1
|
Hao Z, Qiu M, Liu Y, Liu Y, Chang M, Liu X, Wang Y, Sun W, Teng X, Tang Y. Co-exposure to ammonia and lipopolysaccharide-induced impaired energy metabolism via the miR-1599/HK2 axis and triggered autophagy, ER stress, and apoptosis in chicken cardiomyocytes. Poult Sci 2025; 104:104965. [PMID: 40043670 PMCID: PMC11927695 DOI: 10.1016/j.psj.2025.104965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/24/2025] Open
Abstract
Ammonia (NH3) and lipopolysaccharide (LPS), common pollutants in poultry farming environments, pose significant health risks by disrupting cellular processes. Although previous studies have demonstrated the individual effect of NH3 or LPS on human and animal health, the mechanisms underlying their combined impact on chicken heart tissue remain poorly understood. In this study, we established a chicken cardiotoxicity model to investigate the effects of NH3 and/or LPS exposure on energy metabolism, autophagy, endoplasmic reticulum (ER) stress, and apoptosis in cardiomyocytes. Our findings indicated that exposure to NH3 or/and LPS reduced ATPase activity and ATP content, led to the downregulation of HK2, PK, PDHX, and SDH, and upregulation of AMPK, resulting in impaired energy metabolism in chicken cardiomyocytes. Additionally, we found the gga-miR-1599/HK2 axis as a key regulator involved in NH3 or/and LPS-induced energy metabolism impairment. The impairment in energy metabolism activated the AMPK/mTOR pathway, which subsequently triggered autophagy, evidenced by the upregulation of Beclin, LC3-I, and LC3-II. Furthermore, decreased mTOR expression induced ER stress, as indicated by the upregulation of key markers such as ATF6, GRP78, IRE1, and PERK. ER stress, in turn, increased CHOP expression, which downregulated Bcl-2 and upregulated Bim, resulting in elevated levels of Bax, caspase-9, and caspase-3, ultimately triggering apoptosis. This study provides valuable insights into the mechanisms of NH3 and LPS co-exposure on poultry heart tissue and identifies potential molecular targets for mitigating these adverse effects.
Collapse
Affiliation(s)
- Zhiyu Hao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150000, China
| | - Minna Qiu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150000, China
| | - Yuhao Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150000, China
| | - Yuhang Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150000, China
| | - Minghang Chang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150000, China
| | - Xiumei Liu
- College of Life Sciences, Yantai University, Yantai, 264005, China
| | - Yan Wang
- School of Public Health, Beihua University, Jilin, 132013, China
| | - Wei Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150000, China.
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150000, China.
| | - You Tang
- College of Electrical and Information Engineering, JiLin Agricultural Science and Technology University, Jilin 132101, China; College of Information Technology, Jilin Agricultural University, Changchun, Jilin, 132101, China.
| |
Collapse
|
2
|
Lei C, Wang J, Zhang X, Ge X, Zhao W, Li X, Jiang W, Ma M, Wang Z, Sun S, Kong Q, Li H, Mu L, Wang J. The wnt/pyruvate kinase, muscle axis plays an essential role in the differentiation of mouse neuroblastoma cells. Neurochem Int 2024; 181:105901. [PMID: 39542042 DOI: 10.1016/j.neuint.2024.105901] [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: 07/30/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Neuronal differentiation and neurite growth are essential processes in nervous system development and are regulated by several factors. Although all-trans retinoic acid (ATRA) has been shown to mediate the differentiation of mouse neuroblastoma cells via the activation of several pathways, including Wnt/β-catenin signaling, the mechanism remains unclear. The pyruvate kinase, muscle (PKM) plays an important role in the glycolysis of neuroblastoma cells and regulates the Wnt signaling pathway in various cancer cells. In this study, we hypothesized that the Wnt/PKM axis regulates the differentiation of neuroblastoma cells (Neuro-2a and N1E-115). To test this hypothesis, we used inhibitors and activators of the Wnt/β-catenin and glycolytic pathways in ATRA-induced differentiated Neuro-2a and N1E-115 cells and established cell lines with silenced or a mutant replacement of Pkm. Western blot and qPCR showed that ATRA treatment activated the Wnt signaling pathway and inhibited PKM-mediated glycolysis. The oxygen consumption rate (indicating oxidative phosphorylation) significantly increased, whereas the extracellular acidification rate (indicating glycolysis) significantly decreased during differentiation; these effects were reversed upon PKM inhibition. The Wnt inhibitor ICG-001 and PKM activator ML-265 inhibited ATRA-induced Neuro-2a and N1E-115 differentiation, whereas RNA interference-mediated Pkm silencing promoted Neuro-2a and N1E-115 differentiation, which was reversed by PKM overexpression. Treatment with the Wnt activator kenpaullone promoted Neuro-2a and N1E-115 differentiation, which was reversed by ML-265 administration. These results indicate that Wnt/β-catenin signaling promotes Neuro-2a and N1E-115 differentiation by inhibiting PKM-mediated glycolysis during ATRA-induced differentiation. These findings may provide a new theoretical basis for the role of glycolysis in nerve differentiation.
Collapse
Affiliation(s)
- Cheng Lei
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Jiaqi Wang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xiaoyu Zhang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xuemin Ge
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Wei Zhao
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xinrong Li
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Wei Jiang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Mingyu Ma
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Zhenhai Wang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Shanshan Sun
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Qingfei Kong
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Hulun Li
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Lili Mu
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| | - Jinghua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
3
|
Yu G, Wu L, Su Q, Ji X, Zhou J, Wu S, Tang Y, Li H. Neurotoxic effects of heavy metal pollutants in the environment: Focusing on epigenetic mechanisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123563. [PMID: 38355086 DOI: 10.1016/j.envpol.2024.123563] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/04/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
The pollution of heavy metals (HMs) in the environment is a significant global environmental issue, characterized by its extensive distribution, severe contamination, and profound ecological impacts. Excessive exposure to heavy metal pollutants can damage the nervous system. However, the mechanisms underlying the neurotoxicity of most heavy metals are not completely understood. Epigenetics is defined as a heritable change in gene function that can influence gene and subsequent protein expression levels without altering the DNA sequence. Growing evidence indicates that heavy metals can induce neurotoxic effects by triggering epigenetic changes and disrupting the epigenome. Compared with genetic changes, epigenetic alterations are more easily reversible. Epigenetic reprogramming techniques, drugs, and certain nutrients targeting specific epigenetic mechanisms involved in gene expression regulation are emerging as potential preventive or therapeutic tools for diseases. Therefore, this review provides a comprehensive overview of epigenetic modifications encompassing DNA/RNA methylation, histone modifications, and non-coding RNAs in the nervous system, elucidating their association with various heavy metal exposures. These primarily include manganese (Mn), mercury (Hg), lead (Pb), cobalt (Co), cadmium (Cd), nickel (Ni), sliver (Ag), toxic metalloids arsenic (As), and etc. The potential epigenetic mechanisms in the etiology, precision prevention, and target therapy of various neurodevelopmental disorders or different neurodegenerative diseases are emphasized. In addition, the current gaps in research and future areas of study are discussed. From a perspective on epigenetics, this review offers novel insights for prevention and treatment of neurotoxicity induced by heavy metal pollutants.
Collapse
Affiliation(s)
- Guangxia Yu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Lingyan Wu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Qianqian Su
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xianqi Ji
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Jinfu Zhou
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Maternity and Child Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
| | - Siying Wu
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Ying Tang
- Fujian Center for Prevention and Control Occupational Diseases and Chemical Poisoning, Fuzhou 350125, China
| | - Huangyuan Li
- Key Lab of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
4
|
Anyachor CP, Dooka DB, Orish CN, Amadi CN, Bocca B, Ruggieri F, Senofonte M, Frazzoli C, Orisakwe OE. Mechanistic considerations and biomarkers level in nickel-induced neurodegenerative diseases: An updated systematic review. IBRO Neurosci Rep 2022; 13:136-146. [PMID: 35989698 PMCID: PMC9382260 DOI: 10.1016/j.ibneur.2022.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/30/2022] [Indexed: 10/27/2022] Open
Abstract
The environment has been implicated to be a strong determinant of brain health with higher risk of neurodegeneration. The drastic rise in the prevalence of neurodegenerative diseases (NDDs) including Alzheimer's disease (AD), Parkinson's disease (PD), autism spectrum disorder (ASD), multiple sclerosis (MS) etc., supports the idea that environmental factors may play a major role in NDDs aetiology. Nickel is one of the listed environmental metals reported to pose a serious threat to human health. This paper reported available studies on nickel level in NDDs covering both animal and human studies. Different databases were searched for articles reporting the main neurotoxicity mechanisms and the concentration of nickel in fluids and tissues of NDDs patients compared to controls. Data were extracted and synthesized by ensuring the articles were related to nickel and NDDs. Various mechanisms were reported as oxidative stress, disturbances in mitochondrial membrane potential, trace elements homeostasis destabilization, etc. Nickel was found elevated in biological fluids as blood, serum/plasma and CSF and in the brain of NDDs, as a consequence of unintentional exposure thorough nickel-contaminated air, food, water, and skin contact. In addition, after exposure to nickel, the concentration of markers of lipid peroxidation were increased, while some antioxidant defence systems decreased. Thus, the reduction in the exposure to nickel contaminant may hold a promise in reducing the incidence of NDDs.
Collapse
Affiliation(s)
- Chidinma Promise Anyachor
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Donatus Baridoo Dooka
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Chinna Nneka Orish
- Department of Anatomy, College of Health Sciences University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Cecilia Nwadiuto Amadi
- Department of Experimental Pharmacology & Toxicology, Faculty of Pharmacy, University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| | - Beatrice Bocca
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Flavia Ruggieri
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Marta Senofonte
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Frazzoli
- Department for Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, Rome Viale Regina Elena, 29900161 Roma, Italy
| | - Orish E. Orisakwe
- African Centre of Excellence for Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
- Department of Anatomy, College of Health Sciences University of Port Harcourt, PMB, 5323 Port Harcourt, Rivers State, Nigeria
| |
Collapse
|
5
|
Zhao L, Islam R, Wang Y, Zhang X, Liu LZ. Epigenetic Regulation in Chromium-, Nickel- and Cadmium-Induced Carcinogenesis. Cancers (Basel) 2022; 14:cancers14235768. [PMID: 36497250 PMCID: PMC9737485 DOI: 10.3390/cancers14235768] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Environmental and occupational exposure to heavy metals, such as hexavalent chromium, nickel, and cadmium, are major health concerns worldwide. Some heavy metals are well-documented human carcinogens. Multiple mechanisms, including DNA damage, dysregulated gene expression, and aberrant cancer-related signaling, have been shown to contribute to metal-induced carcinogenesis. However, the molecular mechanisms accounting for heavy metal-induced carcinogenesis and angiogenesis are still not fully understood. In recent years, an increasing number of studies have indicated that in addition to genotoxicity and genetic mutations, epigenetic mechanisms play critical roles in metal-induced cancers. Epigenetics refers to the reversible modification of genomes without changing DNA sequences; epigenetic modifications generally involve DNA methylation, histone modification, chromatin remodeling, and non-coding RNAs. Epigenetic regulation is essential for maintaining normal gene expression patterns; the disruption of epigenetic modifications may lead to altered cellular function and even malignant transformation. Therefore, aberrant epigenetic modifications are widely involved in metal-induced cancer formation, development, and angiogenesis. Notably, the role of epigenetic mechanisms in heavy metal-induced carcinogenesis and angiogenesis remains largely unknown, and further studies are urgently required. In this review, we highlight the current advances in understanding the roles of epigenetic mechanisms in heavy metal-induced carcinogenesis, cancer progression, and angiogenesis.
