Bax, Bak, p53)/antiapoptotic (Bcl-2) mRNA ratio, cytochrome c release, annexin V-Cy3 binding, caspase-3 activity, and caspase-3/-7/-9 activation. Exposure of RIN-m5F cells to MeHg (2 μM) also induced protein expression of endoplasmic reticulum (ER) stress-related signaling molecules, including C/EBP homologous protein (CHOP), X-box binding protein (XBP-1), and caspase-12. Pretreatment with 4-phenylbutyric acid (4-PBA; an ER stress inhibitor) and specific siRNAs for CHOP and XBP-1 significantly inhibited their expression and caspase-3/-12 activation in MeHg-exposed RIN-mF cells. MeHg could also evoke c-Jun N-terminal kinase (JNK) activation and reactive oxygen species (ROS) generation. Antioxidant N-acetylcysteine (NAC; 1mM) or 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (trolox; 100 μM) markedly prevented MeH-induced ROS generation and decreased cell viability in RIN-m5F cells. Furthermore, pretreatment of cells with SP600125 (JNK inhibitor; 10 μM) or NAC (1 mM) or transfection with JNK-specific siRNA obviously attenuated the MeHg-induced JNK phosphorylation, CHOP and XBP-1 protein expression, apoptotic events, and insulin secretion dysfunction. NAC significantly inhibited MeHg-activated JNK signaling, but SP600125 could not effectively reduce MeHg-induced ROS generation. Collectively, these findings demonstrate that the induction of ROS-activated JNK signaling is a crucial mechanism underlying MeHg-induced mitochondria- and ER stress-dependent apoptosis, ultimately leading to β-cell death.
Collapse
Affiliation(s)
- Ching-Yao Yang
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan;
- Department of Surgery, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan;
| | - Kai-Min Fang
- Department of Otolaryngology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
| | - Tsung-Yuan Yang
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
- School of Medicine, Institute of Medicine, Chung-Shan Medical University, Taichung 402, Taiwan
| | - Jui-Ming Liu
- Department of Urology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330, Taiwan;
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ya-Wen Chen
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Kai-Chih Chang
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan;
| | - Haw-Ling Chuang
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan;
| | - Cheng-Tien Wu
- Department of Nutrition and Master Program of Food and Drug Safety, China Medical University, Taichung 40402, Taiwan;
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan;
| | - Chun-Fa Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Department of Nursing, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| |
Collapse
|
19
|
García-Aguilar A, Guillén C. Targeting pancreatic beta cell death in type 2 diabetes by polyphenols. Front Endocrinol (Lausanne) 2022; 13:1052317. [PMID: 36465657 PMCID: PMC9712222 DOI: 10.3389/fendo.2022.1052317] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes is a very complex disease which is characterized by the appearance of insulin resistance that is primarily compensated by an increase in pancreatic beta cell mass, generating hyperinsulinemia. After time, pancreatic beta cells die by apoptosis appearing in the second phase of the disease, and characterized by hypoinsulinemia. There are multiple conditions that can alter pancreatic beta cell homeostasis and viability, being the most relevant ones; ER stress, cytotoxicity by amylin, mTORC1 hyperactivity, oxidative stress, mitochondrial dysfunction, inflammation and alterations in autophagy/mitophagy flux. In addition, the possible effects that different polyphenols could exert in the modulation of these mechanisms and regulating pancreatic beta cell viability are analyzed. It is necessary a profound analysis and understanding of all the possible mechanisms involved in the control and maintenance of pancreatic beta cell viability to develop more accurate and target treatments for controlling beta cell homeostasis and preventing or even reversing type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ana García-Aguilar
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Diabetes and Associated Metabolic Diseases Networking Biomedical Research Centre Centro de Investigación Biomédica en Red. Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Guillén
- Diabetes and Associated Metabolic Diseases Networking Biomedical Research Centre Centro de Investigación Biomédica en Red. Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- *Correspondence: Carlos Guillén,
| |
Collapse
|
20
|
