1
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Yu X, Li Y, Tian X, Zang X, Yang S, Qiao H, Zhu C, Moussian B, Wang Y. Pb exposure causes non-linear accumulation of Pb in D. melanogaster controlled by metallothionein B and exerts ecological effects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165680. [PMID: 37499811 DOI: 10.1016/j.scitotenv.2023.165680] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/09/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Pb pollution can harm human health and the ecosystem. Therefore, it is worthwhile to study the metabolic processes of heavy metals in individual bodies and their influence on ecological systems. In this work, we analyzed the genetic responses and physiological changes of D. melanogaster which took diets exposed to different doses of Pb using transcriptomic analysis, ICP-MS, and various other physiological methods. We found that the Pb accumulated in D. melanogaster in a nonlinear pattern with the increase of Pb content in food. Metallothioneins (Mtns), especially the MtnB directly affects the accumulation and excretion of metal Pb in D. melanogaster, and causes the nonlinear accumulation. Metal regulatory transcription factor-1 (MTF-1) is involved in the regulation of Pb-induced high expressions of Mtns. Furthermore, an interaction between the metal metabolism pathway and xenobiotic response pathway leads to the cross-tolerances of Pb-exposed D. melanogaster to insecticides and other toxins. The oxidative stress induced by Pb toxicity may be the bridge between them. Our findings provide a physiological and molecular genetic basis for further study of the accumulation and metabolism of Pb in D. melanogaster.
Collapse
Affiliation(s)
- Xiaoyu Yu
- School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, China
| | - Ying Li
- School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, China
| | - Xiaohan Tian
- School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, China
| | - Xiya Zang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, China
| | - Shuyu Yang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, China
| | - Huanhuan Qiao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072 Tianjin, China
| | - Chunfeng Zhu
- School of Life Sciences, Tianjin University, 300072 Tianjin, China
| | - Bernard Moussian
- Animal Genetics, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, China.
| |
Collapse
|
3
|
Khan S, Lang M. A Comprehensive Review on the Roles of Metals Mediating Insect-Microbial Pathogen Interactions. Metabolites 2023; 13:839. [PMID: 37512546 PMCID: PMC10384549 DOI: 10.3390/metabo13070839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Insects and microbial pathogens are ubiquitous and play significant roles in various biological processes, while microbial pathogens are microscopic organisms that can cause diseases in multiple hosts. Insects and microbial pathogens engage in diverse interactions, leveraging each other's presence. Metals are crucial in shaping these interactions between insects and microbial pathogens. However, metals such as Fe, Cu, Zn, Co, Mo, and Ni are integral to various physiological processes in insects, including immune function and resistance against pathogens. Insects have evolved multiple mechanisms to take up, transport, and regulate metal concentrations to fight against pathogenic microbes and act as a vector to transport microbial pathogens to plants and cause various plant diseases. Hence, it is paramount to inhibit insect-microbe interaction to control pathogen transfer from one plant to another or carry pathogens from other sources. This review aims to succinate the role of metals in the interactions between insects and microbial pathogens. It summarizes the significance of metals in the physiology, immune response, and competition for metals between insects, microbial pathogens, and plants. The scope of this review covers these imperative metals and their acquisition, storage, and regulation mechanisms in insect and microbial pathogens. The paper will discuss various scientific studies and sources, including molecular and biochemical studies and genetic and genomic analysis.
Collapse
Affiliation(s)
- Subhanullah Khan
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minglin Lang
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- College of Life Science, Agricultural University of Hebei, Baoding 071000, China
| |
Collapse
|
4
|
Pandey P, Wall PK, Lopez SR, Dubuisson OS, Zunica ERM, Dantas WS, Kirwan JP, Axelrod CL, Johnson AE. A familial natural short sleep mutation promotes healthy aging and extends lifespan in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538137. [PMID: 37163058 PMCID: PMC10168263 DOI: 10.1101/2023.04.25.538137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Sleep loss typically imposes negative effects on animal health. However, humans with a rare genetic mutation in the dec2 gene ( dec2 P384R ) present an exception; these individuals sleep less without the usual effects associated with sleep deprivation. Thus, it has been suggested that the dec2 P384R mutation activates compensatory mechanisms that allows these individuals to thrive with less sleep. To test this directly, we used a Drosophila model to study the effects of the dec2 P384R mutation on animal health. Expression of human dec2 P384R in fly sleep neurons was sufficient to mimic the short sleep phenotype and, remarkably, dec2 P384R mutants lived significantly longer with improved health despite sleeping less. The improved physiological effects were enabled, in part, by enhanced mitochondrial fitness and upregulation of multiple stress response pathways. Moreover, we provide evidence that upregulation of pro-health pathways also contributes to the short sleep phenotype, and this phenomenon may extend to other pro-longevity models.
Collapse
|
5
|
Yu Y, Chen J, An L, Huang T, Wang W, Cheng Z, Wang L, Xu X, Zhao Z, Fu X, Ma J. Knockdown of phosphatases of regenerating liver-1 prolongs the lifespan of Caenorhabditis elegans via activating DAF-16/FOXO. FASEB J 2023; 37:e22844. [PMID: 36906287 DOI: 10.1096/fj.202202003r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/20/2023] [Accepted: 02/15/2023] [Indexed: 03/13/2023]
Abstract
Phosphatases of regenerating liver (PRLs) are dual-specificity protein phosphatases. The aberrant expression of PRLs threatens human health, but their biological functions and pathogenic mechanisms are unclear yet. Herein, the structure and biological functions of PRLs were investigated using the Caenorhabditis elegans (C. elegans). Structurally, this phosphatase in C. elegans, named PRL-1, consisted of a conserved signature sequence WPD loop and a single C(X)5 R domain. Besides, by Western blot, immunohistochemistry and immunofluorescence staining, PRL-1 was proved to mainly express in larval stages and express in intestinal tissues. Afterward, by feeding-based RNA-interference method, knockdown of prl-1 prolonged the lifespan of C. elegans but also improved their healthspan, such as locomotion, pharyngeal pumping frequency, and defecation interval time. Furthermore, the above effects of prl-1 appeared to be taken without acting on germline signaling, diet restriction pathway, insulin/insulin-like growth factor 1 signaling pathway, and SIR-2.1 but through a DAF-16-dependent pathway. Moreover, knockdown of prl-1 induced the nuclear translocation of DAF-16, and upregulated the expression of daf-16, sod-3, mtl-1, and ctl-2. Finally, suppression of prl-1 also reduced the ROS. In conclusion, suppression of prl-1 enhanced the lifespan and survival quality of C. elegans, which provides a theoretical basis for the pathogenesis of PRLs in related human diseases.
Collapse
Affiliation(s)
- Yaoru Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jing Chen
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lu An
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tianci Huang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Wenbo Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Ziqi Cheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xuesong Xu
- Clinical Laboratory of China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zhizhuang Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xueqi Fu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Junfeng Ma
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
6
|
Zabihihesari A, Parand S, Coulthard AB, Molnar A, Hilliker AJ, Rezai P. An in-vivo microfluidic assay reveals cardiac toxicity of heavy metals and the protective effect of metal responsive transcription factor (MTF-1) in Drosophila model. 3 Biotech 2022; 12:279. [PMID: 36275358 PMCID: PMC9478020 DOI: 10.1007/s13205-022-03336-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/23/2022] [Indexed: 11/01/2022] Open
Abstract
Previous toxicity assessments of heavy metals on Drosophila are limited to investigating the survival, development rate, and climbing behaviour by oral administration while cardiac toxicity of these elements have not been investigated. We utilized a microfluidic device to inject known dosages of zinc (Zn) or cadmium (Cd) into the larvae's hemolymph to expose their heart directly and study their heart rate and arrhythmicity. The effect of heart-specific overexpression of metal responsive transcription factor (MTF-1) on different heartbeat parameters and survival of Drosophila larvae was investigated. The heart rate of wild-type larvae decreased by 24.8% or increased by 11.9%, 15 min after injection of 40 nL of 100 mM Zn or 10 mM Cd solution, respectively. The arrhythmicity index of wild-type larvae increased by 58.2% or 76.8%, after injection of Zn or Cd, respectively. MTF-1 heart overexpression ameliorated these effects completely. Moreover, it increased larvae's survival to pupal and adulthood stages and prolonged the longevity of flies injected with Zn and Cd. Our microfluidic-based cardiac toxicity assay illustrated that heart is an acute target of heavy metals toxicity, and MTF-1 overexpression in this tissue can ameliorate cardiac toxicity of Zn and Cd. The method can be used for cardiotoxicity assays with other pollutants in the future. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03336-7.