Collapse
|
6
|
Role of Oxidative Stress in the Pathogenesis of Atherothrombotic Diseases. Antioxidants (Basel) 2022; 11:antiox11071408. [PMID: 35883899 PMCID: PMC9312358 DOI: 10.3390/antiox11071408] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Oxidative stress is generated by the imbalance between reactive oxygen species (ROS) formation and antioxidant scavenger system’s activity. Increased ROS, such as superoxide anion, hydrogen peroxide, hydroxyl radical and peroxynitrite, likely contribute to the development and complications of atherosclerotic cardiovascular diseases (ASCVD). In genetically modified mouse models of atherosclerosis, the overexpression of ROS-generating enzymes and uncontrolled ROS formation appear to be associated with accelerated atherosclerosis. Conversely, the overexpression of ROS scavenger systems reduces or stabilizes atherosclerotic lesions, depending on the genetic background of the mouse model. In humans, higher levels of circulating biomarkers derived from the oxidation of lipids (8-epi-prostaglandin F2α, and malondialdehyde), as well as proteins (oxidized low-density lipoprotein, nitrotyrosine, protein carbonyls, advanced glycation end-products), are increased in conditions of high cardiovascular risk or overt ASCVD, and some oxidation biomarkers have been reported as independent predictors of ASCVD in large observational cohorts. In animal models, antioxidant supplementation with melatonin, resveratrol, Vitamin E, stevioside, acacetin and n-polyunsaturated fatty acids reduced ROS and attenuated atherosclerotic lesions. However, in humans, evidence from large, placebo-controlled, randomized trials or prospective studies failed to show any athero-protective effect of antioxidant supplementation with different compounds in different CV settings. However, the chronic consumption of diets known to be rich in antioxidant compounds (e.g., Mediterranean and high-fish diet), has shown to reduce ASCVD over decades. Future studies are needed to fill the gap between the data and targets derived from studies in animals and their pathogenetic and therapeutic significance in human ASCVD.
Collapse
|
7
|
Missawi O, Venditti M, Cappello T, Zitouni N, Marco GDE, Boughattas I, Bousserrhine N, Belbekhouche S, Minucci S, Maisano M, Banni M. Autophagic event and metabolomic disorders unveil cellular toxicity of environmental microplastics on marine polychaete Hediste diversicolor. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 302:119106. [PMID: 35248622 DOI: 10.1016/j.envpol.2022.119106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Although the hazards of microplastics (MPs) have been quite well explored, the aberrant metabolism and the involvement of the autophagy pathway as an adverse response to environmental MPs in benthic organisms are still unclear. The present work aims to assess the impact of different environmental MPs collected from the south coast of the Mediterranean Sea, composed by polyethylene (PE), polyethylene vinyl acetate (PEVA), low-density polyethylene (LDPE), high-density polyethylene (HDPE), polypropylene (PP) and polyamide (PA) on the metabolome and proteome of the marine polychaete Hediste diversicolor. As a result, all the microplastic types were detected with Raman microspectroscopy in polychaetes tissues, causing cytoskeleton damage and induced autophagy pathway manifested by immunohistochemical labeling of specific targeted proteins, through Tubulin (Tub), Microtubule-associated protein light chain 3 (LC3), and p62 (also named Sequestosome 1). Metabolomics was conducted to further investigate the metabolic alterations induced by the environmental MPs-mixture in polychaetes. A total of 28 metabolites were differentially expressed between control and MPs-treated polychaetes, which showed elevated levels of amino acids, glucose, ATP/ADP, osmolytes, glutathione, choline and phosphocholine, and reduced concentration of aspartate. These novel findings extend our understanding given the toxicity of environmental microplastics and unravel their underlying mechanisms.
Collapse
Affiliation(s)
- Omayma Missawi
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia.
| | - Massimo Venditti
- Department of Experimental Medicine, Section Human Physiology and Integrated Biological Functions "F. Bottazzi", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Tiziana Cappello
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, 98166 Messina, Italy
| | - Nesrine Zitouni
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia
| | - Giuseppe DE Marco
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, 98166 Messina, Italy
| | - Iteb Boughattas
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia; Regional Field Crops Research Center of Beja, Tunisia
| | - Noureddine Bousserrhine
- University Paris-Est Creteil, Laboratory of Water, Environment and Urban Systems, Faculty of Science and Technology, Creteil Cedex, France
| | - Sabrina Belbekhouche
- CNRS, University of Paris-Est Creteil, Institute of Chemistry and Materials Paris-Est ICMPE, UMR7182, 94320 Thiais, France
| | - Sergio Minucci
- Department of Experimental Medicine, Section Human Physiology and Integrated Biological Functions "F. Bottazzi", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Maisano
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, 98166 Messina, Italy
| | - Mohamed Banni
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia; Higher Institute of Biotechnology Monastir, University of Monastir, Monastir, Tunisia
| |
Collapse
|
8
|
Zou X, Yang H, Li Q, Li N, Hou Y, Wang X, Meng X, Yu J, Zhang Y, Tang C, Kuang T. Protective Effect of Brassica rapa Polysaccharide against Acute High-Altitude Hypoxia-Induced Brain Injury and Its Metabolomics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3063899. [PMID: 39282147 PMCID: PMC11401678 DOI: 10.1155/2022/3063899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/24/2022] [Accepted: 03/15/2022] [Indexed: 09/18/2024]
Abstract
Brassica rapa L., a traditional Tibetan medicine, has been wildly used for treating plateau disease. Polysaccharide is an important chemical component in B. rapa. The present study aimed to evaluate the effect of B. rapa polysaccharide (BRP) against acute high-altitude hypoxia (AHH) induced brain injury and its metabolic mechanism. The rats were randomly divided into six groups: control group, AHH group, Hongjingtian oral liquid group, and three BRP groups (38, 75, and 150 mg/kg/d). Serum levels of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), oxidized glutathione (GSSG), and lactate dehydrogenase (LDH) were detected by commercial biochemical kits. Hippocampus and cortex histopathological changes were observed by H&E staining and Nissl staining. Neuronal apoptosis was observed by TUNEL staining. The protein and gene expression of Caspase-3, Bax, Bcl-2, p-PI3K, PI3K, p-Akt, Akt, HIF-1α, microRNA 210, ISCU1/2, and COX10 were detected by western blotting and qRT-PCR. Then, a brain metabolomics method based on UPLC-Q-Exactive-MS was performed to discover potential biomarkers and analyze metabolic pathways. It was found that BRP decreased levels of MDA, LDH, and GSSG, increased GSH and SOD, reduced the pathological changes, inhibited apoptosis, and activated the PI3K/Akt/HIF-1α signaling pathway as evidenced by increased phosphorylation of PI3K and Akt, enhanced protein expression of HIF-1α and gene levels of microRNA210, ISCU1/2, and COX10. Furthermore, 15 endogenous potential biomarkers were identified in the brain through metabolomics analysis. BRP can regulate 7 potential biomarkers and the corresponding metabolic pathways were mainly associated with pyruvate metabolism and glycolysis/gluconeogenesis. Collectively, BRP has a clear protective effect on AHH-induced brain injury and its mechanisms may be related to ameliorate oxidative stress injury, inhibit apoptosis by activating PI3K/Akt/HIF-1α signaling pathway, and reverse metabolic pathway disturbances.
Collapse
Affiliation(s)
- Xuemei Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hailing Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qiuyue Li
- Pharmacy Intravenous Admixture Services, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646600, China
| | - Ning Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ya Hou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jia Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ce Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingting Kuang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
9
|
Nickel's Role in Pancreatic Ductal Adenocarcinoma: Potential Involvement of microRNAs. TOXICS 2022; 10:toxics10030148. [PMID: 35324773 PMCID: PMC8952337 DOI: 10.3390/toxics10030148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 02/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancer types with a limited overall survival rate due to the asymptomatic progression of symptoms in metastatic stages of the malignancy and the lack of an early reliable diagnostic biomarker. MicroRNAs (miRs/miRNAs) are small (~18–24 nucleotides), endogenous, non-coding RNAs, which are closely linked to the development of numerous malignancies comprising PDAC. Recent studies have described the role of environmental pollutants such as nickel (Ni) in PDAC, but the mechanisms of Ni-mediated toxicity in cancer are still not completely understood. Specifically, Ni has been found to alter the expression and function of miRs in several malignancies, leading to changes in target gene expression. In this study, we found that levels of Ni were significantly higher in cancerous tissue, thus implicating Ni in pancreatic carcinogenesis. Hence, in vitro studies followed by using both normal and pancreatic tumor cell lines and increasing Ni concentration increased lethality. Comparing LC50 values, Ni-acetate groups demonstrated lower values needed than in NiCl2 groups, suggesting greater Ni-acetate. Panc-10.05 cell line appeared the most sensitive to Ni compounds. Exposure to Ni-acetate resulted in an increased phospho-AKT, and decreased FOXO1 expression in Panc-10.05 cells, while NiCl2 also increased PTEN expression in Panc-10.05 cells. Specifically, following NiCl2 exposure to PDAC cells, the expression levels of miR-221 and miR-155 were significantly upregulated, while the expression levels of miR-126 were significantly decreased. Hence, our study has suggested pilot insights to indicate that the environmental pollutant Ni plays an important role in the progression of PDAC by promoting an association between miRs and Ni exposure during PDAC pathogenesis.
Collapse
|
10
|
Chai T, Qiu C, Xian Z, Lu Y, Zeng Y, Li J. A narrative review of research advances in hypoxic pulmonary hypertension. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:230. [PMID: 35280399 PMCID: PMC8908157 DOI: 10.21037/atm-22-259] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/18/2022] [Indexed: 11/23/2022]
Abstract
Background and Objective Hypoxic pulmonary hypertension (HPH) is a pathological syndrome characterized by pulmonary vasoconstriction and pulmonary vascular remodeling caused by hypoxia, which eventually leads to right heart failure or death. There are 2 stages of onset of HPH: hypoxic pulmonary vasoconstriction (HPV) and hypoxic pulmonary vascular remodeling (HPVR). It is an important pathophysiological link in the pathogenesis of chronic obstructive pulmonary disease (COPD) and chronic mountain sickness (CMS), and its severity is closely related to the course and prognosis of COPD and CMS. However, there is a lack of systematic review on the diagnosis, pathogenesis and treatment of HPH. The objective of this paper is to review the diagnosis, pathogenesis, treatment of HPH. Methods In this paper, the method of literature review is adopted to obtain the information about HPH. Based on the literature, comprehensive and systematic review is made. The diagnosis, pathogenesis, treatment of HPH are summarized. Key Content and Findings Right heart catheterization is the gold standard for diagnosing HPH. Hypoxia-inducible factor, oxidative stress, metal metabolism, ion channel, inflammatory cytokines, cell apoptosis and vascular factors are the main pathogenesis of HPH. The treatment of HPH includes long-term oxygen therapy, statins, prostaglandins, phosphodiesterase inhibitor and ET receptor antagonists. Conclusions Although great progress has been made in the pathophysiology and molecular biology of HPH, it is still unclear which factors play a leading role in the pathogenesis of HPH, and no breakthrough has been made in the treatment of HPH. It is believed that the specific mechanism will be revealed as the research continues, and earlier diagnosis and the development of more effective targeted drugs will be the focus of future research.