Dai SD, Wang S, Qin YN, Zhu JC. Multiomics Landscape Uncovers the Molecular Mechanism of the Malignant Evolution of Lung Adenocarcinoma Cells to Chronic Low Dose Cadmium Exposure. Front Oncol 2021; 11:654687. [PMID: 34858801 PMCID: PMC8631903 DOI: 10.3389/fonc.2021.654687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022] Open
Abstract
Cadmium (Cd) from cigarette smoke and polluted air can lead to lung adenocarcinoma after long-term inhalation. However, most studies are based on short-term exposure to this toxic metal at high concentrations. Here, we investigate the effects of long-term exposure of A549 cells (lung adenocarcinoma) to cadmium at low concentrations using morphological and multiomics analyses. First, we treated A549 cells continuously with CdCl2 at 1μM for 8 months and found that CdCl2 promoted cellular migration and invasion. After that, we applied transmission electron and fluorescence microscopies and did not observe significant morphological changes in Golgi apparatus, endoplasmic reticulum, lysosomes, or mitochondria on Cd treated cells; microfilaments, in contrast, accumulated in lamellipodium and adhesion plaques, which suggested that Cd enhanced cellular activity. Second, by using whole-exome sequencing (WES) we detected 4222 unique SNPs in Cd-treated cells, which included 382 unique non-synonymous mutation sites. The corresponding mutated genes, after GO and KEGG enrichments, were involved mainly in cell adhesion, movement, and metabolic pathways. Third, by RNA-seq analysis, we showed that 1250 genes (784 up and 466 down), 1623 mRNAs (1023 up and 591 down), and 679 lncRNAs (375 up and 304 down) were expressed differently. Furthermore, GO enrichment of these RNA-seq results suggested that most differentially expressed genes were related to cell adhesion and organization of the extracellular matrix in biological process terms; KEGG enrichment revealed that the differentially expressed genes took part in 26 pathways, among which the metabolic pathway was the most significant. These findings could be important for unveiling mechanisms of Cd-related cancers and for developing cancer therapies in the future.
Collapse
Affiliation(s)
- Shun-Dong Dai
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Shuang Wang
- Department of Pathology, Shenyang Red Cross Hospital, Shenyang, China
| | - Ya-Nan Qin
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jin-Chao Zhu
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
An W, Huang Y, Chen S, Teng T, Shi Y, Sun Z, Xu Y. Mechanisms of Rhizoma Coptidis against type 2 diabetes mellitus explored by network pharmacology combined with molecular docking and experimental validation. Sci Rep 2021; 11:20849. [PMID: 34675276 PMCID: PMC8531350 DOI: 10.1038/s41598-021-00293-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/05/2021] [Indexed: 02/08/2023] Open
Abstract
This study systematically explored the underlying mechanism of Rhizoma Coptidis against type 2 diabetes mellitus (T2DM) by using network pharmacology and molecular docking and experimental validation. We retrieved and screened active compounds of Rhizoma Coptidis and corresponding T2DM-related targets across multiple databases. PPI networks of the genes were constructed using STRING, and the core targets were screened via topological analysis. GO and KEGG enrichment analyses were performed by using DAVID. Finally, molecular docking and experimental studies were performed after bioinformatic analysis for verification. There were 14 active compounds and 19 core targets of Rhizoma Coptidis-T2DM, of which quercetin was identified as the main compound and IL6, VEGFA and TNF were the most significant core targets. GO and KEGG enrichment analyses showed that Rhizoma Coptidis ameliorated T2DM by regulating multiple biological processes and pathways. Docking studies indicated that IL6, VEGFA and TNF could stably bind with all active compounds of Rhizoma Coptidis. The results of our experiments revealed that Rhizoma Coptidis could inhibit the expression of IL6 and TNFα and enhance islet cell viability. This study suggests anti-inflammatory therapeutic effects of Rhizoma Coptidis on T2DM, thereby providing a scientific basis and new insight for further research on the antidiabetic effect of Rhizoma Coptidis.
Collapse
Affiliation(s)
- Wenrong An
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yanqin Huang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Shouqiang Chen
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 1 Jingba Road, Jinan, 250001, China
| | - Tao Teng
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 1 Jingba Road, Jinan, 250001, China
| | - Yingning Shi
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhenhai Sun
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 1 Jingba Road, Jinan, 250001, China.