Collapse
Affiliation(s)
- Alireza Zabihihesari
- Department of Mechanical Engineering, York University, BRG 433B, 4700 Keele St, Toronto, ON M3J 1P3 Canada
| | - Shahrzad Parand
- Department of Psychology, Faculty of Health, York University, Toronto, ON Canada
| | | | | | | | - Pouya Rezai
- Department of Mechanical Engineering, York University, BRG 433B, 4700 Keele St, Toronto, ON M3J 1P3 Canada
| |
Collapse
|
7
|
Xiao G. Molecular physiology of zinc in Drosophila melanogaster. CURRENT OPINION IN INSECT SCIENCE 2022; 51:100899. [PMID: 35276390 DOI: 10.1016/j.cois.2022.100899] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 06/14/2023]
Abstract
New research in Drosophila melanogaster has revealed the molecular mechanisms of zinc involvement in many biological processes. A newly discovered Metallothionein is predicted to have a higher zinc specificity than the other isoforms. Zinc negatively regulates tyrosine hydroxylase activity by antagonizing iron binding, thus rendering the enzyme ineffective or non-functional. The identification of a new chaperone of the protein disulfide isomerase family provided mechanistic insight into the protein trafficking defects caused by zinc dyshomeostasis in the secretory pathway. Insect models of tumor pathogenesis indicate that zinc regulates the structural stabilization of cells by transcriptionally regulating matrix metalloproteinases while zinc dyshomeostasis in the secretory pathway modulates cell signaling through endoplastic recticulum stress.
Collapse
Affiliation(s)
- Guiran Xiao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China.
| |
Collapse
|
8
|
Hao LY, Zhang M, Tao Y, Xu H, Liu Q, Yang K, Wei R, Zhou H, Jin T, Liu XD, Xue Z, Shen W, Cao JL, Pan Z. miRNA-22 Upregulates Mtf1 in Dorsal Horn Neurons and Is Essential for Inflammatory Pain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8622388. [PMID: 35242280 PMCID: PMC8886789 DOI: 10.1155/2022/8622388] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 11/27/2022]
Abstract
Chronic inflammatory pain seriously affects patients' quality of life because of a paucity of effective clinical treatments caused, at least in part, by lack of full understanding of the underlying mechanisms. miRNAs are known to be involved in inflammatory pain via silencing or degrading of target mRNA in the cytoplasm. The present study provides a novel mechanism by which miRNA-22 positively regulates metal-regulatory transcription factor 1 (Mtf1) in the nuclei of neurons in the dorsal horn of the spinal cord. We found that miRNA-22 was significantly increased in the dorsal horn of mice with either inflammatory pain induced by plantar injection of complete Freund's adjuvant (CFA) or neuropathic pain induced by unilateral sciatic nerve chronic constrictive injury (CCI). Knocking down or blocking miRNA-22 alleviated CFA-induced mechanical allodynia and heat hyperalgesia, whereas overexpressing miRNA-22 produced pain-like behaviors. Mechanistically, the increased miRNA-22 binds directly to the Mtf1 promoter to recruit RNA polymerase II and elevate Mtf1 expression. The increased Mtf1 subsequently enhances spinal central sensitization, as evidenced by increased expression of p-ERK1/2, GFAP, and c-Fos in the dorsal horn. Our findings suggest that the miRNA-22-Mtf1 signaling axis in the dorsal horn plays a critical role in the induction and maintenance of inflammatory pain. This signaling pathway may be a promising therapeutic target in inflammatory pain.
Collapse
Affiliation(s)
- Ling-Yun Hao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Ming Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Yang Tao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Hengjun Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Qiaoqiao Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Kehui Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Runa Wei
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Huimin Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Tong Jin
- Department of Pain, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xiao-Dan Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Zhouya Xue
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| | - Wen Shen
- Department of Pain, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Zhiqiang Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221004, China
| |
Collapse
|
9
|
Slobodian MR, Petahtegoose JD, Wallis AL, Levesque DC, Merritt TJS. The Effects of Essential and Non-Essential Metal Toxicity in the Drosophila melanogaster Insect Model: A Review. TOXICS 2021; 9:269. [PMID: 34678965 PMCID: PMC8540122 DOI: 10.3390/toxics9100269] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
The biological effects of environmental metal contamination are important issues in an industrialized, resource-dependent world. Different metals have different roles in biology and can be classified as essential if they are required by a living organism (e.g., as cofactors), or as non-essential metals if they are not. While essential metal ions have been well studied in many eukaryotic species, less is known about the effects of non-essential metals, even though essential and non-essential metals are often chemically similar and can bind to the same biological ligands. Insects are often exposed to a variety of contaminated environments and associated essential and non-essential metal toxicity, but many questions regarding their response to toxicity remain unanswered. Drosophila melanogaster is an excellent insect model species in which to study the effects of toxic metal due to the extensive experimental and genetic resources available for this species. Here, we review the current understanding of the impact of a suite of essential and non-essential metals (Cu, Fe, Zn, Hg, Pb, Cd, and Ni) on the D. melanogaster metal response system, highlighting the knowledge gaps between essential and non-essential metals in D. melanogaster. This review emphasizes the need to use multiple metals, multiple genetic backgrounds, and both sexes in future studies to help guide future research towards better understanding the effects of metal contamination in general.
Collapse
Affiliation(s)
| | | | | | | | - Thomas J. S. Merritt
- Faculty of Science and Engineering, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON P3E 2C6, Canada; (M.R.S.); (J.D.P.); (A.L.W.); (D.C.L.)
| |
Collapse
|
10
|
Talukder M, Bi SS, Jin HT, Ge J, Zhang C, Lv MW, Li JL. Cadmium induced cerebral toxicity via modulating MTF1-MTs regulatory axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 285:117083. [PMID: 33965856 DOI: 10.1016/j.envpol.2021.117083] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/04/2021] [Accepted: 04/01/2021] [Indexed: 06/12/2023]
Abstract
Metal-responsive transcription factor 1 (MTF1) participates in redox homeostasis and heavy metals detoxification via regulating the expression of metal responsive genes. However, the exact role of MTF1 in Cd-induced cerebral toxicity remains unclear. Herein, we explored the mechanism of Cd-elicited cerebral toxicity through modulating MTF1/MTs pathway in chicken cerebrum exposed to different concentrations of Cd (35 mg, 70 mg, and 140 mg/kg CdCl2) via diet. Notably, cerebral tissues showed varying degrees of microstructural changes under Cd exposure. Cd exposure significantly up-regulated the expression of metal transporters (DMT1, ZIP8, and ZIP10) with concomitant elevated Cd level, as determined by ICP-MS. Cd significantly altered other cerebral biometals concentrations (particularly, Zn, Fe, Se, Cr, Mo, and Pb) and redox balance, resulting in increased cerebral oxidative stress. More importantly, Cd exposure suppressed MTF1 mRNA and nuclear protein levels and its target metal-responsive genes, notably metallothioneins (MT1 and MT2), and Fe and Cu transporter genes (FPN1, ATOX1, and XIAP). Moreover, Cd disrupted the regulation of expression of selenoproteome (particularly, GPxs and SelW), and cerebral Se level. Overall, our data revealed that molecular mechanisms associated with Cd-induced cerebral damage might include over-expression of DMT1, ZIP8 and ZIP10, and suppression of MTF1 and its main target metal-responsive genes as well as several selenoproteins.
Collapse
Affiliation(s)
- Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh
| | - Shao-Shuai Bi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hai-Tao Jin
- Quality and Safety Institute of Agricultural Products, Heilongjiang Academy of Agricultural Sciences, Harbin, 150010, China
| | - Jing Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Cong Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, 450046, Zhengzhou, Henan, China
| | - Mei-Wei Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
11
|
Torres-Velarde JM, Kolora SRR, Khudyakov JI, Crocker DE, Sudmant PH, Vázquez-Medina JP. Elephant seal muscle cells adapt to sustained glucocorticoid exposure by shifting their metabolic phenotype. Am J Physiol Regul Integr Comp Physiol 2021; 321:R413-R428. [PMID: 34260302 DOI: 10.1152/ajpregu.00052.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022]
Abstract
Elephant seals experience natural periods of prolonged food deprivation while breeding, molting, and undergoing postnatal development. Prolonged food deprivation in elephant seals increases circulating glucocorticoids without inducing muscle atrophy, but the cellular mechanisms that allow elephant seals to cope with such conditions remain elusive. We generated a cellular model and conducted transcriptomic, metabolic, and morphological analyses to study how seal cells adapt to sustained glucocorticoid exposure. Seal muscle progenitor cells differentiate into contractile myotubes with a distinctive morphology, gene expression profile, and metabolic phenotype. Exposure to dexamethasone at three ascending concentrations for 48 h modulated the expression of six clusters of genes related to structural constituents of muscle and pathways associated with energy metabolism and cell survival. Knockdown of the glucocorticoid receptor (GR) and downstream expression analyses corroborated that GR mediates the observed effects. Dexamethasone also decreased cellular respiration, shifted the metabolic phenotype toward glycolysis, and induced mitochondrial fission and dissociation of mitochondria-endoplasmic reticulum (ER) interactions without decreasing cell viability. Knockdown of DNA damage-inducible transcript 4 (DDIT4), a GR target involved in the dissociation of mitochondria-ER membranes, recovered respiration and modulated antioxidant gene expression in myotubes treated with dexamethasone. These results show that adaptation to sustained glucocorticoid exposure in elephant seal myotubes involves a metabolic shift toward glycolysis, which is supported by alterations in mitochondrial morphology and a reduction in mitochondria-ER interactions, resulting in decreased respiration without compromising cell survival.