Collapse
Affiliation(s)
- Tianci Chai
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Chen Qiu
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Zhihong Xian
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Yongzhen Lu
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Yuwei Zeng
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Jie Li
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| |
Collapse
|
11
|
Wang L, Liu LZ, Jiang BH. Dysregulation of microRNAs in metal-induced angiogenesis and carcinogenesis. Semin Cancer Biol 2021; 76:279-286. [PMID: 34428550 DOI: 10.1016/j.semcancer.2021.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small endogenous non-coding RNAs that regulate cancer initiation, development, angiogenesis, and therapeutic resistance. Metal exposure widely occurs through air, water, soil, food, and industrial contaminants. Hundreds of millions of people may have metal exposure associated with toxicity, serious health problems, and cancer occurrence. Metal exposure is found to induce oxidative stress, DNA damage and repair, and activation of multiple signaling pathways. However, molecular mechanisms of metal-induced carcinogenesis remain to be elucidated. Recent studies demonstrated that the exposure of metals such as arsenic, hexavalent chromium, cadmium, and nickel caused dysregulation of microRNAs that are implicated to play an important role in cell transformation, tumor growth and angiogenesis. This review focuses on the recent studies that show metal-induced miRNA dysregulation and underlined mechanisms in cell malignant transformation, angiogenesis and tumor growth.
Collapse
Affiliation(s)
- Lin Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China; Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| | - Ling-Zhi Liu
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, United States.
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States.
| |
Collapse
|
12
|
Du H, Zhao Y, Li H, Wang DW, Chen C. Roles of MicroRNAs in Glucose and Lipid Metabolism in the Heart. Front Cardiovasc Med 2021; 8:716213. [PMID: 34368265 PMCID: PMC8339264 DOI: 10.3389/fcvm.2021.716213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that participate in heart development and pathological processes mainly by silencing gene expression. Overwhelming evidence has suggested that miRNAs were involved in various cardiovascular pathological processes, including arrhythmias, ischemia-reperfusion injuries, dysregulation of angiogenesis, mitochondrial abnormalities, fibrosis, and maladaptive remodeling. Various miRNAs could regulate myocardial contractility, vascular proliferation, and mitochondrial function. Meanwhile, it was reported that miRNAs could manipulate nutrition metabolism, especially glucose and lipid metabolism, by regulating insulin signaling pathways, energy substrate transport/metabolism. Recently, increasing studies suggested that the abnormal glucose and lipid metabolism were closely associated with a broad spectrum of cardiovascular diseases (CVDs). Therefore, maintaining glucose and lipid metabolism homeostasis in the heart might be beneficial to CVD patients. In this review, we summarized the present knowledge of the functions of miRNAs in regulating cardiac glucose and lipid metabolism, as well as highlighted the miRNA-based therapies targeting cardiac glucose and lipid metabolism.
Collapse
Affiliation(s)
- Hengzhi Du
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yanru Zhao
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Huaping Li
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Wosnick N, Niella Y, Hammerschlag N, Chaves AP, Hauser-Davis RA, da Rocha RCC, Jorge MB, de Oliveira RWS, Nunes JLS. Negative metal bioaccumulation impacts on systemic shark health and homeostatic balance. MARINE POLLUTION BULLETIN 2021; 168:112398. [PMID: 33906010 DOI: 10.1016/j.marpolbul.2021.112398] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 02/28/2021] [Accepted: 04/01/2021] [Indexed: 06/12/2023]
Abstract
Contamination by metals is among the most pervasive anthropogenic threats to the environment. Despite the ecological importance of marine apex predators, the potential negative impacts of metal bioaccumulation and biomagnification on the health of higher trophic level species remains unclear. To date, most toxicology studies in sharks have focused on measuring metal concentrations in muscle tissues associating human consumption and food safety, without further investigating potential impacts on shark health. To help address this knowledge gap, the present study evaluated metal concentrations in the gills, muscle, liver and rectal gland of coastal sharks opportunistically sampled from Brazilian waters and tested for potential relationships between metal bioaccumulation and general shark health and homeostatic balance metrics. Results revealed high metal concentrations in all four tissue types, with levels varying in relation to size, sex, and life-stage. Metal concentrations were also associated with serum biomarkers (urea, lactate, ALT, triglycerides, alkaline phosphatase, and phosphorus) and body condition, suggesting potential negative impacts on organismal health.
Collapse
Affiliation(s)
- Natascha Wosnick
- Programa de Pós-Graduação em Zoologia, Universidade Federal do Paraná, Brazil.
| | - Yuri Niella
- Department of Biological Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, Australia
| | - Neil Hammerschlag
- Rosenstiel School of Marine and Atmospheric Science, University of Miami, Florida, United States
| | - Ana Paula Chaves
- Analytical and System Toxicology Laboratory, Faculdade de Ciências Farmacêuticas de Ribeirão Preto (USP), SP, Brazil
| | - Rachel Ann Hauser-Davis
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
14
|
Das S, Reddy RC, Chadchan KS, Patil AJ, Biradar MS, Das KK. Nickel and Oxidative Stress: Cell Signaling Mechanisms and Protective Role of Vitamin C. Endocr Metab Immune Disord Drug Targets 2021; 20:1024-1031. [PMID: 31804169 DOI: 10.2174/1871530319666191205122249] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Nickel activates the signaling pathways through the oxygen sensing mechanism and the signaling cascades that control hypoxia-inducible transcriptional gene expressions through oxidative stress. This review emphasizes on the recent updates of nickel toxicities on oxidant and antioxidant balance, molecular interaction of nickel and its signal transduction through low oxygen microenvironment in the in-vivo physiological system. DISCUSSION Nickel alters intracellular chemical microenvironment by increasing ionized calcium concentration, lipid peroxidation, cyclooxygenase, constitutive nitric oxide synthase, leukotriene B4, prostaglandin E2, interleukins, tumor necrosis factor-α, caspases, complement activation, heat shock protein 70 kDa and hypoxia-inducible factor-1α. The oxidative stress induced by nickel is responsible for the progression of metastasis. It has been observed that nickel exposure induces the generation of reactive oxygen species which leads to the increased expression of p53, NF-kβ, AP-1, and MAPK. Ascorbic acid (vitamin C) prevents lipid peroxidation, oxidation of low-density lipoproteins and advanced oxidation protein products. The mechanism involves that vitamin C is capable of reducing ferric iron to ferrous iron in the duodenum, thus the availability of divalent ferrous ion increases which competes with nickel (a divalent cation itself) and reduces its intestinal absorption and nickel toxicities. CONCLUSION Reports suggested the capability of ascorbic acid as a regulatory factor to influence gene expression, apoptosis and other cellular functions of the living system exposed to heavy metals, including nickel.
Collapse
Affiliation(s)
- Swastika Das
- Department of Chemistry, BLDEA's V. P. Dr. P. G. Halakatti College of Engineering and Technology, Vijayapur- 586103, Karnataka, India
| | - Rachamalla C Reddy
- Department of Physiology, Laboratory of Vascular Physiology and Medicine, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Vijayapur-586103, Karnataka, India
| | - Kailash S Chadchan
- Department of Chemistry, BLDEA's V. P. Dr. P. G. Halakatti College of Engineering and Technology, Vijayapur- 586103, Karnataka, India
| | - Arun J Patil
- Department of Biochemistry, Krishna Institute of Medical Sciences "Deemed to be University", Karad- 415539, Maharashtra, India
| | - Mallanagouda S Biradar
- Department of Physiology, Laboratory of Vascular Physiology and Medicine, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Vijayapur-586103, Karnataka, India
| | - Kusal K Das
- Department of Physiology, Laboratory of Vascular Physiology and Medicine, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Vijayapur-586103, Karnataka, India
| |
Collapse
|
15
|
Study on the Relationship between the miRNA-centered ceRNA Regulatory Network and Fatigue. J Mol Neurosci 2021; 71:1967-1974. [PMID: 33993410 PMCID: PMC8500871 DOI: 10.1007/s12031-021-01845-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/13/2021] [Indexed: 10/25/2022]
Abstract
In recent years, the incidence of fatigue has been increasing, and the effective prevention and treatment of fatigue has become an urgent problem. As a result, the genetic research of fatigue has become a hot spot. Transcriptome-level regulation is the key link in the gene regulatory network. The transcriptome includes messenger RNAs (mRNAs) and noncoding RNAs (ncRNAs). MRNAs are common research targets in gene expression profiling. Noncoding RNAs, including miRNAs, lncRNAs, circRNAs and so on, have been developed rapidly. Studies have shown that miRNAs are closely related to the occurrence and development of fatigue. MiRNAs can regulate the immune inflammatory reaction in the central nervous system (CNS), regulate the transmission of nerve impulses and gene expression, regulate brain development and brain function, and participate in the occurrence and development of fatigue by regulating mitochondrial function and energy metabolism. LncRNAs can regulate dopaminergic neurons to participate in the occurrence and development of fatigue. This has certain value in the diagnosis of chronic fatigue syndrome (CFS). CircRNAs can participate in the occurrence and development of fatigue by regulating the NF-κB pathway, TNF-α and IL-1β. The ceRNA hypothesis posits that in addition to the function of miRNAs in unidirectional regulation, mRNAs, lncRNAs and circRNAs can regulate gene expression by competitive binding with miRNAs, forming a ceRNA regulatory network with miRNAs. Therefore, we suggest that the miRNA-centered ceRNA regulatory network is closely related to fatigue. At present, there are few studies on fatigue-related ncRNA genes, and most of these limited studies are on miRNAs in ncRNAs. However, there are a few studies on the relationship between lncRNAs, cirRNAs and fatigue. Less research is available on the pathogenesis of fatigue based on the ceRNA regulatory network. Therefore, exploring the complex mechanism of fatigue based on the ceRNA regulatory network is of great significance. In this review, we summarize the relationship between miRNAs, lncRNAs and circRNAs in ncRNAs and fatigue, and focus on exploring the regulatory role of the miRNA-centered ceRNA regulatory network in the occurrence and development of fatigue, in order to gain a comprehensive, in-depth and new understanding of the essence of the fatigue gene regulatory network.
Collapse
|
16
|
EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Ntzani E, Petersen A, Sand S, Schwerdtle T, Vleminckx C, Wallace H, Guérin T, Massanyi P, Van Loveren H, Baert K, Gergelova P, Nielsen E. Update of the risk assessment of nickel in food and drinking water. EFSA J 2020; 18:e06268. [PMID: 33193868 PMCID: PMC7643711 DOI: 10.2903/j.efsa.2020.6268] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The European Commission asked EFSA to update its previous Opinion on nickel in food and drinking water, taking into account new occurrence data, the updated benchmark dose (BMD) Guidance and newly available scientific information. More than 47,000 analytical results on the occurrence of nickel were used for calculating chronic and acute dietary exposure. An increased incidence of post-implantation loss in rats was identified as the critical effect for the risk characterisation of chronic oral exposure and a BMDL 10 of 1.3 mg Ni/kg body weight (bw) per day was selected as the reference point for the establishment of a tolerable daily intake (TDI) of 13 μg/kg bw. Eczematous flare-up reactions in the skin elicited in nickel-sensitised humans, a condition known as systemic contact dermatitis, was identified as the critical effect for the risk characterisation of acute oral exposure. A BMDL could not be derived, and therefore, the lowest-observed-adverse-effect-level of 4.3 μg Ni/kg bw was selected as the reference point. The margin of exposure (MOE) approach was applied and an MOE of 30 or higher was considered as being indicative of a low health concern. The mean lower bound (LB)/upper bound (UB) chronic dietary exposure was below or at the level of the TDI. The 95th percentile LB/UB chronic dietary exposure was below the TDI in adolescents and in all adult age groups, but generally exceeded the TDI in toddlers and in other children, as well as in infants in some surveys. This may raise a health concern in these young age groups. The MOE values for the mean UB acute dietary exposure and for the 95th percentile UB raises a health concern for nickel-sensitised individuals. The MOE values for an acute scenario regarding consumption of a glass of water on an empty stomach do not raise a health concern.