| |
Collapse
|
22
|
Jackson TW, Ryherd GL, Scheibly CM, Sasser AL, Guillette TC, Belcher SM. Gestational Cd Exposure in the CD-1 Mouse Induces Sex-Specific Hepatic Insulin Insensitivity, Obesity, and Metabolic Syndrome in Adult Female Offspring. Toxicol Sci 2021; 178:264-280. [PMID: 33259630 DOI: 10.1093/toxsci/kfaa154] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There is compelling evidence that developmental exposure to toxic metals increases risk for obesity and obesity-related morbidity including cardiovascular disease and type 2 diabetes. To explore the hypothesis that developmental Cd exposure increases risk of obesity later in life, male, and female CD-1 mice were maternally exposed to 500 ppb CdCl2 in drinking water during a human gestational equivalent period (gestational day 0-postnatal day 10 [GD0-PND10]). Hallmark indicators of metabolic disruption, hepatic steatosis, and metabolic syndrome were evaluated prior to birth through adulthood. Maternal blood Cd levels were similar to those observed in human pregnancy cohorts, and Cd was undetected in adult offspring. There were no observed impacts of exposure on dams or pregnancy-related outcomes. Results of glucose and insulin tolerance testing revealed that Cd exposure impaired offspring glucose homeostasis on PND42. Exposure-related increases in circulating triglycerides and hepatic steatosis were apparent only in females. By PND120, Cd-exposed females were 30% heavier with 700% more perigonadal fat than unexposed control females. There was no evidence of dyslipidemia, steatosis, increased weight gain, nor increased adiposity in Cd-exposed male offspring. Hepatic transcriptome analysis on PND1, PND21, and PND42 revealed evidence for female-specific increases in oxidative stress and mitochondrial dysfunction with significant early disruption of retinoic acid signaling and altered insulin receptor signaling consistent with hepatic insulin sensitivity in adult females. The observed steatosis and metabolic syndrome-like phenotypes resulting from exposure to 500 ppb CdCl2 during the pre- and perinatal period of development equivalent to human gestation indicate that Cd acts developmentally as a sex-specific delayed obesogen.
Collapse
Affiliation(s)
- Thomas W Jackson
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| | - Garret L Ryherd
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| | - Chris M Scheibly
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| | - Aubrey L Sasser
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| | - T C Guillette
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| | - Scott M Belcher
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
23
|
Salcedo-Bellido I, Gómez-Peña C, Pérez-Carrascosa FM, Vrhovnik P, Mustieles V, Echeverría R, Fiket Ž, Pérez-Díaz C, Barrios-Rodríguez R, Jiménez-Moleón JJ, Arrebola JP. Adipose tissue cadmium concentrations as a potential risk factor for insulin resistance and future type 2 diabetes mellitus in GraMo adult cohort. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 780:146359. [PMID: 34030321 DOI: 10.1016/j.scitotenv.2021.146359] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 06/12/2023]
Abstract
Adipose tissue has been recently highlighted as a promising matrix for evaluation of cadmium's (Cd) long-term exposure although not frequently considered in epidemiological studies. The association between Cd exposure and type 2 Diabetes Mellitus (T2DM) remains unclear. This work aimed to explore the association between adipose tissue Cd levels and T2DM incidence over a 16-year follow-up in an adult cohort from Southern Spain considering smoking status. We also performed complementary cross-sectional analyses focused on subclinical markers of glucose homeostasis at recruitment. Clinical information was obtained from hospital databases. Socio-demographic characteristics, lifestyle and diet were collected by face-to-face interviews. Homeostatic model assessment (HOMA) values of insulin sensitivity/resistance and β-cell function were calculated using fasting serum glucose, insulin, and C-peptide levels at recruitment. Adipose tissue Cd concentrations were quantified by inductively coupled plasma mass spectrometry. Statistical analyses were performed by means of Cox-regression and multivariable linear regression models. Participants in the 4th quartile (Q4) of Cd concentrations showed a non statistically-significant increased T2DM risk (Hazard Ratio (HR) Q4 vs Q1: 1.97; 95% Confidence Interval (CI): 0.69, 5.66). This association was particularly strong and suggestive in current smokers (HR: 2.19; 95% CI: 0.98, 4.98). Interestingly, smokers in the 2nd tertile (T2) of adipose tissue Cd levels showed increased log-transformed insulin resistance (beta T2 vs T1: 0.52; 95% CI: 0.07, 0.97), as well as higher log-transformed insulin levels (beta T2 vs T1: 0.52; 95% CI: 0.08, 0.95). We found evidences supporting that Cd exposure, particularly from tobacco smoking, could be a risk factor for T2DM. In addition, our results support the potential relevance of adipose tissue as a matrix for Cd exposure assessment.
Collapse
Affiliation(s)
- Inmaculada Salcedo-Bellido
- Universidad de Granada. Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain
| | - Celia Gómez-Peña
- Unidad de Gestión Clínica de Farmacia Hospitalaria, Hospital Universitario San Cecilio, Granada, Spain
| | - Francisco M Pérez-Carrascosa
- Universidad de Granada. Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain; Radiation Oncology Department, Virgen de las Nieves University Hospital, Granada, Spain
| | - Petra Vrhovnik
- Slovenian National Building and Civil Engineering Institute (ZAG), Ljubljana, Slovenia
| | - Vicente Mustieles
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain; University of Granada, Center for Biomedical Research (CIBM), Granada, Spain
| | - Ruth Echeverría
- Universidad de Granada. Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
| | - Željka Fiket
- Ruđer Bošković Institute, Division for Marine and Environmental Research, Zagreb, Croatia
| | - Celia Pérez-Díaz
- Universidad de Granada. Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
| | - Rocío Barrios-Rodríguez
- Universidad de Granada. Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain.