Collapse
Affiliation(s)
| | | | - Jane I Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, California
| | - Daniel E Crocker
- Department of Biology, Sonoma State University, Rohnert Park, California
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, Berkeley, California
| | | |
Collapse
|
12
|
Everman ER, Cloud-Richardson KM, Macdonald SJ. Characterizing the genetic basis of copper toxicity in Drosophila reveals a complex pattern of allelic, regulatory, and behavioral variation. Genetics 2021; 217:1-20. [PMID: 33683361 PMCID: PMC8045719 DOI: 10.1093/genetics/iyaa020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/16/2020] [Indexed: 11/13/2022] Open
Abstract
A range of heavy metals are required for normal cell function and homeostasis. However, the anthropogenic release of metal compounds into soil and water sources presents a pervasive health threat. Copper is one of many heavy metals that negatively impacts diverse organisms at a global scale. Using a combination of quantitative trait locus (QTL) mapping and RNA sequencing in the Drosophila Synthetic Population Resource, we demonstrate that resistance to the toxic effects of ingested copper in D. melanogaster is genetically complex and influenced by allelic and expression variation at multiple loci. QTL mapping identified several QTL that account for a substantial fraction of heritability. Additionally, we find that copper resistance is impacted by variation in behavioral avoidance of copper and may be subject to life-stage specific regulation. Gene expression analysis further demonstrated that resistant and sensitive strains are characterized by unique expression patterns. Several of the candidate genes identified via QTL mapping and RNAseq have known copper-specific functions (e.g., Ccs, Sod3, CG11825), and others are involved in the regulation of other heavy metals (e.g., Catsup, whd). We validated several of these candidate genes with RNAi suggesting they contribute to variation in adult copper resistance. Our study illuminates the interconnected roles that allelic and expression variation, organism life stage, and behavior play in copper resistance, allowing a deeper understanding of the diverse mechanisms through which metal pollution can negatively impact organisms.
Collapse
Affiliation(s)
- Elizabeth R Everman
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | | | - Stuart J Macdonald
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
- Center for Computational Biology, University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
13
|
Alavijeh ES, Khajehali J, Snoeck S, Panteleri R, Ghadamyari M, Jonckheere W, Bajda S, Saalwaechter C, Geibel S, Douris V, Vontas J, Van Leeuwen T, Dermauw W. Molecular and genetic analysis of resistance to METI-I acaricides in Iranian populations of the citrus red mite Panonychus citri. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2020; 164:73-84. [PMID: 32284140 DOI: 10.1016/j.pestbp.2019.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/22/2019] [Accepted: 12/27/2019] [Indexed: 06/11/2023]
Abstract
The citrus red mite, Panonychus citri, is a major pest on citrus all around the world. Mitochondrial Electron Transport Inhibitors of complex I (METI-I) acaricides such as fenpyroximate have been used extensively to control P. citri populations, which resulted in multiple reports of METI-I resistant populations in the field. In this study, biochemical and molecular mechanisms of fenpyroximate resistance were investigated in P. citri. Seven populations were collected from Northern provinces of Iran. Resistance ratios were determined and reached up to 75-fold in comparison to a fenpyroximate susceptible population. Cross-resistance to two additional METI-I acaricides, pyridaben and tebufenpyrad, was detected. PBO synergism experiments, in vivo enzyme assays and gene expression analysis suggest a minor involvement of cytochrome P450 monooxygenases in fenpyroximate resistance, which is in contrast with many reported cases for the closely related Tetranychus urticae. Next, we determined the frequency of a well-known mutation in the target-site of METI-Is, the PSST subunit, associated with METI-I resistance. Indeed, the H92R substitution was detected in a highly fenpyroximate resistant P. citri population. Additionally, a new amino acid substitution at a conserved site in the PSST subunit was detected, A94V, with higher allele frequencies in a moderately resistant population. Marker-assisted back-crossing in a susceptible background confirmed the potential involvement of the newly discovered A94V mutation in fenpyroximate resistance. However, introduction of the A94V mutation in the PSST homologue of D. melanogaster using CRISPR-Cas9 did not result in fenpyroximate resistant flies. In addition, differences in binding curves between METI-Is and complex I measured directly, in isolated transgenic and wildtype mitochondria preparations, could not be found.
Collapse
Affiliation(s)
- Elaheh Shafiei Alavijeh
- Department of Plant Protection, Faculty of Agricultural Sciences, University of Guilan, Rasht, Iran
| | - Jahangir Khajehali
- Department of Plant Protection, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Simon Snoeck
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure links 653, 9000 Ghent, Belgium
| | - Rafaela Panteleri
- Laboratory of Molecular Entomology, Department of Biology, University of Crete, GR-70013 Heraklion, Crete, Greece; Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology (FORTH), Nikolaou Plastira Street 100, 70013 Heraklion, Crete, Greece
| | - Mohammad Ghadamyari
- Department of Plant Protection, Faculty of Agricultural Sciences, University of Guilan, Rasht, Iran
| | - Wim Jonckheere
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure links 653, 9000 Ghent, Belgium
| | - Sabina Bajda
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure links 653, 9000 Ghent, Belgium
| | | | - Sven Geibel
- Bayer AG, CropScience Division, 40789 Monheim, Germany
| | - Vassilis Douris
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology (FORTH), Nikolaou Plastira Street 100, 70013 Heraklion, Crete, Greece; Department of Biological Applications and Technology, University of Ioannina, 451 10 Ioannina, Greece
| | - John Vontas
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology (FORTH), Nikolaou Plastira Street 100, 70013 Heraklion, Crete, Greece; Pesticide Science Laboratory, Department of Crop Science, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - Thomas Van Leeuwen
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure links 653, 9000 Ghent, Belgium.
| | - Wannes Dermauw
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure links 653, 9000 Ghent, Belgium.
| |
Collapse
|
14
|
Chen PH, Wu J, Ding CKC, Lin CC, Pan S, Bossa N, Xu Y, Yang WH, Mathey-Prevot B, Chi JT. Kinome screen of ferroptosis reveals a novel role of ATM in regulating iron metabolism. Cell Death Differ 2020; 27:1008-1022. [PMID: 31320750 PMCID: PMC7206124 DOI: 10.1038/s41418-019-0393-7] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 06/22/2019] [Accepted: 07/01/2019] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis is a specialized iron-dependent cell death that is associated with lethal lipid peroxidation. Modulation of ferroptosis may have therapeutic potential since it has been implicated in various human diseases as well as potential antitumor activities. However, much remains unknown about the underlying mechanisms and genetic determinants of ferroptosis. Given the critical role of kinases in most biological processes and the availability of various kinase inhibitors, we sought to systemically identify kinases essential for ferroptosis. We performed a forward genetic-based kinome screen against ferroptosis in MDA-MB-231 cells triggered by cystine deprivation. This screen identified 34 essential kinases involved in TNFα and NF-kB signaling. Unexpectedly, the DNA damage response serine/threonine kinase ATM (mutated in Ataxia-Telangiectasia) was found to be essential for ferroptosis. The pharmacological or genetic inhibition of ATM consistently rescued multiple cancer cells from ferroptosis triggered by cystine deprivation or erastin. Instead of the canonical DNA damage pathways, ATM inhibition rescued ferroptosis by increasing the expression of iron regulators involved in iron storage (ferritin heavy and light chain, FTH1 and FTL) and export (ferroportin, FPN1). The coordinated changes of these iron regulators during ATM inhibition resulted in a lowering of labile iron and prevented the iron-dependent ferroptosis. Furthermore, we found that ATM inhibition enhanced the nuclear translocation of metal-regulatory transcription factor 1 (MTF1), responsible for regulating expression of Ferritin/FPN1 and ferroptosis protection. Genetic depletion of MTF-1 abolished the regulation of iron-regulatory elements by ATM and resensitized the cells to ferroptosis. Together, we have identified an unexpected ATM-MTF1-Ferritin/FPN1 regulatory axis as novel determinants of ferroptosis through regulating labile iron levels.