Collapse
|
17
|
Akinwumi KA, Jubril AJ, Olaniyan OO, Umar YY. Ethanol extract of Nigella sativa has antioxidant and ameliorative effect against nickel chloride-induced hepato-renal injury in rats. CLINICAL PHYTOSCIENCE 2020. [DOI: 10.1186/s40816-020-00205-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Nickel exposure causes hepato-renal toxicity via oxidative stress. Medicinal plants with antioxidants properties are being explored as treatment options. In this study, the effect of ethanol extract of Nigella sativa (ENS) on nickel chloride (NiCl2)-induced hepato-renal damage was evaluated by monitoring biochemical and oxidative stress markers. Additionally, the antioxidant capacity and phytochemical constituents of ENS were quantified using HPLC and GC-MS.
Result
NiCl2 significantly increased (p < 0.05) aspartate aminotransferase, creatinine, sodium ion, chloride ion and malondialdehyde levels, while antioxidant enzymes were decreased in the organs except for kidney glutathione-S-transferase when compared to the control. However, ENS exerted inhibitory effect against NiCl2 toxicity in both organs by reversing the biomarkers towards control levels. ENS has a high antioxidant capacity and is rich in antioxidants including gallic acid, quercetin, eucalyptol and levomenthol that may have accounted for the improvement of hepato-renal health in co-exposed rats.
Conclusion
Our result suggests that amelioration of nickel chloride-induced hepato-renal pathology by ethanol extract of Nigella sativa was related to its antioxidant properties. Therefore, Nigella sativa could be valuable in the management of nickel-induced toxicity.
Collapse
|
18
|
Melatonin Antagonizes Nickel-Induced Aerobic Glycolysis by Blocking ROS-Mediated HIF-1 α/miR210/ISCU Axis Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5406284. [PMID: 32566089 PMCID: PMC7275958 DOI: 10.1155/2020/5406284] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/10/2020] [Accepted: 04/29/2020] [Indexed: 11/23/2022]
Abstract
Nickel and its compounds, which are well-documented carcinogens, induce the Warburg effect in normal cells by stabilizing hypoxia-inducible factor 1α (HIF-1α). Melatonin has shown diverse anticancer properties for its reactive oxygen species- (ROS-) scavenging ability. Our aim was to explore how melatonin antagonized a nickel-induced increment in aerobic glycolysis. In the current work, a normal human bronchial epithelium cell line (BEAS-2B) was exposed to a series of nonlethal doses of NiCl2, with or without 1 mM melatonin. Melatonin attenuated nickel-enhanced aerobic glycolysis. The inhibition effects on aerobic glycolysis were attributed to the capability of melatonin to suppress the regulatory axis comprising HIF-1α, microRNA210 (miR210), and iron-sulfur cluster assembly scaffold protein (ISCU1/2). N-Acetylcysteine (NAC) manifested similar effects as melatonin in scavenging ROS, maintaining prolyl-hydroxylase activity, and mitigating HIF-1α transcriptional activity in nickel-exposed cells. Our results indicated that ROS generation contributed to nickel-caused HIF-1α stabilization and downstream signal activation. Melatonin could antagonize HIF-1α-controlled aerobic glycolysis through ROS scavenging.
Collapse
|
19
|
Chi Q, Luan Y, Zhang Y, Hu X, Li S. The regulatory effects of miR-138-5p on selenium deficiency-induced chondrocyte apoptosis are mediated by targeting SelM. Metallomics 2020; 11:845-857. [PMID: 30869711 DOI: 10.1039/c9mt00006b] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Apoptosis is a common paradigm of cell death and plays a key role in cartilage damage and selenium (Se) deficiency. Selenoproteins play major roles in determining the biological effects of Se, and are potentially involved in the pathophysiological processes in bone tissue. MicroRNAs (miRNAs) play important roles in cell proliferation, differentiation, apoptosis and tumorigenesis. Based on the preliminary results, the expression of selenoprotein M (SelM) was significantly decreased (69%) in chicken cartilage tissues with Se deficiency, and we subsequently screened and verified that SelM is one of the target genes of miR-138-5p in chicken cartilage using a dual luciferase reporter assay and real-time quantitative PCR (qRT-PCR). The expression of miR-138-5p was increased in response to Se deficiency, and the overexpression of miR-138-5p increased caspase-3, caspase-9, BAX and BAK levels, while the BCL-2 level was decreased, suggesting that miR-138-5p induced apoptosis via the mitochondrial pathway in vivo and in vitro. We explored whether oxidative stress, mitochondrial fission and fusion, and energy metabolism might trigger apoptosis to obtain an understanding of the mechanisms underlying the effects of miR-138-5p on Se deficiency-induced apoptosis in cartilage. The levels of indicators of oxidative stress, mitochondrial dynamics and energy metabolism were changed as well. This study confirmed that SelM is one of the target genes of miR-138-5p, and the overexpression of miR-138-5p induced by Se deficiency triggered oxidative stress, an imbalance in mitochondrial fission and fusion, and energy metabolism dysfunction. Therefore, miR-138-5p is involved in the mitochondrial apoptosis pathway via targeting SelM in chicken chondrocytes.
Collapse
Affiliation(s)
- Qianru Chi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| | | | | | | | | |
Collapse
|
20
|
Wang X, Hou Y, Li Q, Li X, Wang W, Ai X, Kuang T, Chen X, Zhang Y, Zhang J, Hu Y, Meng X. Rhodiola crenulata attenuates apoptosis and mitochondrial energy metabolism disorder in rats with hypobaric hypoxia-induced brain injury by regulating the HIF-1α/microRNA 210/ISCU1/2(COX10) signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:111801. [PMID: 30878546 DOI: 10.1016/j.jep.2019.03.028] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/06/2019] [Accepted: 03/10/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhodiola crenulata, a traditional Tibetan medicine, has shown promise in the treatment of hypobaric hypoxia (HH)-induced brain injury. However, the underlying mechanisms remain unclear. This study investigated the protective effects of R. crenulata aqueous extract (RCAE) on HH-induced brain injury in rats. MATERIALS AND METHODS An animal model of high-altitude hypoxic brain injury was established in SD rats using an animal decompression chamber for 24 h. Serum and hippocampus levels of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), oxidized glutathione (GSSG), and lactate dehydrogenase (LDH) were then determined using commercial biochemical kits. Neuron morphology and vitality were also evaluated using H&E and Nissl staining, and TUNEL staining was used to examine apoptosis. Gene and protein expression of HIF-1α, microRNA 210, ISCU1/2, COX10, Apaf-1, cleaved Caspase-3, Caspase-3, Bax, Bcl-2, and Cyto-c were determined by western blot, immunohistochemical and qRT-PCR analysis. RESULTS RCAE administration attenuated HH-induced brain injury as evidenced by decreased levels of MDA, LDH, and GSSG, increased GSH and SOD, improvements in hippocampus histopathological changes, increased cell vitality and ATP level, and reduced apoptotic cell numbers. RCAE treatment also enhanced HIF-1α, ISCU1/2, COX10, and Bcl-2 protein expression, while dramatically inhibiting expression of Apaf-1, Bax, Cyto-c, and cleaved Caspase-3. Treatment also increased gene levels of HIF-1α, microRNA 210, ISCU1/2, and COX10, and decreased Caspase-3 gene production. CONCLUSIONS RCAE attenuated HH-induced brain injury by regulating apoptosis and mitochondrial energy metabolism via the HIF-1α/microRNA 210/ISCU1/2 (COX10) signaling pathway.
Collapse
Affiliation(s)
- Xiaobo Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China; Interdisciplinary Laboratory of Exercise and Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ya Hou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China; Interdisciplinary Laboratory of Exercise and Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Qiuyue Li
- Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Xuanhao Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China; Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Wenxiang Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Xiaopeng Ai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Tingting Kuang
- Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Xiaorui Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Yi Zhang
- Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Jing Zhang
- Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| | - Yao Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China; Interdisciplinary Laboratory of Exercise and Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| | - Xianli Meng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| |
Collapse
|
21
|
Abstract
Nickel is a naturally occurring element found in the Earth’s crust and an International Agency for Research on Cancer (IARC)-classified human carcinogen. While low levels found in the natural environment pose a minor concern, the extensive use of nickel in industrial settings such as in the production of stainless steel and various alloys complicate human exposure and health effects. Notably, interactions with nickel macromolecules, primarily through inhalation, have been demonstrated to promote lung cancer. Mechanisms of nickel-carcinogenesis range from oxidative stress, DNA damage, and hypoxia-inducible pathways to epigenetic mechanisms. Recently, non-coding RNAs have drawn increased attention in cancer mechanistic studies. Specifically, nickel has been found to disrupt expression and functions of micro-RNAs and long-non-coding RNAs, resulting in subsequent changes in target gene expression levels, some of which include key cancer genes such as p53, MDM2, c-myc, and AP-1. Non-coding RNAs are also involved in well-studied mechanisms of nickel-induced lung carcinogenesis, such as the hypoxia-inducible factor (HIF) pathway, oxidative stress, DNA damage and repair, DNA hypermethylation, and alterations in tumor suppressors and oncogenes. This review provides a summary of the currently known epigenetic mechanisms involved in nickel-induced lung carcinogenesis, with a particular focus on non-coding RNAs.
Collapse
|
22
|
Ullmann P, Nurmik M, Begaj R, Haan S, Letellier E. Hypoxia- and MicroRNA-Induced Metabolic Reprogramming of Tumor-Initiating Cells. Cells 2019; 8:E528. [PMID: 31159361 PMCID: PMC6627778 DOI: 10.3390/cells8060528] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC), the second most common cause of cancer mortality in the Western world, is a highly heterogeneous disease that is driven by a rare subpopulation of tumorigenic cells, known as cancer stem cells (CSCs) or tumor-initiating cells (TICs). Over the past few years, a plethora of different approaches, aimed at identifying and eradicating these self-renewing TICs, have been described. A focus on the metabolic and bioenergetic differences between TICs and less aggressive differentiated cancer cells has thereby emerged as a promising strategy to specifically target the tumorigenic cell compartment. Extrinsic factors, such as nutrient availability or tumor hypoxia, are known to influence the metabolic state of TICs. In this review, we aim to summarize the current knowledge on environmental stress factors and how they affect the metabolism of TICs, with a special focus on microRNA (miRNA)- and hypoxia-induced effects on colon TICs.
Collapse
Affiliation(s)
- Pit Ullmann
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg.
| | - Martin Nurmik
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg.
| | - Rubens Begaj
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg.
| | - Serge Haan
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg.