| | - José Juan Jiménez-Moleón
- Universidad de Granada. Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain
| | - Juan Pedro Arrebola
- Universidad de Granada. Departamento de Medicina Preventiva y Salud Pública, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain.
| |
Collapse
|
24
|
Huang CC, Yang CY, Su CC, Fang KM, Yen CC, Lin CT, Liu JM, Lee KI, Chen YW, Liu SH, Huang CF. 4-Methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene, a Major Active Metabolite of Bisphenol A, Triggers Pancreatic β-Cell Death via a JNK/AMPKα Activation-Regulated Endoplasmic Reticulum Stress-Mediated Apoptotic Pathway. Int J Mol Sci 2021; 22:ijms22094379. [PMID: 33922211 PMCID: PMC8122752 DOI: 10.3390/ijms22094379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), a major active metabolite of bisphenol A (BPA), is generated in the mammalian liver. Some studies have suggested that MBP exerts greater toxicity than BPA. However, the mechanism underlying MBP-induced pancreatic β-cell cytotoxicity remains largely unclear. This study demonstrated the cytotoxicity of MBP in pancreatic β-cells and elucidated the cellular mechanism involved in MBP-induced β-cell death. Our results showed that MBP exposure significantly reduced cell viability, caused insulin secretion dysfunction, and induced apoptotic events including increased caspase-3 activity and the expression of active forms of caspase-3/-7/-9 and PARP protein. In addition, MBP triggered endoplasmic reticulum (ER) stress, as indicated by the upregulation of GRP 78, CHOP, and cleaved caspase-12 proteins. Pretreatment with 4-phenylbutyric acid (4-PBA; a pharmacological inhibitor of ER stress) markedly reversed MBP-induced ER stress and apoptosis-related signals. Furthermore, exposure to MBP significantly induced the protein phosphorylation of JNK and AMP-activated protein kinase (AMPK)α. Pretreatment of β-cells with pharmacological inhibitors for JNK (SP600125) and AMPK (compound C), respectively, effectively abrogated the MBP-induced apoptosis-related signals. Both JNK and AMPK inhibitors also suppressed the MBP-induced activation of JNK and AMPKα and of each other. In conclusion, these findings suggest that MBP exposure exerts cytotoxicity on β-cells via the interdependent activation of JNK and AMPKα, which regulates the downstream apoptotic signaling pathway.
Collapse
Affiliation(s)
- Cheng-Chin Huang
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan; (C.-C.H.); (J.-M.L.); (K.-IL.)
| | - Ching-Yao Yang
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan;
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kai-Min Fang
- Department of Otolaryngology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
| | - Cheng-Chieh Yen
- Department of Occupational Safety and Health, College of Health Care and Management, Chung Shan Medical University, Taichung 402, Taiwan;
| | - Ching-Ting Lin
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
| | - Jui-Min Liu
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan; (C.-C.H.); (J.-M.L.); (K.-IL.)
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan; (C.-C.H.); (J.-M.L.); (K.-IL.)
| | - Ya-Wen Chen
- Department of Physiology and Graduate Institute of Basic Medical Science, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan;
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Correspondence: (S.-H.L.); (C.-F.H.)
| | - Chun-Fa Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
- Department of Nursing, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
- Correspondence: (S.-H.L.); (C.-F.H.)