Collapse
Affiliation(s)
- Po-Han Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Jianli Wu
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Chien-Kuang Cornelia Ding
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Chao-Chieh Lin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Samuel Pan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Nathan Bossa
- Department of Civil and Environmental Engineering, Duke University, Durham, NC, USA
| | - Yitong Xu
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Wen-Hsuan Yang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Bernard Mathey-Prevot
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA. .,Duke Center for Genomic and Computational Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
15
|
Zeng J, Huynh N, Phelps B, King-Jones K. Snail synchronizes endocycling in a TOR-dependent manner to coordinate entry and escape from endoreplication pausing during the Drosophila critical weight checkpoint. PLoS Biol 2020; 18:e3000609. [PMID: 32097403 PMCID: PMC7041797 DOI: 10.1371/journal.pbio.3000609] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 01/28/2020] [Indexed: 12/30/2022] Open
Abstract
The final body size of any given individual underlies both genetic and environmental constraints. Both mammals and insects use target of rapamycin (TOR) and insulin signaling pathways to coordinate growth with nutrition. In holometabolous insects, the growth period is terminated through a cascade of peptide and steroid hormones that end larval feeding behavior and trigger metamorphosis, a nonfeeding stage during which the larval body plan is remodeled to produce an adult. This irreversible decision, termed the critical weight (CW) checkpoint, ensures that larvae have acquired sufficient nutrients to complete and survive development to adulthood. How insects assess body size via the CW checkpoint is still poorly understood on the molecular level. We show here that the Drosophila transcription factor Snail plays a key role in this process. Before and during the CW checkpoint, snail is highly expressed in the larval prothoracic gland (PG), an endocrine tissue undergoing endoreplication and primarily dedicated to the production of the steroid hormone ecdysone. We observed two Snail peaks in the PG, one before and one after the molt from the second to the third instar. Remarkably, these Snail peaks coincide with two peaks of PG cells entering S phase and a slowing of DNA synthesis between the peaks. Interestingly, the second Snail peak occurs at the exit of the CW checkpoint. Snail levels then decline continuously, and endoreplication becomes nonsynchronized in the PG after the CW checkpoint. This suggests that the synchronization of PG cells into S phase via Snail represents the mechanistic link used to terminate the CW checkpoint. Indeed, PG-specific loss of snail function prior to the CW checkpoint causes larval arrest due to a cessation of endoreplication in PG cells, whereas impairing snail after the CW checkpoint no longer affected endoreplication and further development. During the CW window, starvation or loss of TOR signaling disrupted the formation of Snail peaks and endocycle synchronization, whereas later starvation had no effect on snail expression. Taken together, our data demonstrate that insects use the TOR pathway to assess nutrient status during larval development to regulate Snail in ecdysone-producing cells as an effector protein to coordinate endoreplication and CW attainment. During Drosophila development, the time window when larvae assess their readiness for metamorphosis is marked by slowing of cell growth in the prothoracic gland that produces the molting hormone; cell growth (via DNA endoreplication) then increases, allowing the production of the amount of hormone required to trigger metamorphosis. This study shows that these processes depend on the transcription factor Snail.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Nhan Huynh
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Brian Phelps
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Kirst King-Jones
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
- * E-mail:
| |
Collapse
|
16
|
Halmenschelager PT, da Rocha JBT. Biochemical CuSO 4 Toxicity in Drosophila melanogaster Depends on Sex and Developmental Stage of Exposure. Biol Trace Elem Res 2019; 189:574-585. [PMID: 30178145 DOI: 10.1007/s12011-018-1475-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/07/2018] [Indexed: 01/01/2023]
Abstract
Copper is a transition metal that exists in different chemical forms (e.g., Cu2+,Cu+, and Cu0) and at high concentrations it is toxic. Here, we investigated the Cu2+-induced toxicity in Drosophila melanogaster, evaluating the survival, locomotion, and the activity of acetylcholinesterase (AChE) and glutathione S-transferase (GST) enzymes. Flies were exposed to Cu2+(0.1-1 mmol CuSO4/kg of diet or approximately 0.1-1 mM Cu2+) and allowed to mate during 24 h. GST and AChE enzymes were evaluated in the larvae and in the head and the body (thorax + abdomen) of the adult male and females flies. The total number of adult females (0.4-1 mM) and males (0.75 and 1 mM) was decreased by CuSO4. The climbing ability was hampered in flies exposed to 1 mM Cu2+. In larvae, Cu2+(0.4-1 mM) increased AChE activity (P < 0.002). In males' heads, 0.4 mM Cu2+ increased the AChE activity (P < 0.01). In adults' bodies, Cu2+inhibited the activity in both sexes, but with greater effectiveness in males (0.1 to 1 mM) than in females (1 mM). Regarding GST activity, 0.1 mM Cu2+increased, but 1 mM decrease GST in larvae. In the head of flies, Cu2+decreased the GST activity at intermediate (0.4 mM) and increased GST at the highest concentration (1 mM) in males. In the bodies, the effect of Cu2+was similar. In conclusion, Cu2+exposure in D. melanogaster disrupted locomotion and enzymatic parameters that can be related to changes in AChE and in the detoxifying GST enzyme.
Collapse
Affiliation(s)
- Paula Tais Halmenschelager
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - João Batista Teixeira da Rocha
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil.
| |
Collapse
|
17
|
Miller SJ, Glatzer JC, Hsieh YC, Rothstein JD. Cortical astroglia undergo transcriptomic dysregulation in the G93A SOD1 ALS mouse model. J Neurogenet 2018; 32:322-335. [PMID: 30398075 PMCID: PMC6444185 DOI: 10.1080/01677063.2018.1513508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 08/06/2018] [Indexed: 12/13/2022]
Abstract
Astroglia are the most abundant glia cell in the central nervous system, playing essential roles in maintaining homeostasis. Key functions of astroglia include, but are not limited to, neurotransmitter recycling, ion buffering, immune modulation, neurotrophin secretion, neuronal synaptogenesis and elimination, and blood-brain barrier maintenance. In neurological diseases, it is well appreciated that astroglia play crucial roles in the disease pathogenesis. In amyotrophic lateral sclerosis (ALS), a motor neuron degenerative disease, astroglia in the spinal cord and cortex downregulate essential transporters, among other proteins, that exacerbate disease progression. Spinal cord astroglia undergo dramatic transcriptome dysregulation. However, in the cortex, it has not been well studied what effects glia, especially astroglia, have on upper motor neurons in the pathology of ALS. To begin to shed light on the involvement and dysregulation that astroglia undergo in ALS, we isolated pure grey-matter cortical astroglia and subjected them to microarray analysis. We uncovered a vast number of genes that show dysregulation at end-stage in the ALS mouse model, G93A SOD1. Many of these genes play essential roles in ion homeostasis and the Wnt-signaling pathway. Several of these dysregulated genes are common in ALS spinal cord astroglia, while many of them are unique. This database serves as an approach for understanding the significance of dysfunctional genes and pathways in cortical astroglia in the context of motor neuron disease, as well as determining regional astroglia heterogeneity, and providing insight into ALS pathogenesis.
Collapse
Affiliation(s)
- Sean J. Miller
- Dept. of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD 21205
| | - Jenna C. Glatzer
- Dept. of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD 21205
| | - Yi-chun Hsieh
- Dept. of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD 21205
| | - Jeffrey D. Rothstein
- Dept. of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD 21205
- Dept. of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
| |
Collapse
|
18
|
Andéol Y, Bonneau J, M Gagné L, Jacquet K, Rivest V, Huot MÉ, Séguin C. The phosphoinositide 3-kinase pathway and glycogen synthase kinase-3 positively regulate the activity of metal-responsive transcription factor-1 in response to zinc ions. Biochem Cell Biol 2018; 96:1-8. [PMID: 29707960 DOI: 10.1139/bcb-2018-0073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
Metal-responsive transcription factor-1 (MTF-1) is a metal-regulatory transcription factor essential for induction of the genes encoding metallothioneins (MTs) in response to transition metal ions. Activation of MTF-1 is dependent on the interaction of zinc with the zinc fingers of the protein. In addition, phosphorylation is essential for MTF-1 transactivation. We previously showed that inhibition of phosphoinositide 3-kinase (PI3K) abrogated Mt expression and metal-induced MTF-1 activation in human hepatocellular carcinoma (HCC) HepG2 and mouse L cells, thus showing that the PI3K signaling pathway positively regulates MTF-1 activity and Mt gene expression. However, it has also been reported that inhibition of PI3K has no significant effects on Mt expression in immortalized epithelial cells and increases Mt expression in HCC cells. To further characterize the role of the PI3K pathway on the activity of MTF-1, transfection experiments were performed in HEK293 and HepG2 cells in presence of glycogen synthase kinase-3 (GSK-3), mTOR-C1, and mTOR-C2 inhibitors, as well as of siRNAs targeting Phosphatase and TENsin homolog (PTEN). We showed that inhibition of the mTOR-C2 complex inhibits the activity of MTF-1 in HepG2 and HEK293 cells, while inhibition of the mTOR-C1 complex or of PTEN stimulates MTF-1 activity in HEK293 cells. These results confirm that the PI3K pathway positively regulates MTF-1 activity. Finally, we showed that GSK-3 is required for MTF-1 activation in response to zinc ions.