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg.
| |
Collapse
|
23
|
Fuschi P, Maimone B, Gaetano C, Martelli F. Noncoding RNAs in the Vascular System Response to Oxidative Stress. Antioxid Redox Signal 2019; 30:992-1010. [PMID: 28683564 DOI: 10.1089/ars.2017.7229] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Redox homeostasis plays a pivotal role in vascular cell function and its imbalance has a causal role in a variety of vascular diseases. Accordingly, the response of mammalian cells to redox cues requires precise transcriptional and post-transcriptional modulation of gene expression patterns. Recent Advances: Mounting evidence shows that nonprotein-coding RNAs (ncRNAs) are important for the functional regulation of most, if not all, cellular processes and tissues. Not surprisingly, a prominent role of ncRNAs has been identified also in the vascular system response to oxidative stress. CRITICAL ISSUES The highly heterogeneous family of ncRNAs has been divided into several groups. In this article we focus on two classes of regulatory ncRNAs: microRNAs and long ncRNAs (lncRNAs). Although knowledge in many circumstances, and especially for lncRNAs, is still fragmentary, ncRNAs are clinically interesting because of their diagnostic and therapeutic potential. We outline ncRNAs that are regulated by oxidative stress as well as ncRNAs that modulate reactive oxygen species production and scavenging. More importantly, we describe the role of these ncRNAs in vascular physiopathology and specifically in disease conditions wherein oxidative stress plays a crucial role, such as hypoxia and ischemia, ischemia reperfusion, inflammation, diabetes mellitus, and atherosclerosis. FUTURE DIRECTIONS The therapeutic potential of ncRNAs in vascular diseases and in redox homeostasis is discussed.
Collapse
Affiliation(s)
- Paola Fuschi
- 1 Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan, Italy
| | - Biagina Maimone
- 1 Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan, Italy
| | - Carlo Gaetano
- 2 Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main, Germany
| | - Fabio Martelli
- 1 Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan, Italy
| |
Collapse
|
24
|
Hou Y, Wang X, Chen X, Zhang J, Ai X, Liang Y, Yu Y, Zhang Y, Meng X, Kuang T, Hu Y. Establishment and evaluation of a simulated high‑altitude hypoxic brain injury model in SD rats. Mol Med Rep 2019; 19:2758-2766. [PMID: 30720143 PMCID: PMC6423628 DOI: 10.3892/mmr.2019.9939] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/25/2019] [Indexed: 12/19/2022] Open
Abstract
This study was conducted to establish a stable hypobaric hypoxia brain injury model. SD rats were randomly separated into control and model groups, and placed outside or inside of a hypobaric chamber, respectively. Subsequent to 24 h anoxic exposure, plasma superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), oxidized glutathione (GSSG) and lactate dehydrogenase (LDH) were measured using commercial biochemical kits. Hematoxylin-eosin (H&E), Nissl's and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining were used to observe the morphology of neurons in the hippocampus. The protein expression levels of apoptotic protease activating factor-1 (Apaf-1), hypoxia inducible factor-1α (HIF-1α), caspase-3, cleaved caspase-3, Bcl-2-associated X protein (Bax) and cytochrome c (cyto-c) were detected using western blot and immunohistochemistry analyses. Hypoxic substantially induced morphological lesions in the hippocampus concomitant with the physical behavioral performance deficit. Furthermore, hypoxia markedly exacerbated the levels of MDA, LDH and GSSG, and restrained GSH (P<0.01) and SOD (P<0.05) levels compared with the control group. In addition, hypoxia significantly induced the protein expression of Apaf-1, HIF-1α, caspase-3, cleaved caspase-3, Bax and Cyto-c (P<0.01) compared with the control group. Finally, a lower number and volume of Nissl bodies were verified in the hypoxic group. TUNEL results demonstrated a greater number of apoptotic cells in the hypoxic group. The present study demonstrates a model of rat hypoxic brain injuries induced by a hypobaric chamber at 9,000 m for 24 h. Furthermore, the redox enzyme, HIF-1α and mitochondrial apoptosis-associated protein, along with H&E and Nissl's staining, may be applied to evaluate the degree of injury.
Collapse
Affiliation(s)
- Ya Hou
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xiaobo Wang
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xiaorui Chen
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Jing Zhang
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xiaopeng Ai
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yusheng Liang
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yangyang Yu
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yi Zhang
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Xianli Meng
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Tingting Kuang
- Department of Tibetan Medicine, Ethnic Medicine College, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| | - Yao Hu
- Department of Pharmacology of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Wenjiang, Chengdu, Sichuan 611137, P.R. China
| |
Collapse
|
25
|
Abstract
A hypoxic environment can be defined as a region of the body or the whole body that is deprived of oxygen. Hypoxia is a feature of many diseases, such as cardiovascular disease, tissue trauma, stroke, and solid cancers. A loss of oxygen supply usually results in cell death; however, when cells gradually become hypoxic, they may survive and continue to thrive as described for conditions that promote metastatic growth. The role of hypoxia in these pathogenic pathways is therefore of great interest, and understanding the effect of hypoxia in regulating these mechanisms is fundamentally important. This chapter gives an extensive overview of these mechanisms. Moreover, given the challenges posed by tumor hypoxia we describe the current methods to simulate and detect hypoxic conditions followed by a discussion on current and experimental therapies that target hypoxic cells.
Collapse
Affiliation(s)
- Elizabeth Bowler
- College of Medicine and Health, University of Exeter Medical School, Exeter, UK.
| | - Michael R Ladomery
- Faculty Health and Applied Sciences, University of the West of England, Bristol, UK
| |
Collapse
|
26
|
Zhang W, Zhang S, Li B, Sun M, Zhang J. Paravertebral dexmedetomidine as an adjuvant to ropivacaine protects against independent lung injury during one-lung ventilation: a preliminary randomized clinical trial. BMC Anesthesiol 2018; 18:67. [PMID: 29907082 PMCID: PMC6003137 DOI: 10.1186/s12871-018-0532-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/28/2018] [Indexed: 12/27/2022] Open
Abstract
Background To investigate the effect of paravertebral dexmedetomidine as an adjuvant to ropivacaine on independent lung injury during one-lung ventilation. Methods In total, 120 patients who underwent elective radical resection of pulmonary carcinoma were randomly assigned to one of six groups (n = 20): normal saline (C group), ropivacaine (R group), intravenous dexmedetomidine (Div group), 0.5 μg/kg paravertebral dexmedetomidine as an adjuvant to ropivacaine (RD0.5 group), 1.0 μg/kg paravertebral dexmedetomidine as an adjuvant to ropivacaine (RD1.0 group), or 2.0 μg/kg paravertebral dexmedetomidine as an adjuvant to ropivacaine (RD2.0 group). Patients in the R, Div, RD0.5, RD1.0 and RD2.0 groups underwent a thoracic paravertebral block, and normal saline was administered as a control to C group. Small marginal lung samples next to the tumor were harvested immediately after the tumor tissues were excised. Lung injury was evaluated as follows: an injury score was determined via light microscopy, and cell apoptosis was determined via a TUNEL assay. TNF-α, IL-6, miRNA-210, HIF-1α, Tom20 and ISCU2 were also detected. Results Both intravenous and paravertebral dexmedetomidine attenuated independent lung injury. Downregulation of HIF-1α and miRNA-210 and upregulation of Tom20 and ISCU2 may be the underlying mechanism. No difference was observed between the Div and RD0.5 groups, and no further improvement of lung injury was found in the RD1.0 and RD2.0 groups with increased paravertebral dexmedetomidine doses. Conclusions Paravertebral dexmedetomidine as an adjuvant to ropivacaine, which is comparable to intravenous dexmedetomidine, could protect against independent lung injury during one-lung ventilation. Trial registration ISRCTN, 13000406; retrospectively registered on 22.05.2018.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Anesthesiology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou City, Henan Province, China
| | - Shanfeng Zhang
- Department of Biochemistry and Molecular Biology, The Academy of Medical Science, Zhengzhou University, No. 100, Science Avenue, Zhengzhou City, Henan Province, China
| | - Bing Li
- Department of Anesthesiology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou City, Henan Province, China
| | - Mingyang Sun
- Department of Anesthesiology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou City, Henan Province, China
| | - Jiaqiang Zhang
- Department of Anesthesiology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou City, Henan Province, China.
| |
Collapse
|
27
|
Ullmann P, Qureshi-Baig K, Rodriguez F, Ginolhac A, Nonnenmacher Y, Ternes D, Weiler J, Gäbler K, Bahlawane C, Hiller K, Haan S, Letellier E. Hypoxia-responsive miR-210 promotes self-renewal capacity of colon tumor-initiating cells by repressing ISCU and by inducing lactate production. Oncotarget 2018; 7:65454-65470. [PMID: 27589845 PMCID: PMC5323168 DOI: 10.18632/oncotarget.11772] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/25/2016] [Indexed: 01/01/2023] Open
Abstract
Low oxygen concentrations (hypoxia) are known to affect the cellular metabolism and have been suggested to regulate a subpopulation of cancer cells with tumorigenic properties, the so-called tumor-initiating cells (TICs). To better understand the mechanism of hypoxia-induced TIC activation, we set out to study the role of hypoxia-responsive miRNAs in recently established colon cancer patient-derived TICs. We were able to show that low oxygen concentrations consistently lead to the upregulation of miR-210 in different primary TIC-enriched cultures. Both stable overexpression of miR-210 and knockdown of its target gene ISCU resulted in enhanced TIC self-renewal. We could validate the tumorigenic properties of miR- 210 in in vivo experiments by showing that ectopic expression of miR-210 results in increased tumor incidence. Furthermore, enhanced miR-210 expression correlated with reduced TCA cycle activity and increased lactate levels. Importantly, by blocking lactate production via inhibition of LDHA, we could reverse the promoting effect of miR-210 on self-renewal capacity, thereby emphasizing the regulatory impact of the glycolytic phenotype on colon TIC properties. Finally, by assessing expression levels in patient tissue, we could demonstrate the clinical relevance of the miR-210/ISCU signaling axis for colorectal carcinoma. Taken together, our study highlights the importance of hypoxia-induced miR-210 in the regulation of colon cancer initiation.
Collapse
Affiliation(s)
- Pit Ullmann
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Komal Qureshi-Baig
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Fabien Rodriguez
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Aurélien Ginolhac
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | | | - Dominik Ternes
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jil Weiler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karoline Gäbler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Christelle Bahlawane
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karsten Hiller
- Luxembourg Centre for Systems Biomedicine, L-4367 Belvaux, Luxembourg
| | - Serge Haan
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Elisabeth Letellier
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| |
Collapse
|
28
|
Wang B, Wang H, Xiong J, Zhou Q, Wu H, Xia L, Li L, Yu Z. A Proteomic Analysis Provides Novel Insights into the Stress Responses of Caenorhabditis elegans towards Nematicidal Cry6A Toxin from Bacillus thuringiensis. Sci Rep 2017; 7:14170. [PMID: 29074967 PMCID: PMC5658354 DOI: 10.1038/s41598-017-14428-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/10/2017] [Indexed: 01/16/2023] Open
Abstract
Cry6A represents a novel family of nematicidal crystal proteins from Bacillus thuringiensis. It has distinctive architecture as well as mechanism of action from Cry5B, a highly focused family of nematicidal crystal proteins, and even from other insecticidal crystal proteins containing the conserved three-domain. However, how nematode defends against Cry6A toxin remains obscure. In this study, the global defense pattern of Caenorhabditis elegans against Cry6Aa2 toxin was investigated by proteomic analysis. In response to Cry6Aa2, 12 proteins with significantly altered abundances were observed from worms, participating in innate immune defense, insulin-like receptor (ILR) signaling pathway, energy metabolism, and muscle assembly. The differentially expressed proteins (DEPs) functioning in diverse biological processes suggest that a variety of defense responses participate in the stress responses of C. elegans to Cry6Aa2. The functional verifications of DEPs suggest that ILR signaling pathway, DIM-1, galectin LEC-6 all are the factors of defense responses to Cry6Aa2. Moreover, Cry6Aa2 also involves in accelerating the metabolic energy production which fulfills the energy demand for the immune responses. In brief, our findings illustrate the global pattern of defense responses of nematode against Cry6A for the first time, and provide a novel insight into the mechanism through which worms respond to Cry6A.