| |
Collapse
|
25
|
Hong H, Xu Y, Xu J, Zhang J, Xi Y, Pi H, Yang L, Yu Z, Wu Q, Meng Z, Ruan WS, Ren Y, Xu S, Lu YQ, Zhou Z. Cadmium exposure impairs pancreatic β-cell function and exaggerates diabetes by disrupting lipid metabolism. ENVIRONMENT INTERNATIONAL 2021; 149:106406. [PMID: 33508533 DOI: 10.1016/j.envint.2021.106406] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/23/2020] [Accepted: 01/13/2021] [Indexed: 06/12/2023]
Abstract
Cadmium is known as an environmental pollutant that contributes to pancreatic damage and the pathogenesis of diabetes. However, less attention has been devoted to elucidating the mechanisms underlying Cd-induced pancreatic β-cell dysfunction and the role of Cd toxicity in the development of diabetes. In this study, we demonstrated that exposure to Cd caused remarkable pancreatic β-cell dysfunction and death, both in vitro and in vivo. Lipidomic analysis of Cd-exposed pancreatic β-cells using high-resolution mass spectrometry revealed that Cd exposure altered the profile and abundance of lipids. Cd exposure induced intracellular lipid accumulation, promoted lipid biogenesis, elevated pro-inflammatory lipid contents and inhibited lipid degradation. Furthermore, Cd exposure upregulated the expression levels of TNF-α, IL-1β and IL-6 in pancreatic β-cells and elevated the TNF-α, IL1-β and IL-6 levels in the serum and pancreas. Taken together, the results of our study demonstrated that environmental relevant Cd exposure causes pro-inflammatory lipids elevation and insulin secretion dysfunction in β-cells and hence exaggerates diabetes development. Combined exposure to environmental hazardous chemicals might markedly increase the probability of developing diabetes in humans. This study provides new metabolic and pharmacological targets for antagonizing Cd toxicity.
Collapse
Affiliation(s)
- Huihui Hong
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yudong Xu
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Xu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, Hangzhou, China
| | - Jingjing Zhang
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Xi
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huifeng Pi
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Qingqian Wu
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhuoxian Meng
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei-Shuyi Ruan
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, Hangzhou, China
| | - Yunzhao Ren
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuzhen Xu
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan-Qiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, Hangzhou, China.
| | - Zhou Zhou
- Department of Environmental Medicine, and Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
26
|
Ye Y, Li X, Wang Z, Ye F, Xu W, Lu R, Shen H, Miao S. 3,3'-Diindolylmethane induces gastric cancer cells death via STIM1 mediated store-operated calcium entry. Int J Biol Sci 2021; 17:1217-1233. [PMID: 33867841 PMCID: PMC8040462 DOI: 10.7150/ijbs.56833] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
3,3'-Diindolylmethane (DIM), a natural phytochemicals isolated from cruciferous vegetables, has been reported to inhibit human gastric cancer cells proliferation and induce cells apoptosis as well as autophagy, but its mechanisms are still unclear. Store-operated calcium entry (SOCE) is a main Ca2+ influx pathway in various of cancers, which is activated by the depletion of endoplasmic reticulum (ER) Ca2+ store. Stromal interaction molecular 1 (STIM1) is the necessary component of SOCE. In this study, we focus on to examine the regulatory mechanism of SOCE on DIM-induced death in gastric cancer. After treating the human BGC-823 and SGC-7901 gastric cancer cells with DIM, cellular proliferation was determined by MTT, apoptosis and autophagy were detected by flow cytometry or Hoechst 33342 staining. The expression levels of related proteins were evaluated by Western blotting. Free cytosolilc Ca2+ level was assessed by fluorescence monitoring under a laser scanning confocal microscope. The data have shown that DIM could significantly inhibit proliferation and induce apoptosis as well as autophagy in two gastric cancer cell lines. After DIM treatment, the STIM1-mediated SOCE was activated by upregulating STIM1 and decreasing ER Ca2+ level. Knockdown STIM1 with siRNA or pharmacological inhibition of SOCE attenuated DIM induced apoptosis and autophagy by inhibiting p-AMPK mediated ER stress pathway. Our data highlighted that the potential of SOCE as a promising target for treating cancers. Developing effective and selective activators targeting STIM1-mediated SOCE pathway will facilitate better therapeutic sensitivity of phytochemicals acting on SOCE in gastric cancer. Moreover, more research should be performed to validate the efficacy of combination chemotherapy of anti-cancer drugs targeting SOCE for clinical application.