Collapse
Affiliation(s)
- Yannick Andéol
- a Équipe Enzymologie de l'ARN, ER6, 9 quai St Bernard, Faculté des Sciences et Technologies, Sorbonne-Université, 75252 Paris, Cedex 05, France
| | - Jessica Bonneau
- b Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Université Laval and Centre de recherche du CHU de Québec, Axe Oncologie, Hôtel Dieu de Québec, 9 rue McMahon, Québec, QC G1R 3S3, Canada
| | - Laurence M Gagné
- b Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Université Laval and Centre de recherche du CHU de Québec, Axe Oncologie, Hôtel Dieu de Québec, 9 rue McMahon, Québec, QC G1R 3S3, Canada
| | - Kevin Jacquet
- b Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Université Laval and Centre de recherche du CHU de Québec, Axe Oncologie, Hôtel Dieu de Québec, 9 rue McMahon, Québec, QC G1R 3S3, Canada
| | - Véronique Rivest
- b Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Université Laval and Centre de recherche du CHU de Québec, Axe Oncologie, Hôtel Dieu de Québec, 9 rue McMahon, Québec, QC G1R 3S3, Canada
| | - Marc-Étienne Huot
- b Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Université Laval and Centre de recherche du CHU de Québec, Axe Oncologie, Hôtel Dieu de Québec, 9 rue McMahon, Québec, QC G1R 3S3, Canada
| | - Carl Séguin
- b Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Université Laval and Centre de recherche du CHU de Québec, Axe Oncologie, Hôtel Dieu de Québec, 9 rue McMahon, Québec, QC G1R 3S3, Canada
| |
Collapse
|
19
|
Calap-Quintana P, Navarro JA, González-Fernández J, Martínez-Sebastián MJ, Moltó MD, Llorens JV. Drosophila melanogaster Models of Friedreich's Ataxia. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5065190. [PMID: 29850527 PMCID: PMC5907503 DOI: 10.1155/2018/5065190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/29/2018] [Accepted: 02/28/2018] [Indexed: 11/17/2022]
Abstract
Friedreich's ataxia (FRDA) is a rare inherited recessive disorder affecting the central and peripheral nervous systems and other extraneural organs such as the heart and pancreas. This incapacitating condition usually manifests in childhood or adolescence, exhibits an irreversible progression that confines the patient to a wheelchair, and leads to early death. FRDA is caused by a reduced level of the nuclear-encoded mitochondrial protein frataxin due to an abnormal GAA triplet repeat expansion in the first intron of the human FXN gene. FXN is evolutionarily conserved, with orthologs in essentially all eukaryotes and some prokaryotes, leading to the development of experimental models of this disease in different organisms. These FRDA models have contributed substantially to our current knowledge of frataxin function and the pathogenesis of the disease, as well as to explorations of suitable treatments. Drosophila melanogaster, an organism that is easy to manipulate genetically, has also become important in FRDA research. This review describes the substantial contribution of Drosophila to FRDA research since the characterization of the fly frataxin ortholog more than 15 years ago. Fly models have provided a comprehensive characterization of the defects associated with frataxin deficiency and have revealed genetic modifiers of disease phenotypes. In addition, these models are now being used in the search for potential therapeutic compounds for the treatment of this severe and still incurable disease.
Collapse
Affiliation(s)
- P. Calap-Quintana
- Department of Genetics, University of Valencia, Campus of Burjassot, Valencia, Spain
| | - J. A. Navarro
- Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - J. González-Fernández
- Department of Genetics, University of Valencia, Campus of Burjassot, Valencia, Spain
- Biomedical Research Institute INCLIVA, Valencia, Spain
| | | | - M. D. Moltó
- Department of Genetics, University of Valencia, Campus of Burjassot, Valencia, Spain
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - J. V. Llorens
- Department of Genetics, University of Valencia, Campus of Burjassot, Valencia, Spain
| |
Collapse
|
20
|
Navarro JA, Schneuwly S. Copper and Zinc Homeostasis: Lessons from Drosophila melanogaster. Front Genet 2017; 8:223. [PMID: 29312444 PMCID: PMC5743009 DOI: 10.3389/fgene.2017.00223] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/11/2017] [Indexed: 01/19/2023] Open
Abstract
Maintenance of metal homeostasis is crucial for many different enzymatic activities and in turn for cell function and survival. In addition, cells display detoxification and protective mechanisms against toxic accumulation of metals. Perturbation of any of these processes normally leads to cellular dysfunction and finally to cell death. In the last years, loss of metal regulation has been described as a common pathological feature in many human neurodegenerative diseases. However, in most cases, it is still a matter of debate whether such dyshomeostasis is a primary or a secondary downstream defect. In this review, we will summarize and critically evaluate the contribution of Drosophila to model human diseases that involve altered metabolism of metals or in which metal dyshomeostasis influence their pathobiology. As a prerequisite to use Drosophila as a model, we will recapitulate and describe the main features of core genes involved in copper and zinc metabolism that are conserved between mammals and flies. Drosophila presents some unique strengths to be at the forefront of neurobiological studies. The number of genetic tools, the possibility to easily test genetic interactions in vivo and the feasibility to perform unbiased genetic and pharmacological screens are some of the most prominent advantages of the fruitfly. In this work, we will pay special attention to the most important results reported in fly models to unveil the role of copper and zinc in cellular degeneration and their influence in the development and progression of human neurodegenerative pathologies such as Parkinson's disease, Alzheimer's disease, Huntington's disease, Friedreich's Ataxia or Menkes, and Wilson's diseases. Finally, we show how these studies performed in the fly have allowed to give further insight into the influence of copper and zinc in the molecular and cellular causes and consequences underlying these diseases as well as the discovery of new therapeutic strategies, which had not yet been described in other model systems.
Collapse
Affiliation(s)
- Juan A. Navarro
- Department of Developmental Biology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | | |
Collapse
|
21
|
Smolentseva O, Gusarov I, Gautier L, Shamovsky I, DeFrancesco AS, Losick R, Nudler E. Mechanism of biofilm-mediated stress resistance and lifespan extension in C. elegans. Sci Rep 2017; 7:7137. [PMID: 28769037 PMCID: PMC5540977 DOI: 10.1038/s41598-017-07222-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/26/2017] [Indexed: 01/16/2023] Open
Abstract
Bacteria naturally form communities of cells known as biofilms. However the physiological roles of biofilms produced by non-pathogenic microbiota remain largely unknown. To assess the impact of a biofilm on host physiology we explored the effect of several non-pathogenic biofilm-forming bacteria on Caenorhabditis elegans. We show that biofilm formation by Bacillus subtilis, Lactobacillus rhamnosus and Pseudomonas fluorescens induces C. elegans stress resistance. Biofilm also protects against pathogenic infection and prolongs lifespan. Total mRNA analysis identified a set of host genes that are upregulated in response to biofilm formation by B. subtilis. We further demonstrate that mtl-1 is responsible for the biofilm-mediated increase in oxidative stress resistance and lifespan extension. Induction of mtl-1 and hsp-70 promotes biofilm-mediated thermotolerance. ilys-2 activity accounts for biofilm-mediated resistance to Pseudomonas aeruginosa killing. These results reveal the importance of non-pathogenic biofilms for host physiology and provide a framework to study commensal biofilms in higher organisms.
Collapse
Affiliation(s)
- Olga Smolentseva
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Ivan Gusarov
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Laurent Gautier
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Ilya Shamovsky
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Alicia S DeFrancesco
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, 02138, USA
| | - Richard Losick
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, 02138, USA
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA.
- Howard Hughes Medical Institute, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
22
|
Moskalev A, Shaposhnikov M, Proshkina E, Belyi A, Fedintsev A, Zhikrivetskaya S, Guvatova Z, Sadritdinova A, Snezhkina A, Krasnov G, Kudryavtseva A. The influence of pro-longevity gene Gclc overexpression on the age-dependent changes in Drosophila transcriptome and biological functions. BMC Genomics 2016; 17:1046. [PMID: 28105938 PMCID: PMC5249042 DOI: 10.1186/s12864-016-3356-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Transcriptional changes that contribute to the organism’s longevity and prevent the age-dependent decline of biological functions are not well understood. Here, we overexpressed pro-longevity gene encoding glutamate-cysteine ligase catalytic subunit (Gclc) and analyzed age-dependent changes in transcriptome that associated with the longevity, stress resistance, locomotor activity, circadian rhythmicity, and fertility. Results Here we reproduced the life extension effect of neuronal overexpression of the Gclc gene and investigated its influence on the age-depended dynamics of transcriptome and biological functions such as fecundity, spontaneous locomotor activity and circadian rhythmicity, as well as on the resistance to oxidative, proteotoxic and osmotic stresses. It was shown that Gclc overexpression reduces locomotor activity in the young and middle ages compared to control flies. Gclc overexpression slowed down the age-dependent decline of locomotor activity and circadian rhythmicity, and resistance to stress treatments. Gclc level demonstrated associations with the expression of genes involved in a variety of cellular processes including Jak-STAT, MAPK, FOXO, Notch, mTOR, TGF-beta signaling pathways, translation, protein processing in endoplasmic reticulum, proteasomal degradation, glycolysis, oxidative phosphorylation, apoptosis, regulation of circadian rhythms, differentiation of neurons, synaptic plasticity and transmission. Conclusions Our study revealed that Gclc overexpression induces transcriptional changes associated with the lifespan extension and uncovered pathways that may be associated with the age-dependent decline of biological functions. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3356-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia. .,Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia. .,Moscow Institute of Physics and Technology, Dolgoprudny, Russia. .,Syktyvkar State University, Syktyvkar, Russia.