Collapse
Affiliation(s)
- Bing Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Haiwen Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Jing Xiong
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Qiaoni Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Huan Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Liqiu Xia
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Lin Li
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Ziquan Yu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China.
| |
Collapse
|
29
|
Song X, Fiati Kenston SS, Kong L, Zhao J. Molecular mechanisms of nickel induced neurotoxicity and chemoprevention. Toxicology 2017; 392:47-54. [PMID: 29032222 DOI: 10.1016/j.tox.2017.10.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/08/2017] [Accepted: 10/10/2017] [Indexed: 01/05/2023]
Abstract
Nickel (Ni) is widely used in many industrial sectors such as alloy, welding, printing inks, electrical and electronics industries. Excessive environmental or occupational exposure to Ni may result in tumor, contact dermatitis, as well as damages to the nervous system. In recent years, more and more research has demonstrated that Ni induced nerve damages are related to mitochondrial dysfunction. In this paper, we try to characterize Ni induced neurotoxicity as well as the underlying mechanisms, and how to find new drugs for chemoprevention, by reviewing chemicals with neuroprotective effects on Ni induced neurotoxicity.
Collapse
Affiliation(s)
- Xin Song
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Samuel Selorm Fiati Kenston
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Lu Kong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Jinshun Zhao
- Department of Preventative Medicine, Zhejiang Key Laboratory of Pathophysiology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang Province, 315211, People's Republic of China.
| |
Collapse
|
30
|
Hypoxia-inducible factor-1 alpha as a therapeutic target for primary effusion lymphoma. PLoS Pathog 2017; 13:e1006628. [PMID: 28922425 PMCID: PMC5619862 DOI: 10.1371/journal.ppat.1006628] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/28/2017] [Accepted: 09/05/2017] [Indexed: 01/08/2023] Open
Abstract
Primary effusion lymphoma (PEL) is an aggressive B-cell lymphoma with poor prognosis caused by Kaposi’s sarcoma-associated herpesvirus (KSHV). Previous studies have revealed that HIF-1α, which mediates much of the cellular response to hypoxia, plays an important role in life cycle of KSHV. KSHV infection promotes HIF-1α activity, and several KSHV genes are in turn activated by HIF-1α. In this study, we investigated the effects of knocking down HIF-1α in PELs. We observed that HIF-1α knockdown in each of two PEL lines leads to a reduction in both aerobic and anaerobic glycolysis as well as lipid biogenesis, indicating that HIF-1α is necessary for maintaining a metabolic state optimal for growth of PEL. We also found that HIF-1α suppression leads to a substantial reduction in activation of lytic KSHV genes, not only in hypoxia but also in normoxia. Moreover, HIF-1α knockdown led to a decrease in the expression of various KSHV latent genes, including LANA, vCyclin, kaposin, and miRNAs, under both normoxic and hypoxic conditions. These observations provide evidence that HIF-1α plays an important role in PEL even in normoxia. Consistent with these findings, we observed a significant inhibition of growth of PEL in normoxia upon HIF-1α suppression achieved by either HIF-1α knockdown or treatment with PX-478, a small molecule inhibitor of HIF-1α. These results offer further evidence that HIF-1α plays a critical role in the pathogenesis of PEL, and that inhibition of HIF-1α can be a potential therapeutic strategy in this disease. Kaposi’s sarcoma-associated herpesvirus (KSHV) is an oncogenic herpesvirus that causes several malignancies including primary effusion lymphoma (PEL). PEL is an aggressive B-cell lymphoma that usually develops in a hypoxic environment. There is no standard treatment for PEL and it carries a poor prognosis. Previous studies have revealed that certain KSHV-encoded genes are activated by hypoxia-inducible factor 1 (HIF-1), an intracellular factor that mediates much of the cellular response to hypoxia. KSHV in turn can upregulate HIF-1, suggesting HIF-1 might play a substantial role in PEL oncogenesis. Here, we report for the first time the effects of suppressing HIF-1α, an oxygen-sensitive subunit of HIF-1, in PEL tumor cells. We demonstrate that suppressing HIF-1α can dramatically affect the oncogenic metabolic signature of PELs, replication of KSHV, expression of KSHV-encoded oncogenes, and the growth of PEL cells. Findings presented here not only provide new insights into the role of HIF-1α in KSHV-induced tumors but also provide a rationale for using anti-HIF-1α agents as a therapeutic strategy for PEL and potentially other KSHV-associated malignancies.
Collapse
|
31
|
Hodjat M, Rahmani S, Khan F, Niaz K, Navaei–Nigjeh M, Mohammadi Nejad S, Abdollahi M. Environmental toxicants, incidence of degenerative diseases, and therapies from the epigenetic point of view. Arch Toxicol 2017; 91:2577-2597. [DOI: 10.1007/s00204-017-1979-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/04/2017] [Indexed: 01/12/2023]
|
32
|
Millan MJ. Linking deregulation of non-coding RNA to the core pathophysiology of Alzheimer's disease: An integrative review. Prog Neurobiol 2017; 156:1-68. [PMID: 28322921 DOI: 10.1016/j.pneurobio.2017.03.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023]
Abstract
The human genome encodes a vast repertoire of protein non-coding RNAs (ncRNA), some specific to the brain. MicroRNAs, which interfere with the translation of target mRNAs, are of particular interest since their deregulation has been implicated in neurodegenerative disorders like Alzheimer's disease (AD). However, it remains challenging to link the complex body of observations on miRNAs and AD into a coherent framework. Using extensive graphical support, this article discusses how a diverse panoply of miRNAs convergently and divergently impact (and are impacted by) core pathophysiological processes underlying AD: neuroinflammation and oxidative stress; aberrant generation of β-amyloid-42 (Aβ42); anomalies in the production, cleavage and post-translational marking of Tau; impaired clearance of Aβ42 and Tau; perturbation of axonal organisation; disruption of synaptic plasticity; endoplasmic reticulum stress and the unfolded protein response; mitochondrial dysfunction; aberrant induction of cell cycle re-entry; and apoptotic loss of neurons. Intriguingly, some classes of miRNA provoke these cellular anomalies, whereas others act in a counter-regulatory, protective mode. Moreover, changes in levels of certain species of miRNA are a consequence of the above-mentioned anomalies. In addition to miRNAs, circular RNAs, piRNAs, long non-coding RNAs and other types of ncRNA are being increasingly implicated in AD. Overall, a complex mesh of deregulated and multi-tasking ncRNAs reciprocally interacts with core pathophysiological mechanisms underlying AD. Alterations in ncRNAs can be detected in CSF and the circulation as well as the brain and are showing promise as biomarkers, with the ultimate goal clinical exploitation as targets for novel modes of symptomatic and course-altering therapy.
Collapse
Affiliation(s)
- Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, institut de recherche Servier, 125 chemin de ronde, 78290 Croissy sur Seine, France.
| |
Collapse
|
33
|
Distinct Effects of miR-210 Reduction on Neurogenesis: Increased Neuronal Survival of Inflammation But Reduced Proliferation Associated with Mitochondrial Enhancement. J Neurosci 2017; 37:3072-3084. [PMID: 28188219 DOI: 10.1523/jneurosci.1777-16.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 01/19/2023] Open
Abstract
Neurogenesis is essential to brain development and plays a central role in the response to brain injury. Stroke and head trauma stimulate proliferation of endogenous neural stem cells (NSCs); however, the survival of young neurons is sharply reduced by postinjury inflammation. Cellular mitochondria are critical to successful neurogenesis and are a major target of inflammatory injury. Mitochondrial protection was shown to improve survival of young neurons. This study tested whether reducing cellular microRNA-210 (miR-210) would enhance mitochondrial function and improve survival of young murine neurons under inflammatory conditions. Several studies have demonstrated the potential of miR-210 inhibition to enhance and protect mitochondrial function through upregulation of mitochondrial proteins. Here, miR-210 inhibition significantly increased neuronal survival and protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase in differentiating NSC cultures exposed to inflammatory mediators. Unexpectedly, we found that reducing miR-210 significantly attenuated NSC proliferation upon induction of differentiation. Further investigation revealed that increased mitochondrial function suppressed the shift to primarily glycolytic metabolism and reduced mitochondrial length characteristic of dividing cells. Activation of AMP-regulated protein kinase-retinoblastoma signaling is important in NSC proliferation and the reduction of this activation observed by miR-210 inhibition is one mechanism contributing to the reduced proliferation. Postinjury neurogenesis occurs as a burst of proliferation that peaks in days, followed by migration and differentiation over weeks. Our studies suggest that mitochondrial protective miR-210 inhibition should be delayed until after the initial burst of proliferation, but could be beneficial during the prolonged differentiation stage.SIGNIFICANCE STATEMENT Increasing the success of endogenous neurogenesis after brain injury holds therapeutic promise. Postinjury inflammation markedly reduces newborn neuron survival. This study found that enhancement of mitochondrial function by reducing microRNA-210 (miR-210) levels could improve survival of young neurons under inflammatory conditions. miR-210 inhibition protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase. Conversely, we observed decreased precursor cell proliferation likely due to suppression of the AMP-regulated protein kinase-retinoblastoma axis with miR-210 inhibition. Therefore, mitochondrial protection is a double-edged sword: early inhibition reduces proliferation, but inhibition later significantly increases neuroblast survival. This explains in part the contradictory published reports of the effects of miR-210 on neurogenesis.
Collapse
|
34
|
Pinti MV, Hathaway QA, Hollander JM. Role of microRNA in metabolic shift during heart failure. Am J Physiol Heart Circ Physiol 2016; 312:H33-H45. [PMID: 27742689 DOI: 10.1152/ajpheart.00341.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 12/16/2022]
Abstract
Heart failure (HF) is an end point resulting from a number of disease states. The prognosis for HF patients is poor with survival rates precipitously low. Energy metabolism is centrally linked to the development of HF, and it involves the proteomic remodeling of numerous pathways, many of which are targeted to the mitochondrion. microRNAs (miRNA) are noncoding RNAs that influence posttranscriptional gene regulation. miRNA have garnered considerable attention for their ability to orchestrate changes to the transcriptome, and ultimately the proteome, during HF. Recently, interest in the role played by miRNA in the regulation of energy metabolism at the mitochondrion has emerged. Cardiac proteome remodeling during HF includes axes impacting hypertrophy, oxidative stress, calcium homeostasis, and metabolic fuel transition. Although it is established that the pathological environment of hypoxia and hemodynamic stress significantly contribute to the HF phenotype, it remains unclear as to the mechanistic underpinnings driving proteome remodeling. The aim of this review is to present evidence highlighting the role played by miRNA in these processes as a means for linking pathological stimuli with proteomic alteration. The differential expression of proteins of substrate transport, glycolysis, β-oxidation, ketone metabolism, the citric acid cycle (CAC), and the electron transport chain (ETC) are paralleled by the differential expression of miRNA species that modulate these processes. Identification of miRNAs that translocate to cardiomyocyte mitochondria (miR-181c, miR-378) influencing the expression of the mitochondrial genome-encoded transcripts as well as suggested import modulators are discussed. Current insights, applications, and challenges of miRNA-based therapeutics are also described.