Collapse
Affiliation(s)
- Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xue Li
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhihua Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Fen Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
- Department of Clinical Laboratory Center, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
- Center for Experimental Research, Affiliated Kunshan Hospital to Jiangsu University School of Medicine, Kunshan, Suzhou, China
| | - Haijun Shen
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shuhan Miao
- Department of Health Care, Zhenjiang Fourth Peoples Hospital, Zhenjiang, China
| |
Collapse
|
27
|
Lewandowski Ł, Urbanowicz I, Kepinska M, Milnerowicz H. Concentration/activity of superoxide dismutase isozymes and the pro-/antioxidative status, in context of type 2 diabetes and selected single nucleotide polymorphisms (genes: INS, SOD1, SOD2, SOD3) - Preliminary findings. Biomed Pharmacother 2021; 137:111396. [PMID: 33761612 DOI: 10.1016/j.biopha.2021.111396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 11/29/2022] Open
Abstract
The alterations in concentration/activity of superoxide dismutase isozymes in the context of type 2 diabetes or obesity are well-described. Moreover, many hereditary factors, including single-nucleotide polymorphisms (SNPs) of genes for coding insulin, insulin receptors, or insulin receptor substrates (INS, INSR, IRS1, IRS2) or superoxide dismutase isozymes (SOD1, SOD2, SOD3), have been linked with the incidence of obesity and diabetes. However, the underlying changes in the plasma concentration/activity of superoxide dismutase isozymes and their potential connection with the said hereditary factors remain unexplored. Previously, we have observed that the plasma concentration/activity of superoxide dismutase isozymes differs in the context of obesity and/or rs2234694 (SOD1) and rs4880 (SOD2) and that the concentrations of SOD1, SOD2, SOD3 are correlated with each other. Intersexual variability of SOD1 concentration was detected regardless of obesity. In this study, the variability of concentration/activity of superoxide dismutase isozymes in plasma is considered in the context of type 2 diabetes and/or SNPs: rs2234694 (SOD1), rs5746105 (SOD2), rs4880 (SOD2), rs927450 (SOD2), rs8192287 (SOD3). Genotypic variability of SNP rs3842729 (INS), previously studied in the context of insulin-dependent diabetes, is investigated in terms of selected clinical parameters associated with type 2 diabetes. This study revealed higher SOD1 concentration in diabetic men compared to women, and extremely high SOD1 concentration, higher total superoxide dismutase, and copper-zinc superoxide dismutase activity, and lower superoxide dismutase and copper-zinc superoxide dismutase activity (when adjusted for the concentration of SODs) in the diabetic group regardless of sex. Multiple logistic regression, applied to explore possible links between the studied SNPs and other factors with the odds of type 2 diabetes or obesity, revealed that the genotypic variability of rs4880 (SOD2) could affect these odds, supporting the findings of several other studies.
Collapse
Affiliation(s)
- Łukasz Lewandowski
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wrocław, Poland.
| | - Iwona Urbanowicz
- Department of Clinical Chemistry and Laboratory Hematology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wrocław, Poland
| | - Marta Kepinska
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wrocław, Poland
| | - Halina Milnerowicz
- Department of Biomedical and Environmental Analyses, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wrocław, Poland
| |
Collapse
|
28
|
Al Doghaither H, Elmorsy E, Al-Ghafari A, Ghulam J. Roles of oxidative stress, apoptosis, and inflammation in metal-induced dysfunction of beta pancreatic cells isolated from CD1 mice. Saudi J Biol Sci 2021; 28:651-663. [PMID: 33424352 PMCID: PMC7785459 DOI: 10.1016/j.sjbs.2020.10.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
The diabetogenic effects of metals including lead (Pb), mercury (Hg), cadmium (Cd), and molybdenum (Mo) have been reported with poorly identified underlying mechanisms. The current study assessed the effect of metals on the roles of oxidative stress, apoptosis, and inflammation in beta pancreatic cells isolated from CD-1 mice, via different biochemical assays. Data showed that the tested metals were cytotoxic to the isolated cells with impaired glucose stimulated insulin secretion (GSIS). This was associated with increased reactive oxygen species (ROS) production, lipid peroxidation, antioxidant enzymes activities, active proapoptotic caspase-3 (cas-3), inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) levels in the intoxicated cells. Furthermore, antioxidant-reduced glutathione (GSH-R), cas-3 inhibitor z-VAD-FMK, IL-6 inhibitor bazedoxifene (BZ), and TNF-α inhibitor etanercept (ET) were found to significantly decrease metal-induced cytotoxicity with improved GSIS in metals' intoxicated cells. In conclusion, oxidative stress, apoptosis, and inflammation can play roles in metals-induced diabetogenic effect.