| | - Mikhail Shaposhnikov
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia.,Syktyvkar State University, Syktyvkar, Russia
| | - Ekaterina Proshkina
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia.,Syktyvkar State University, Syktyvkar, Russia
| | - Alexey Belyi
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia.,Syktyvkar State University, Syktyvkar, Russia
| | | | | | - Zulfiya Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Asiya Sadritdinova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
23
|
Camargo G, Elizalde A, Trujillo X, Montoya-Pérez R, Mendoza-Magaña ML, Hernandez-Chavez A, Hernandez L. Inactivation of GABAA receptor is related to heat shock stress response in organism model Caenorhabditis elegans. Cell Stress Chaperones 2016; 21:763-72. [PMID: 27230213 PMCID: PMC5003793 DOI: 10.1007/s12192-016-0701-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 04/29/2016] [Accepted: 05/11/2016] [Indexed: 01/08/2023] Open
Abstract
The mechanisms underlying oxidative stress (OS) resistance are not completely clear. Caenorhabditis elegans (C. elegans) is a good organism model to study OS because it displays stress responses similar to those in mammals. Among these mechanisms, the insulin/IGF-1 signaling (IIS) pathway is thought to affect GABAergic neurotransmission. The aim of this study was to determine the influence of heat shock stress (HS) on GABAergic activity in C. elegans. For this purpose, we tested the effect of exposure to picrotoxin (PTX), gamma-aminobutyric acid (GABA), hydrogen peroxide, and HS on the occurrence of a shrinking response (SR) after nose touch stimulus in N2 (WT) worms. Moreover, the effect of HS on the expression of UNC-49 (GABAA receptor ortholog) in the EG1653 strain and the effect of GABA and PTX exposure on HSP-16.2 expression in the TJ375 strain were analyzed. PTX 1 mM- or H2O2 0.7 mM-exposed worms displayed a SR in about 80 % of trials. GABA exposure did not cause a SR. HS prompted the occurrence of a SR as did PTX 1 mM or H2O2 0.7 mM exposure. In addition, HS increased UNC-49 expression, and PTX augmented HSP-16.2 expression. Thus, the results of the present study suggest that oxidative stress, through either H2O2 exposure or application of heat shock, inactivates the GABAergic system, which subsequently would affect the oxidative stress response, perhaps by enhancing the activity of transcription factors DAF-16 and HSF-1, both regulated by the IIS pathway and related to hsp-16.2 expression.
Collapse
Affiliation(s)
- Gabriela Camargo
- Laboratorio de Neurofisiología, Departamento de Fisiología, Centro Universitario de Ciencias dela Salud, Universidad de Guadalajara, Sierra Mojada # 950, Guadalajara, 44340, Jalisco, Mexico
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima Av. 25 de Julio # 965, Colima, 28045, Colima, Mexico
- Laboratorio de Biotecnología, Departamento de Botánica y Zoología, Centro Universitariode Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez # 2100, Zapopan, 45110, Jalisco, Mexico
| | - Alejandro Elizalde
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima Av. 25 de Julio # 965, Colima, 28045, Colima, Mexico
| | - Xochitl Trujillo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima Av. 25 de Julio # 965, Colima, 28045, Colima, Mexico
| | - Rocío Montoya-Pérez
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Francisco J. Mújica S/N, Morelia, 58030, Michoacán, Mexico
| | - María Luisa Mendoza-Magaña
- Laboratorio de Neurofisiología, Departamento de Fisiología, Centro Universitario de Ciencias dela Salud, Universidad de Guadalajara, Sierra Mojada # 950, Guadalajara, 44340, Jalisco, Mexico
| | - Abel Hernandez-Chavez
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, SierraMojada # 950, Guadalajara, 44340, Jalisco, Mexico
| | - Leonardo Hernandez
- Laboratorio de Neurofisiología, Departamento de Fisiología, Centro Universitario de Ciencias dela Salud, Universidad de Guadalajara, Sierra Mojada # 950, Guadalajara, 44340, Jalisco, Mexico.
| |
Collapse
|
24
|
Soriano S, Calap-Quintana P, Llorens JV, Al-Ramahi I, Gutiérrez L, Martínez-Sebastián MJ, Botas J, Moltó MD. Metal Homeostasis Regulators Suppress FRDA Phenotypes in a Drosophila Model of the Disease. PLoS One 2016; 11:e0159209. [PMID: 27433942 PMCID: PMC4951068 DOI: 10.1371/journal.pone.0159209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/28/2016] [Indexed: 11/19/2022] Open
Abstract
Friedreich's ataxia (FRDA), the most commonly inherited ataxia in populations of European origin, is a neurodegenerative disorder caused by a decrease in frataxin levels. One of the hallmarks of the disease is the accumulation of iron in several tissues including the brain, and frataxin has been proposed to play a key role in iron homeostasis. We found that the levels of zinc, copper, manganese and aluminum were also increased in a Drosophila model of FRDA, and that copper and zinc chelation improve their impaired motor performance. By means of a candidate genetic screen, we identified that genes implicated in iron, zinc and copper transport and metal detoxification can restore frataxin deficiency-induced phenotypes. Taken together, these results demonstrate that the metal dysregulation in FRDA includes other metals besides iron, therefore providing a new set of potential therapeutic targets.
Collapse
Affiliation(s)
- Sirena Soriano
- Department of Genetics, University of Valencia, Burjassot, Valencia, Spain
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | | | | | - Ismael Al-Ramahi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lucía Gutiérrez
- Department of Biomaterials and Bioinspired Materials, Instituto de Ciencia de Materiales de Madrid/CSIC, Madrid, Spain
| | | | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - María Dolores Moltó
- Department of Genetics, University of Valencia, Burjassot, Valencia, Spain
- CIBERSAM, INCLIVA, Valencia, Spain
| |
Collapse
|
25
|
Lee D, Hwang W, Artan M, Jeong DE, Lee SJ. Effects of nutritional components on aging. Aging Cell 2015; 14:8-16. [PMID: 25339542 PMCID: PMC4326908 DOI: 10.1111/acel.12277] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2014] [Indexed: 12/11/2022] Open
Abstract
Nutrients including carbohydrates, proteins, lipids, vitamins, and minerals regulate various physiological processes and are essential for the survival of organisms. Reduced overall caloric intake delays aging in various organisms. However, the role of each nutritional component in the regulation of lifespan is not well established. In this review, we describe recent studies focused on the regulatory role of each type of nutrient in aging. Moreover, we will discuss how the amount or composition of each nutritional component may influence longevity or health in humans.