Collapse
Affiliation(s)
- Mark V Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; and Mitochondria, Metabolism, and Bioenergentics Working Group, Morgantown, West Virginia
| | - Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; and Mitochondria, Metabolism, and Bioenergentics Working Group, Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; and Mitochondria, Metabolism, and Bioenergentics Working Group, Morgantown, West Virginia
| |
Collapse
|
35
|
Sato T, Kishimoto Y, Asakawa S, Mizuno N, Hiratsuka M, Hirasawa N. Involvement of COX-2 in nickel elution from a wire implanted subcutaneously in mice. Toxicology 2016; 363-364:37-45. [PMID: 27452194 DOI: 10.1016/j.tox.2016.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/21/2016] [Accepted: 07/21/2016] [Indexed: 11/24/2022]
Abstract
Many types of medical alloys include nickel (Ni), and the elution of Ni ions from these materials causes toxicities and inflammation. We have previously reported that inflammation enhances Ni elution, although the molecular mechanisms underlying this effect remain unclear. In this study, we investigated how inflammatory responses enhanced Ni elution in a wire-implantation mouse model. Subcutaneous implantation of Ni wire induced the expression of cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase-1 (mPGES-1) mRNA in the surrounding tissues. Immunostaining analysis showed that cells expressing COX-2 were mainly fibroblast-like cells 8h after implantation of a Ni wire, but were mainly infiltrated leukocytes at 24h. NiCl2 induced the expression of COX-2 mRNA in primary fibroblasts, neutrophils, RAW 264 cells, and THP-1 cells, indicating that Ni ions can induce COX-2 expression in various types of cells. The elution of Ni ions from the implanted Ni wire at 8h was reduced by dexamethasone (Dex), indomethacin (Ind), or celecoxib (Cel) treatment. Ni wire implantation induced an increase in mRNA levels for anaerobic glycolytic pathway components glucose transporter 1 (GLUT1), hexokinase 2 (HK2), lactate dehydrogenase A (LDHA), and monocarboxylate transporter 4 (MCT4); the expression of these genes was also inhibited by Dex, Ind, and Cel. In primary fibroblasts, the expression of these mRNAs and the production of lactate were induced by NiCl2 and further potentiated by PGE2. Furthermore, Ni wire-induced infiltration of inflammatory leukocytes was significantly reduced by Dex, Ind, or Cel. Depletion of neutrophils with a specific antibody caused reduction of both leukocyte infiltration and Ni elution. These results indicate that Ni ions eluted from wire induced COX-2 expression, which further promoted elution of Ni ions by increasing lactate production and leukocyte infiltration. Since COX inhibitors and Dex reduced the elution of Ni ions, these drugs may be useful for prevention of metal-related inflammation and allergy.
Collapse
Affiliation(s)
- Taiki Sato
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yu Kishimoto
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Sanki Asakawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Natsumi Mizuno
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masahiro Hiratsuka
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
36
|
Ma Q, Chen C, Deng P, Zhu G, Lin M, Zhang L, Xu S, He M, Lu Y, Duan W, Pi H, Cao Z, Pei L, Li M, Liu C, Zhang Y, Zhong M, Zhou Z, Yu Z. Extremely Low-Frequency Electromagnetic Fields Promote In Vitro Neuronal Differentiation and Neurite Outgrowth of Embryonic Neural Stem Cells via Up-Regulating TRPC1. PLoS One 2016; 11:e0150923. [PMID: 26950212 PMCID: PMC4780708 DOI: 10.1371/journal.pone.0150923] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/22/2016] [Indexed: 02/07/2023] Open
Abstract
Exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) can enhance hippocampal neurogenesis in adult mice. However, little is focused on the effects of ELF-EMFs on embryonic neurogenesis. Here, we studied the potential effects of ELF-EMFs on embryonic neural stem cells (eNSCs). We exposed eNSCs to ELF-EMF (50 Hz, 1 mT) for 1, 2, and 3 days with 4 hours per day. We found that eNSC proliferation and maintenance were significantly enhanced after ELF-EMF exposure in proliferation medium. ELF-EMF exposure increased the ratio of differentiated neurons and promoted the neurite outgrowth of eNSC-derived neurons without influencing astrocyes differentiation and the cell apoptosis. In addition, the expression of the proneural genes, NeuroD and Ngn1, which are crucial for neuronal differentiation and neurite outgrowth, was increased after ELF-EMF exposure. Moreover, the expression of transient receptor potential canonical 1 (TRPC1) was significantly up-regulated accompanied by increased the peak amplitude of intracellular calcium level induced by ELF-EMF. Furthermore, silencing TRPC1 expression eliminated the up-regulation of the proneural genes and the promotion of neuronal differentiation and neurite outgrowth induced by ELF-EMF. These results suggest that ELF-EMF exposure promotes the neuronal differentiation and neurite outgrowth of eNSCs via up-regulation the expression of TRPC1 and proneural genes (NeuroD and Ngn1). These findings also provide new insights in understanding the effects of ELF-EMF exposure on embryonic brain development.
Collapse
Affiliation(s)
- Qinlong Ma
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Gang Zhu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Min Lin
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Shangcheng Xu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Mindi He
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Weixia Duan
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Zhengwang Cao
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Liping Pei
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Min Li
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Chuan Liu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yanwen Zhang
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Min Zhong
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
- * E-mail: (ZY); (ZZ)
| | - Zhengping Yu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
- Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education of China, Third Military Medical University, Chongqing, China
- * E-mail: (ZY); (ZZ)
| |
Collapse
|
37
|
Chen S, McKinney GJ, Nichols KM, Colbourne JK, Sepúlveda MS. Novel Cadmium Responsive MicroRNAs in Daphnia pulex. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:14605-13. [PMID: 26550707 DOI: 10.1021/acs.est.5b03988] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Daphnia pulex is a widely used toxicological model and is known for its sensitivity to cadmium (Cd). Recent research suggests that microRNAs (miRNAs) play a critical role in animal responses to heavy metals. To investigate the functions of D. pulex miRNAs under Cd exposure, we analyzed the miRNA profiles of D. pulex after 48 h using miRNA microarrays and validated our findings by q-PCR. miRNA dpu-let-7 was identified as a stably expressed gene and used as a reference. We identified 22 and 21 differentially expressed miRNAs under low (20 μg/L CdCl2) and high-exposure (40 μg/L CdCl2) concentrations compared to controls, respectively. Cellular functions of predicted miRNA target Cd-responsive genes included oxidative stress, ion transport, mitochondrial damage, and DNA repair. An insulin-related network was also identified in relation to several Cd-responsive miRNAs. The expression of three predicted target genes for miR-71 and miR-210 were evaluated, and expression of two of them (SCN2A and SLC31A1) was negatively correlated with the expression of their regulator miRNAs. We show miR-210 is hypoxia-responsive in D. pulex and propose Cd and hypoxia induce miR-210 via a same HIF1α modulated pathway. Collectively, this research advances our understanding on the role of miRNAs in response to heavy-metal exposure.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Forestry and Natural Resources, Purdue University , West Lafayette, Indiana, 47907 United States
| | - Garrett J McKinney
- School of Aquatic and Fishery Sciences, University of Washington , Seattle, Washington, 98195 United States
| | - Krista M Nichols
- Department of Forestry and Natural Resources, Purdue University , West Lafayette, Indiana, 47907 United States
- Conservation Biology Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Ocean and Atmospheric Administration , Seattle, Washington, 98112 United States
| | - John K Colbourne
- Environmental Genomics Group, School of Biosciences, University of Birmingham , Edgbaston, Birmingham B15 2TT, U.K
| | - Maria S Sepúlveda
- Department of Forestry and Natural Resources, Purdue University , West Lafayette, Indiana, 47907 United States
| |
Collapse
|
38
|
Millan MJ. The epigenetic dimension of Alzheimer's disease: causal, consequence, or curiosity? DIALOGUES IN CLINICAL NEUROSCIENCE 2015. [PMID: 25364287 PMCID: PMC4214179 DOI: 10.31887/dcns.2014.16.3/mmillan] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Early-onset, familial Alzheimer's disease (AD) is rare and may be attributed to disease-causinq mutations. By contrast, late onset, sporadic (non-Mendelian) AD is far more prevalent and reflects the interaction of multiple genetic and environmental risk factors, together with the disruption of epigenetic mechanisms controlling gene expression. Accordingly, abnormal patterns of histone acetylation and methylation, as well as anomalies in global and promoter-specific DNA methylation, have been documented in AD patients, together with a deregulation of noncoding RNA. In transgenic mouse models for AD, epigenetic dysfunction is likewise apparent in cerebral tissue, and it has been directly linked to cognitive and behavioral deficits in functional studies. Importantly, epigenetic deregulation interfaces with core pathophysiological processes underlying AD: excess production of Aβ42, aberrant post-translational modification of tau, deficient neurotoxic protein clearance, axonal-synaptic dysfunction, mitochondrial-dependent apoptosis, and cell cycle re-entry. Reciprocally, DNA methylation, histone marks and the levels of diverse species of microRNA are modulated by Aβ42, oxidative stress and neuroinflammation. In conclusion, epigenetic mechanisms are broadly deregulated in AD mainly upstream, but also downstream, of key pathophysiological processes. While some epigenetic shifts oppose the evolution of AD, most appear to drive its progression. Epigenetic changes are of irrefutable importance for AD, but they await further elucidation from the perspectives of pathogenesis, biomarkers and potential treatment.
Collapse
Affiliation(s)
- Mark J Millan
- Pole of Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy-sur-Seine, France
| |
Collapse
|
39
|
Uppala R, McKinney RW, Brant KA, Fabisiak JP, Goetzman ES. Nickel inhibits mitochondrial fatty acid oxidation. Biochem Biophys Res Commun 2015; 463:806-10. [PMID: 26051273 DOI: 10.1016/j.bbrc.2015.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/02/2015] [Indexed: 12/17/2022]
Abstract
Nickel exposure is associated with changes in cellular energy metabolism which may contribute to its carcinogenic properties. Here, we demonstrate that nickel strongly represses mitochondrial fatty acid oxidation-the pathway by which fatty acids are catabolized for energy-in both primary human lung fibroblasts and mouse embryonic fibroblasts. At the concentrations used, nickel suppresses fatty acid oxidation without globally suppressing mitochondrial function as evidenced by increased glucose oxidation to CO2. Pre-treatment with l-carnitine, previously shown to prevent nickel-induced mitochondrial dysfunction in neuroblastoma cells, did not prevent the inhibition of fatty acid oxidation. The effect of nickel on fatty acid oxidation occurred only with prolonged exposure (>5 h), suggesting that direct inhibition of the active sites of metabolic enzymes is not the mechanism of action. Nickel is a known hypoxia-mimetic that activates hypoxia inducible factor-1α (HIF1α). Nickel-induced inhibition of fatty acid oxidation was blunted in HIF1α knockout fibroblasts, implicating HIF1α as one contributor to the mechanism. Additionally, nickel down-regulated the protein levels of the key fatty acid oxidation enzyme very long-chain acyl-CoA dehydrogenase (VLCAD) in a dose-dependent fashion. In conclusion, inhibition of fatty acid oxidation by nickel, concurrent with increased glucose metabolism, represents a form of metabolic reprogramming that may contribute to nickel-induced carcinogenesis.