Collapse
Affiliation(s)
- Huda Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Pathology Department, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Ayat Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Cancer Metabolism and Epigenetics Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Cancer and Mutagenesis Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jihan Ghulam
- General Education Department, Dar Al-Hekma University, Jeddah Saudi Arabia
| |
Collapse
|
29
|
Elmorsy E, Al-Ghafari A, Al Doghaither H, Ghulam J. Effects of environmental metals on mitochondrial bioenergetics of the CD-1 mice pancreatic beta-cells. Toxicol In Vitro 2020; 70:105015. [PMID: 33038468 DOI: 10.1016/j.tiv.2020.105015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/19/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Environmental metals are believed to have diabetogenic effects without any clear underlying mechanisms. The study investigated the effects of metals, lead (Pb), mercury (Hg), cadmium (Cd), and molybdenum (Mo), on the bioenergetics of isolated pancreatic β-cells from CD-1 mice via different functional and structural techniques. The tested metals caused significant decrease in ATP production in concentration and exposure duration-dependent pattern; Cd was the most potent cytotoxic metal. In ATP assay estimated effective concentration 50 (EC50) (25, 40, 20, and 100 μM for Pb, Hg, Cd, and Mo, respectively), the metals also significantly inhibited the glucose-stimulated insulin secretion (GSIS), mitochondrial complexes activity, mitochondrial membranes potential, and oxygen consumption rates of the treated cells with parallel increases in their lactate production and in the mitochondrial swelling and permeation of their inner mitochondrial membranes to potassium (K+) and hydrogen (H+) ions. In addition, Cd, Pb, and Hg produced significant increases in mitochondrial membrane fluidity (MMF) with significant decreases in saturated/unsaturated fatty acid ratios. In 10 μM concentration, away from Mo, the three metals showed inhibitory effects on the mitochondrial functions to variable degrees. Only Cd showed significant effect on MMF and fatty acid ratios at a concentration of 10 μM. In conclusion, the tested metals significantly affected the bioenergetics of the pancreatic β-cells with significant effect on GSIS. Cd showed the most significant functional and structural effects on their mitochondria followed by Pb, then Hg, while Mo was almost safe up to 10 μM concentration. Hence, bioenergetic mitochondrial disruption can be considered as an underlying mechanism of the diabetogenic effects of the tested metals.
Collapse
Affiliation(s)
- Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Pathology Department, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia.
| | - Ayat Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Cancer Metabolism and Epigenetics Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia; Cancer and Mutagenesis Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jihan Ghulam
- General Education Department, Dar Al-Hekma University, Jeddah, Saudi Arabia
| |
Collapse
|
30
|
Buha A, Đukić-Ćosić D, Ćurčić M, Bulat Z, Antonijević B, Moulis JM, Goumenou M, Wallace D. Emerging Links between Cadmium Exposure and Insulin Resistance: Human, Animal, and Cell Study Data. TOXICS 2020; 8:E63. [PMID: 32867022 PMCID: PMC7560347 DOI: 10.3390/toxics8030063] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Recent research has helped clarify the role of cadmium (Cd) in various pathological states. We have demonstrated Cd involvement in pancreatic cancer, as well as the bioaccumulation of Cd in the pancreas. Bioaccumulation and increased toxicity suggest that Cd may also be involved in other pancreas-mediated diseases, like diabetes. Cd falls into the category of "hyperglycemic" metals, i.e., metals that increase blood glucose levels, which could be due to increased gluconeogenesis, damage to β-cells leading to reduced insulin production, or insulin resistance at target tissue resulting in a lack of glucose uptake. This review addresses the current evidence for the role of Cd, leading to insulin resistance from human, animal, and in vitro studies. Available data have shown that Cd may affect normal insulin function through multiple pathways. There is evidence that Cd exposure results in the perturbation of the enzymes and modulatory proteins involved in insulin signal transduction at the target tissue and mutations of the insulin receptor. Cd, through well-described mechanisms of oxidative stress, inflammation, and mitochondrial damage, may also alter insulin production in β-cells. More work is necessary to elucidate the mechanisms associated with Cd-mediated insulin resistance.
Collapse
Affiliation(s)
- Aleksandra Buha
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia; (D.Đ.-Ć.); (M.Ć.); (Z.B.); (B.A.)
| | - Danijela Đukić-Ćosić
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia; (D.Đ.-Ć.); (M.Ć.); (Z.B.); (B.A.)
| | - Marijana Ćurčić
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia; (D.Đ.-Ć.); (M.Ć.); (Z.B.); (B.A.)
| | - Zorica Bulat
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia; (D.Đ.-Ć.); (M.Ć.); (Z.B.); (B.A.)
| | - Biljana Antonijević
- Department of Toxicology “Akademik Danilo Soldatović”, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia; (D.Đ.-Ć.); (M.Ć.); (Z.B.); (B.A.)