Collapse
Affiliation(s)
- Dongyeop Lee
- Department of Life Sciences; Pohang University of Science and Technology; Pohang Gyeongbuk South Korea
| | - Wooseon Hwang
- Department of Life Sciences; Pohang University of Science and Technology; Pohang Gyeongbuk South Korea
| | - Murat Artan
- Information Technology Convergence Engineering; Pohang University of Science and Technology; Pohang Gyeongbuk South Korea
| | - Dae-Eun Jeong
- Department of Life Sciences; Pohang University of Science and Technology; Pohang Gyeongbuk South Korea
| | - Seung-Jae Lee
- Department of Life Sciences; Pohang University of Science and Technology; Pohang Gyeongbuk South Korea
- Information Technology Convergence Engineering; Pohang University of Science and Technology; Pohang Gyeongbuk South Korea
- School of Interdisciplinary Bioscience and Bioengineering; Pohang University of Science and Technology; Pohang Gyeongbuk 790-784 South Korea
| |
Collapse
|
26
|
Montgomery SL, Vorojeikina D, Huang W, Mackay TFC, Anholt RRH, Rand MD. Genome-wide association analysis of tolerance to methylmercury toxicity in Drosophila implicates myogenic and neuromuscular developmental pathways. PLoS One 2014; 9:e110375. [PMID: 25360876 PMCID: PMC4215868 DOI: 10.1371/journal.pone.0110375] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/11/2014] [Indexed: 11/30/2022] Open
Abstract
Methylmercury (MeHg) is a persistent environmental toxin present in seafood that can compromise the developing nervous system in humans. The effects of MeHg toxicity varies among individuals, despite similar levels of exposure, indicating that genetic differences contribute to MeHg susceptibility. To examine how genetic variation impacts MeHg tolerance, we assessed developmental tolerance to MeHg using the sequenced, inbred lines of the Drosophila melanogaster Genetic Reference Panel (DGRP). We found significant genetic variation in the effects of MeHg on development, measured by eclosion rate, giving a broad sense heritability of 0.86. To investigate the influence of dietary factors, we measured MeHg toxicity with caffeine supplementation in the DGRP lines. We found that caffeine counteracts the deleterious effects of MeHg in the majority of lines, and there is significant genetic variance in the magnitude of this effect, with a broad sense heritability of 0.80. We performed genome-wide association (GWA) analysis for both traits, and identified candidate genes that fall into several gene ontology categories, with enrichment for genes involved in muscle and neuromuscular development. Overexpression of glutamate-cysteine ligase, a MeHg protective enzyme, in a muscle-specific manner leads to a robust rescue of eclosion of flies reared on MeHg food. Conversely, mutations in kirre, a pivotal myogenic gene identified in our GWA analyses, modulate tolerance to MeHg during development in accordance with kirre expression levels. Finally, we observe disruptions of indirect flight muscle morphogenesis in MeHg-exposed pupae. Since the pathways for muscle development are evolutionarily conserved, it is likely that the effects of MeHg observed in Drosophila can be generalized across phyla, implicating muscle as an additional hitherto unrecognized target for MeHg toxicity. Furthermore, our observations that caffeine can ameliorate the toxic effects of MeHg show that nutritional factors and dietary manipulations may offer protection against the deleterious effects of MeHg exposure.
Collapse
Affiliation(s)
- Sara L. Montgomery
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Wen Huang
- Department of Biological Sciences, Genetics Program, and W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Trudy F. C. Mackay
- Department of Biological Sciences, Genetics Program, and W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Robert R. H. Anholt
- Department of Biological Sciences, Genetics Program, and W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Matthew D. Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
27
|
Afshar N, Argunhan B, Bettedi L, Szular J, Missirlis F. A recessive X-linked mutation causes a threefold reduction of total body zinc accumulation in Drosophila melanogaster laboratory strains. FEBS Open Bio 2013; 3:302-4. [PMID: 23951551 PMCID: PMC3741916 DOI: 10.1016/j.fob.2013.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 11/25/2022] Open
Abstract
A newly identified human locus on chromosome 15 was recently associated with zinc accumulation. Based on a prior report of a threefold difference in zinc accumulation between fumble1 heterozygous mutants and control fly strains, it was suggested that phosphopantothenoylcysteine decarboxylase might affect zinc status through its effects on vitamin B5 (pantothenate) metabolism. We report here that outcrossed fumble1 heterozygous mutant flies with low zinc content have been recovered, suggesting that pantothenate metabolism did not alter zinc homeostasis in fumble1 heterozygous flies. We show instead that the Drosophila condition of low body zinc accumulation is an X-chromosome-linked recessive trait. Flies with a threefold reduction in zinc accumulation remain viable and fertile. There is no causal association between zinc accumulation and fly pantothenate kinase mutants. A widespread X-linked mutation causes a threefold reduction in zinc accumulation in Drosophila.
Collapse
Affiliation(s)
- Negar Afshar
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | | | | | | | | |
Collapse
|
28
|
Huyck RW, Keightley A, Laity JH. Expression and purification of full length mouse metal response element binding transcription factor-1 using Pichia pastoris. Protein Expr Purif 2012; 85:86-93. [PMID: 22780964 DOI: 10.1016/j.pep.2012.06.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 05/31/2012] [Accepted: 06/28/2012] [Indexed: 12/01/2022]
Abstract
The metal response element binding transcription factor-1 (MTF-1) is an important stress response, heavy metal detoxification, and zinc homeostasis factor in eukaryotic organisms from Drosophila to humans. MTF-1 transcriptional regulation is primarily mediated by elevated levels of labile zinc, which direct MTF-1 to bind the metal response element (MRE). This process involves direct zinc binding to the MTF-1 zinc fingers, and zinc dependent interaction of the MTF-1 acidic region with the p300 coactivator protein. Here, the first recombinant expression system for mutant and wild type (WT) mouse MTF-1 (mMTF-1) suitable for biochemical and biophysical studies in vitro is reported. Using the methyltropic yeast Pichia pastoris, nearly half-milligram recombinant WT and mutant mMTF-1 were produced per liter of P. pastoris cell culture, and purified by a FLAG-tag epitope. Using a first pass ammonium sulfate purification, followed by anti-FLAG affinity resin, mMTF-1 was purified to >95% purity. This recombinant mMTF-1 was then assayed for direct protein-protein interactions with p300 by co-immunoprecipitation. Surface plasmon resonance studies on mMTF-1 provided the first quantitative DNA binding affinity measurements to the MRE promotor element (K(d)=5±3 nM). Both assays demonstrated the functional activity of the recombinant mMTF-1, while elucidating the molecular basis for mMTF-1-p300 functional synergy, and provided new insights into the mMTF-1 domain specific roles in DNA binding. Overall, this production system provides accessibility for the first time to a multitude of in vitro studies using recombinant mutant and WT mMTF-1, which greatly facilitates new approaches to understanding the complex and varied functions of this protein.
Collapse
Affiliation(s)
- Ryan W Huyck
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110-2499, United States.
| | | | | |
Collapse
|
29
|
Günther V, Lindert U, Schaffner W. The taste of heavy metals: gene regulation by MTF-1. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1823:1416-25. [PMID: 22289350 DOI: 10.1016/j.bbamcr.2012.01.005] [Citation(s) in RCA: 238] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/08/2012] [Accepted: 01/11/2012] [Indexed: 11/22/2022]
Abstract
The metal-responsive transcription factor-1 (MTF-1, also termed MRE-binding transcription factor-1 or metal regulatory transcription factor-1) is a pluripotent transcriptional regulator involved in cellular adaptation to various stress conditions, primarily exposure to heavy metals but also to hypoxia or oxidative stress. MTF-1 is evolutionarily conserved from insects to humans and is the main activator of metallothionein genes, which encode small cysteine-rich proteins that can scavenge toxic heavy metals and free radicals. MTF-1 has been suggested to act as an intracellular metal sensor but evidence for direct metal sensing was scarce. Here we review recent advances in our understanding of MTF-1 regulation with a focus on the mechanism underlying heavy metal responsiveness and transcriptional activation mediated by mammalian or Drosophila MTF-1. This article is part of a Special Issue entitled: Cell Biology of Metals.
Collapse
Affiliation(s)
- Viola Günther
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | | | |
Collapse
|
30
|
N-acetyl cysteine suppresses the foam cell formation that is induced by oxidized low density lipoprotein via regulation of gene expression. Mol Biol Rep 2012; 39:3001-7. [PMID: 21681422 DOI: 10.1007/s11033-011-1062-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 06/08/2011] [Indexed: 12/14/2022]
Abstract
Foam cells derived from macrophages have been implicated as markers of early stage atherosclerosis development. In this study, we found that N-acetyl cysteine (NAC), a well-known inhibitor of reactive oxygen species (ROS), decreased the generation of ROS and suppressed foam cell formation in the presence of oxidized low density lipoprotein through down-regulation of cluster of differentiation 36 expression. We investigated gene expression profiles in order to determine the effects of NAC on foam cell formation using a microarray analysis. The level of apolipoprotein E, which is involved in lipid efflux, was increased and the levels of the antioxidant genes glutathione peroxidase 1 and 3 were also increased. The expression levels of the oxidative stress response and the DNA repair genes were decreased. These results were confirmed using quantitative real-time PCR. Our results indicate that oxidative stress plays an important role in foam cell formation, and that regulation of oxidation using antioxidants is a potential therapeutic method for blocking atherosclerosis development.
Collapse
|
31
|
Apolipoprotein E genotype affects tissue metallothionein levels: studies in targeted gene replacement mice. GENES AND NUTRITION 2012; 7:247-55. [PMID: 22328270 DOI: 10.1007/s12263-012-0282-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 01/24/2012] [Indexed: 12/14/2022]
Abstract
The apolipoprotein E (APOE) genotype is an important risk factor for ageing and age-related diseases. The APOE4 genotype (in contrast to APOE3) has been shown to be associated with oxidative stress and chronic inflammation. Metallothioneins (MT) exhibit antioxidant and anti-inflammatory activity, and MT overexpression has been shown to increase lifespan in mice. Interactions between APOE and MT, however, are largely unknown. Hence, we determined the effect of the APOE4 versus APOE3 genotype on MT levels in targeted gene replacement mice. APOE4 versus APOE3 mice exhibited significantly lower hepatic MT1 and MT2 mRNA as well as lower MT protein levels. The decrease in hepatic MT protein levels in APOE4 as compared to APOE3 mice was accompanied by lower nuclear Nrf1, a protein partly controlling MT gene expression. Cell culture experiments using hepatocytes identified allyl-isothiocyanate (AITC) as a potent MT inductor in vitro. Therefore, we supplemented APOE3 and APOE4 mice with AITC. However, AITC (15 mg/kg b.w.) could only partly correct for decreased MT1 and MT2 gene expression in APOE4 mice in vivo. Furthermore, cholesterol significantly decreased both Nrf1 and MT mRNA levels in Huh7 cells indicating that differences in MT gene expression between the two genotypes could be related to differences in hepatic cholesterol concentrations. Overall, present data suggest that the APOE genotype is an important determinant of tissue MT levels in mice and that MT gene expression may be impaired by the APOE4 genotype.