Collapse
Affiliation(s)
- Radha Uppala
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Richard W McKinney
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Kelly A Brant
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15219, USA
| | - James P Fabisiak
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15219, USA
| | - Eric S Goetzman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA.
| |
Collapse
|
40
|
Li Y, Wei Y, Guo J, Cheng Y, He W. Interactional role of microRNAs and bHLH-PAS proteins in cancer (Review). Int J Oncol 2015; 47:25-34. [PMID: 25997457 DOI: 10.3892/ijo.2015.3007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/08/2015] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are recognized as an emerging class of master regulators that regulate human gene expression at the post-transcriptional level and are involved in many normal and pathological cellular processes. Mammalian basic HLH (helix-loop-helix)-PER-ARNT-SIM (bHLH-PAS) proteins are heterodimeric transcriptional regulators that sense and respond to environmental signals (such as chemical pollutants) or to physiological signals (for instance hypoxia). In the normal state, bHLH-PAS proteins are responsible for multiple critical aspects of physiology to ensure the cell accurate homeostasis, but dysregulation of these proteins has been shown to contribute to carcinogenic events such as tumor initiation, promotion, and progression. Increasing epidemiological and experimental studies have shown that bHLH-PAS proteins regulate a panel of miRNAs, whereas some miRNAs also target bHLH-PAS proteins. The interaction between miRNAs and certain bHLH-PAS proteins [hypoxia-inducible factor (HIF) and aryl hydrocarbon receptor (AHR)] is relevant to many vital events associated with tumorigenesis. This review will summarize recent findings on the interesting and complicated underlying mechanisms that miRNAs interact with HIFs or AHR in tumors, hopefully to benefit the discovery of novel drug-interfering targets for cancer therapy.
Collapse
Affiliation(s)
- Yumin Li
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yucai Wei
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Jiwu Guo
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yusheng Cheng
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Wenting He
- The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
41
|
Duan WX, He MD, Mao L, Qian FH, Li YM, Pi HF, Liu C, Chen CH, Lu YH, Cao ZW, Zhang L, Yu ZP, Zhou Z. NiO nanoparticles induce apoptosis through repressing SIRT1 in human bronchial epithelial cells. Toxicol Appl Pharmacol 2015; 286:80-91. [PMID: 25840356 DOI: 10.1016/j.taap.2015.03.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 03/20/2015] [Accepted: 03/21/2015] [Indexed: 02/06/2023]
Abstract
With application of nano-sized nickel-containing particles (Nano-Ni) expanding, the health concerns about their adverse effects on the pulmonary system are increasing. However, the mechanisms for the pulmonary toxicity of these materials remain unclear. In the present study, we focused on the impacts of NiO nanoparticles (NiONPs) on sirtuin1 (SIRT1), a NAD-dependent deacetylase, and investigated whether SIRT1 was involved in NiONPs-induced apoptosis. Although the NiONPs tended to agglomerate in fluid medium, they still entered into the human bronchial epithelial cells (BEAS-2B) and released Ni(2+) inside the cells. NiONPs at doses of 5, 10, and 20μg/cm(2) inhibited the cell viability. NiONPs' produced cytotoxicity was demonstrated through an apoptotic process, indicated by increased numbers of Annexin V positive cells and caspase-3 activation. The expression of SIRT1 was markedly down-regulated by the NiONPs, accompanied by the hyperacetylation of p53 (tumor protein 53) and overexpression of Bax (Bcl-2-associated X protein). However, overexpression of SIRT1 through resveratrol treatment or transfection clearly attenuated the NiONPs-induced apoptosis and activation of p53 and Bax. Our results suggest that the repression of SIRT1 may underlie the NiONPs-induced apoptosis via p53 hyperacetylation and subsequent Bax activation. Because SIRT1 participates in multiple biologic processes by deacetylation of dozens of substrates, this knowledge of the impact of NiONPs on SIRT1 may lead to an improved understanding of the toxic mechanisms of Nano-Ni and provide a molecular target to antagonize Nano-Ni toxicity.
Collapse
Affiliation(s)
- Wei-Xia Duan
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Min-Di He
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Lin Mao
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Feng-Hua Qian
- Department of Hematology, Southwest Hospital, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Yu-Ming Li
- Institute of Hepatobiliary Surgery, XinQiao Hospital, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Hui-Feng Pi
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Chuan Liu
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Chun-Hai Chen
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Yong-Hui Lu
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Zheng-Wang Cao
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Lei Zhang
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Zheng-Ping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Zhou Zhou
- Department of Occupational Health, Third Military Medical University, Chongqing 400038, People's Republic of China.
| |
Collapse
|
42
|
Dang K, Myers KA. The role of hypoxia-induced miR-210 in cancer progression. Int J Mol Sci 2015; 16:6353-72. [PMID: 25809609 PMCID: PMC4394536 DOI: 10.3390/ijms16036353] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 12/23/2022] Open
Abstract
Prolonged hypoxia, the event of insufficient oxygen, is known to upregulate tumor development and growth by promoting the formation of a neoplastic environment. The recent discovery that a subset of cellular microRNAs (miRs) are upregulated during hypoxia, where they function to promote tumor development, highlights the importance of hypoxia-induced miRs as targets for continued investigation. miRs are short, non-coding transcripts involved in gene expression and regulation. Under hypoxic conditions, miR-210 becomes highly upregulated in response to hypoxia inducing factors (HIFs). HIF-1α drives miR-210’s overexpression and the resultant alteration of cellular processes, including cell cycle regulation, mitochondria function, apoptosis, angiogenesis and metastasis. Here we discuss hypoxia-induced dysregulation of miR-210 and the resultant changes in miR-210 protein targets that regulate cancer progression. Potential methods of targeting miR-210 as a therapeutic tool are also explored.
Collapse
Affiliation(s)
- Kyvan Dang
- Department of Biological Sciences, University of the Sciences, 600 S. 43rd Str., Philadelphia, PA 19104, USA.
| | - Kenneth A Myers
- Department of Biological Sciences, University of the Sciences, 600 S. 43rd Str., Philadelphia, PA 19104, USA.
| |
Collapse
|
43
|
Shi Z, Yu H, Sun Y, Yang C, Lian H, Cai P. The Energy Metabolism in Caenorhabditis elegans under The Extremely Low-Frequency Electromagnetic Field Exposure. Sci Rep 2015; 5:8471. [PMID: 25683579 PMCID: PMC4329544 DOI: 10.1038/srep08471] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/21/2015] [Indexed: 02/08/2023] Open
Abstract
A literal mountain of documentation generated in the past five decades showing unmistakable health hazards associated with extremely low-frequency electromagnetic fields (ELF-EMFs) exposure. However, the relation between energy mechanism and ELF-EMF exposure is poorly understood. In this study, Caenorhabditis elegans was exposed to 50 Hz ELF-EMF at intensities of 0.5, 1, 2, and 3 mT, respectively. Their metabolite variations were analyzed by GC-TOF/MS-based metabolomics. Although minimal metabolic variations and no regular pattern were observed, the contents of energy metabolism-related metabolites such as pyruvic acid, fumaric acid, and L-malic acid were elevated in all the treatments. The expressions of nineteen related genes that encode glycolytic enzymes were analyzed by using quantitative real-time PCR. Only genes encoding GAPDH were significantly upregulated (P < 0.01), and this result was further confirmed by western blot analysis. The enzyme activity of GAPDH was increased (P < 0.01), whereas the total intracellular ATP level was decreased. While no significant difference in lifespan, hatching rate and reproduction, worms exposed to ELF-EMF exhibited less food consumption compared with that of the control (P < 0.01). In conclusion, C. elegans exposed to ELF-EMF have enhanced energy metabolism and restricted dietary, which might contribute to the resistance against exogenous ELF-EMF stress.
Collapse
Affiliation(s)
- Zhenhua Shi
- 1] Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China [2] University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, P. R. China
| | - Hui Yu
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| | - Yongyan Sun
- 1] Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China [2] University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, P. R. China
| | - Chuanjun Yang
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| | - Huiyong Lian
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| | - Peng Cai
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| |
Collapse
|
44
|
The neuroprotective effects of taurine against nickel by reducing oxidative stress and maintaining mitochondrial function in cortical neurons. Neurosci Lett 2015; 590:52-7. [PMID: 25637701 DOI: 10.1016/j.neulet.2015.01.065] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/16/2015] [Accepted: 01/26/2015] [Indexed: 11/20/2022]
Abstract
Previous studies have indicated that oxidative stress and mitochondrial dysfunction are involved in the toxicity of nickel. Taurine is recognized as an efficient antioxidant and is essential for mitochondrial function. To investigate whether taurine could protect against the neurotoxicity of nickel, we exposed primary cultured cortical neurons to various concentrations of nickel chloride (NiCl2; 0.5mM, 1mM and 2mM) for 24h or to 1mM NiCl2 for various periods (0 h, 12h, 24h and 48 h). Our results showed that taurine efficiently reduced lactate dehydrogenase (LDH) release induced by NiCl2. Along with this protective effect, taurine pretreatment not only significantly reversed the increase of ROS production and mitochondrial superoxide concentration, but also attenuated the decrease of superoxide dismutase (SOD) activity and glutathione (GSH) concentration in neurons exposed to NiCl2 for 24h. Moreover, nickel exposure reduced ATP production, disrupted the mitochondrial membrane potential and decreased mtDNA content. These types of oxidative damage in the mitochondria were efficiently ameliorated by taurine pretreatment. Taken together, our results indicate that the neuroprotective effects of taurine against the toxicity of nickel might largely depend on its roles in reducing oxidative stress and improving mitochondrial function. Taurine may have great pharmacological potential in treating the adverse effects of nickel in the nervous system.
Collapse
|
45
|
Millan MJ. The epigenetic dimension of Alzheimer's disease: causal, consequence, or curiosity? DIALOGUES IN CLINICAL NEUROSCIENCE 2014; 16:373-93. [PMID: 25364287 PMCID: PMC4214179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/10/2024]
Abstract
Early-onset, familial Alzheimer's disease (AD) is rare and may be attributed to disease-causinq mutations. By contrast, late onset, sporadic (non-Mendelian) AD is far more prevalent and reflects the interaction of multiple genetic and environmental risk factors, together with the disruption of epigenetic mechanisms controlling gene expression. Accordingly, abnormal patterns of histone acetylation and methylation, as well as anomalies in global and promoter-specific DNA methylation, have been documented in AD patients, together with a deregulation of noncoding RNA. In transgenic mouse models for AD, epigenetic dysfunction is likewise apparent in cerebral tissue, and it has been directly linked to cognitive and behavioral deficits in functional studies. Importantly, epigenetic deregulation interfaces with core pathophysiological processes underlying AD: excess production of Aβ42, aberrant post-translational modification of tau, deficient neurotoxic protein clearance, axonal-synaptic dysfunction, mitochondrial-dependent apoptosis, and cell cycle re-entry. Reciprocally, DNA methylation, histone marks and the levels of diverse species of microRNA are modulated by Aβ42, oxidative stress and neuroinflammation. In conclusion, epigenetic mechanisms are broadly deregulated in AD mainly upstream, but also downstream, of key pathophysiological processes. While some epigenetic shifts oppose the evolution of AD, most appear to drive its progression. Epigenetic changes are of irrefutable importance for AD, but they await further elucidation from the perspectives of pathogenesis, biomarkers and potential treatment.
Collapse
Affiliation(s)
- Mark J. Millan
- Pole of Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy-sur-Seine, France
| |
Collapse
|