| | - Jean-Marc Moulis
- Alternative Energies and Atomic Energy Commission—Fundamental Research Division—Interdisciplinary Research Institute of Grenoble (CEA-IRIG), University of Grenoble Alpes, F-38000 Grenoble, France;
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), University of Grenoble Alpes, Inserm U1055, F-38000 Grenoble, France
| | - Marina Goumenou
- Centre of Toxicology and Forensic Sciences, Medicine School, University of Crete, 70013 Heraklion, Greece;
- General Chemical State Laboratory of Greek Republic, 71202 Heraklion, Greece
| | - David Wallace
- Department of Pharmacology & Toxicology, Oklahoma State University Center for Health Sciences, Tulsa, OK 74107, USA;
| |
Collapse
|
31
|
Theoretical Modeling of Oral Glucose Tolerance Tests Guides the Interpretation of the Impact of Perinatal Cadmium Exposure on the Offspring's Glucose Homeostasis. TOXICS 2020; 8:toxics8020030. [PMID: 32326427 PMCID: PMC7357044 DOI: 10.3390/toxics8020030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Oral glucose tolerance tests, in which the concentration of glucose is monitored in the circulation over 2 h after ingesting a bolus, probe diabetic or pre-diabetic conditions. The resulting glucose curves inform about glucose turnover, insulin production and sensitivity, and other parameters. However, extracting the relevant parameters from a single complex curve is not straightforward. We propose a simple modeling method recapitulating the most salient features of the role of insulin-secreting pancreatic β-cells and insulin sensitive tissues. This method implements four ordinary differential equations with ten parameters describing the time-dependence of glucose concentration, its removal rate, and the circulating and stored insulin concentrations. From the initial parameter set adjusted to a reference condition, fitting is done by minimizing a weighted least-square residual. In doing so, the sensitivity of β-cells to glucose was identified as the most likely impacted function at weaning for the progeny of rats that were lightly exposed to cadmium in the perigestational period. Later in life, after young rats received non-contaminated carbohydrate enriched food, differences are more subtle, but modeling agrees with long-lasting perturbation of glucose homeostasis.
Collapse
|
32
|
Li X, Huang Y, Xing Y, Hu C, Zhang W, Tang Y, Su W, Huo X, Zhou A, Xia W, Xu S, Chen D, Li Y. Association of urinary cadmium, circulating fatty acids, and risk of gestational diabetes mellitus: A nested case-control study in China. ENVIRONMENT INTERNATIONAL 2020; 137:105527. [PMID: 32007690 DOI: 10.1016/j.envint.2020.105527] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Previous studies have observed that cadmium (Cd) exposure of pregnant women was associated with increased risk of gestational diabetes mellitus (GDM). However, the potential mechanism still remains unclear. In addition, various animal studies have suggested that Cd exposure could affect fatty acids (FAs) metabolism, but data on humans are scant. OBJECTIVES We conducted a nested case-control study to investigate the associations of urinary Cd concentrations with levels of circulating FAs and risk of GDM in pregnant women, and further to examine the role of FAs in mediating the relationship between Cd exposure and risk of GDM. METHODS A total of 305 GDM cases were matched to 305 controls on pregnant women's age (±2 years) and infant's gender from a birth cohort study conducted in Wuhan, China. Urinary Cd concentrations and levels of plasma FAs between 10 and 16 gestational weeks were measured using inductively coupled plasma mass spectrometry and gas chromatography-mass spectrometry, respectively. Conditional logistic regressions models were used to estimate the associations of Cd concentrations and levels of FAs with the risk of GDM. Multiple linear regression models were applied to estimate the associations between Cd concentrations and levels of FAs. Mediation analysis was used to assess the mediating role of FAs in the association of Cd with the risk of GDM. RESULTS Urinary concentrations of Cd in cases (median: 0.69 μg/L) were significantly higher than controls (median: 0.59 μg/L, P < 0.05). Cd concentrations were positively associated with the risk of GDM (Ptrend = 0.003). Compared to the first tertile of Cd, the adjusted odds ratios (95% confidence intervals) of GDM risk were 2.08 (1.29, 3.36) for the second tertile and 2.09 (1.32, 3.33) for the third tertile. Cd concentrations were positively correlated with levels of eicosadienoic acid and arachidonic acid/eicosapentaenoic acid ratio, but negatively correlated with levels of stearic acid, eicosapentaenoic acid, total odd-chain saturated fatty acids, total n-3 polyunsaturated fatty acids (PUFAs), and n-3 PUFAs/n-6 PUFAs ratio. We did not observe evidence that the association of Cd exposure and risk of GDM was mediated through FAs. CONCLUSIONS Our findings confirmed the association of higher Cd exposure with increased risk of GDM in pregnant women, and provided forceful epidemiological evidence for the relation of Cd concentrations and levels of FAs.
Collapse
Affiliation(s)
- Xinping Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yichao Huang
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Yuling Xing
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Chen Hu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Wenxin Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yi Tang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Weijie Su
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Xia Huo
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Aifen Zhou
- Wuhan Medical & Healthcare Center for Women and Children, Wuhan, Hubei, People's Republic of China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Da Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, People's Republic of China.
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|