Collapse
|
32
|
Saini N, Georgiev O, Schaffner W. The parkin mutant phenotype in the fly is largely rescued by metal-responsive transcription factor (MTF-1). Mol Cell Biol 2011; 31:2151-61. [PMID: 21383066 PMCID: PMC3133352 DOI: 10.1128/mcb.05207-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Accepted: 02/21/2011] [Indexed: 11/20/2022] Open
Abstract
The gene for Parkin, an E3 ubiquitin ligase, is mutated in some familial forms of Parkinson's disease, a severe neurodegenerative disorder. A homozygous mutant of the Drosophila ortholog of human parkin is viable but results in severe motoric impairment including an inability to fly, female and male sterility, and a decreased life span. We show here that a double mutant of the genes for Parkin and the metal-responsive transcription factor 1 (MTF-1) is not viable. MTF-1, which is conserved from insects to mammals, is a key regulator of heavy metal homeostasis and detoxification and plays additional roles in other stress conditions, notably oxidative stress. In contrast to the synthetic lethality of the double mutant, elevated expression of MTF-1 dramatically ameliorates the parkin mutant phenotype, as evidenced by a prolonged life span, motoric improvement including short flight episodes, and female fertility. At the cellular level, muscle and mitochondrial structures are substantially improved. A beneficial effect is also seen with a transgene encoding human MTF-1. We propose that Parkin and MTF-1 provide complementary functions in metal homeostasis, oxidative stress and other cellular stress responses. Our findings also raise the possibility that MTF-1 gene polymorphisms in humans could affect the severity of Parkinson's disease.
Collapse
Affiliation(s)
- Nidhi Saini
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, Zürich CH-8051, Switzerland
| | - Oleg Georgiev
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, Zürich CH-8051, Switzerland
| | - Walter Schaffner
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, Zürich CH-8051, Switzerland
| |
Collapse
|
33
|
Mocchegiani E, Costarelli L, Giacconi R, Piacenza F, Basso A, Malavolta M. Zinc, metallothioneins and immunosenescence: effect of zinc supply as nutrigenomic approach. Biogerontology 2011; 12:455-65. [PMID: 21503725 DOI: 10.1007/s10522-011-9337-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 04/10/2011] [Indexed: 01/09/2023]
Abstract
Ageing is an inevitable biological process associated with gradual and spontaneous biochemical and physiological changes and increased susceptibility to diseases. Nutritional factor, zinc, known to be involved in improving immunity, may remodel some of the age-associated changes, leading to a healthy ageing. "In Vitro" studies involving human lymphocytes exposed to endotoxins, and "in vivo" studies comparing old and young mice fed with low dietary zinc suggest that zinc is important for both innate and adaptive immune efficiency, and more optimal inflammatory/immune response. The intracellular zinc homeostasis is mainly regulated by Metallothioneins (MT), via ion release through the reduction of thiol groups in MT molecule. These processes are crucial because mediating the zinc signalling within the immune cells assigning to zinc a role of "second messenger". Zinc homeostasis is altered in ageing partly due to higher expression levels of MT, leading to an increased sequestration of zinc, resulting in less availability of free intracellular zinc. Improvement of immune functions and stress response systems occurs in elderly after physiological zinc supplementation. The main reason behind these effects seems to be related to a like "hormetic" response induced by zinc. However, the choice of old subjects for zinc supplementation has to be performed in relationship to the specific genetic background of MT and pro-inflammatory cytokine (IL-6) because the latter is involved both in MT gene expression and in intracellular zinc homeostasis. Old subjects carrying GG genotypes (termed C- carriers) in IL-6--174G/C locus display increased IL-6 production, low intracellular zinc ion availability, impaired innate immune response and enhanced MT. By contrast, old subjects carrying GC and CC genotypes (termed C+ carriers) in the same IL-6--174 locus displayed satisfactory intracellular zinc and innate immune response. Moreover, male carriers of C+ allele are more prone to reach centenarian age than C- ones. Therefore, old C- subjects are likely to benefit more from zinc supplementation restoring NK cell cytotoxicity and improving the zinc status. Plasma zinc deficiency and the altered immune response is more evident when the genetic variations of IL-6 polymorphism are associated with the genetic variations of MT1A in position +647, suggesting that the genetic variations of IL-6 and MT1A are very useful tools for the identification of old people who effectively need zinc supplementation.
Collapse
Affiliation(s)
- Eugenio Mocchegiani
- Nutrition and Ageing Centre, Italian National Research Centres on Ageing (INRCA), Via Birarelli 8, 60121, Ancona, Italy.
| | | | | | | | | | | |
Collapse
|
34
|
McHugh PC, Wright JA, Brown DR. Transcriptional regulation of the beta-synuclein 5'-promoter metal response element by metal transcription factor-1. PLoS One 2011; 6:e17354. [PMID: 21386983 PMCID: PMC3046239 DOI: 10.1371/journal.pone.0017354] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 01/29/2011] [Indexed: 12/24/2022] Open
Abstract
The progression of many human neurodegenerative disorders is associated with an accumulation of alpha-synuclein. Alpha-synuclein belongs to the homologous synuclein family, which includes beta-synuclein. It has been proposed that beta-synuclein may be a natural regulator of alpha-synuclein. Therefore controlling beta-synuclein expression may control the accumulation of alpha-synuclein and ultimately prevent disease progression. The regulation of synucleins is poorly understood. We investigated the transcriptional regulation of beta-synuclein, with the aim of identifying molecules that differentially control beta-synuclein expression levels. To investigate transcriptional regulation of beta-synuclein, we used reporter gene assays and bioinformatics. We identified a region -1.1/-0.6 kb upstream of the beta-synuclein translational start site to be a key regulatory region of beta-synuclein 5'-promoter activity in human dopaminergic cells (SH-SY5Y). Within this key promoter region we identified a metal response element pertaining to a putative Metal Transcription Factor-1 (MTF-1) binding site. We demonstrated that MTF-1 binds to this 5'-promoter region using EMSA analysis. Moreover, we showed that MTF-1 differentially regulates beta-synuclein promoter binding site, as well as beta-synuclein mRNA and protein expression. This effect of MTF-1 on expression was found to be specific to beta-synuclein when compared to alpha-synuclein. Understanding the regulation of synucleins and how they interact may point to molecular targets that could be manipulated for therapeutic benefit. In this study we showed that MTF-1 differentially controls the expression of beta-synuclein when compared to its homolog alpha-synuclein. This could potentially provide a novel targets or pathways for therapeutic intervention and/or treatment of synucleinopathies.
Collapse
Affiliation(s)
- Patrick C. McHugh
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | - Josephine A. Wright
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| |
Collapse
|
35
|
Swindell WR. Metallothionein and the biology of aging. Ageing Res Rev 2011; 10:132-45. [PMID: 20933613 DOI: 10.1016/j.arr.2010.09.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 09/24/2010] [Accepted: 09/24/2010] [Indexed: 12/22/2022]
Abstract
Metallothionein (MT) is a low molecular weight protein with anti-apoptotic properties that has been demonstrated to scavenge free radicals in vitro. MT has not been extensively investigated within the context of aging biology. The purpose of this review, therefore, is to discuss findings on MT that are relevant to basic aging mechanisms and to draw attention to the possible role of MT in pro-longevity interventions. MT is one of just a handful of proteins that, when overexpressed, has been demonstrated to increase mouse lifespan. MT also protects against development of obesity in mice provided a high fat diet as well as diet-induced oxidative stress damage. Abundance of MT is responsive to caloric restriction (CR) and inhibition of the insulin/insulin-like signaling (IIS) pathway, and elevated MT gene expression has been observed in tissues from fasted and CR-fed mice, long-lived dwarf mice, worms maintained under CR conditions, and long-lived daf-2 mutant worms. The dysregulation of MT in these systems is likely to have tissue-specific effects on aging outcomes. Further investigation will therefore be needed to understand how MT contributes to the response of invertebrates and mice to CR and the endocrine mutations studied by aging researchers.
Collapse
Affiliation(s)
- William R Swindell
- Department of Genetics, Harvard Medical School New Research Building, Room 0464, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|