201
|
Lonergan ZR, Skaar EP. Nutrient Zinc at the Host-Pathogen Interface. Trends Biochem Sci 2019; 44:1041-1056. [PMID: 31326221 PMCID: PMC6864270 DOI: 10.1016/j.tibs.2019.06.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/13/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022]
Abstract
Zinc is an essential cofactor required for life and, as such, mechanisms exist for its homeostatic maintenance in biological systems. Despite the evolutionary distance between vertebrates and microbial life, there are parallel mechanisms to balance the essentiality of zinc with its inherent toxicity. Vertebrates regulate zinc homeostasis through a complex network of metal transporters and buffering systems that respond to changes in nutritional zinc availability or inflammation. Fine-tuning of this network becomes crucial during infections, where host nutritional immunity attempts to limit zinc availability to pathogens. However, accumulating evidence demonstrates that pathogens have evolved mechanisms to subvert host-mediated zinc withholding, and these metal homeostasis systems are important for survival within the host. We discuss here the mechanisms of vertebrate and bacterial zinc homeostasis and mobilization, as well as recent developments in our understanding of microbial zinc acquisition.
Collapse
Affiliation(s)
- Zachery R Lonergan
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Microbe-Host Interactions Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
202
|
Metallothionein induction attenuates the progression of lung injury in mice exposed to long-term intermittent hypoxia. Inflamm Res 2019; 69:15-26. [DOI: 10.1007/s00011-019-01287-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 09/19/2019] [Accepted: 09/24/2019] [Indexed: 01/09/2023] Open
|
203
|
Walther B, Lett AM, Bordoni A, Tomás‐Cobos L, Nieto JA, Dupont D, Danesi F, Shahar DR, Echaniz A, Re R, Fernandez AS, Deglaire A, Gille D, Schmid A, Vergères G. GutSelf: Interindividual Variability in the Processing of Dietary Compounds by the Human Gastrointestinal Tract. Mol Nutr Food Res 2019; 63:e1900677. [PMID: 31483113 PMCID: PMC6900003 DOI: 10.1002/mnfr.201900677] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/25/2019] [Indexed: 12/19/2022]
Abstract
Nutritional research is currently entering the field of personalized nutrition, to a large extent driven by major technological breakthroughs in analytical sciences and biocomputing. An efficient launching of the personalized approach depends on the ability of researchers to comprehensively monitor and characterize interindividual variability in the activity of the human gastrointestinal tract. This information is currently not available in such a form. This review therefore aims at identifying and discussing published data, providing evidence on interindividual variability in the processing of the major nutrients, i.e., protein, fat, carbohydrates, vitamins, and minerals, along the gastrointestinal tract, including oral processing, intestinal digestion, and absorption. Although interindividual variability is not a primary endpoint of most studies identified, a significant number of publications provides a wealth of information on this topic for each category of nutrients. This knowledge remains fragmented, however, and understanding the clinical relevance of most of the interindividual responses to food ingestion described in this review remains unclear. In that regard, this review has identified a gap and sets the base for future research addressing the issue of the interindividual variability in the response of the human organism to the ingestion of foods.
Collapse
Affiliation(s)
- Barbara Walther
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| | - Aaron M. Lett
- Section for Nutrition ResearchDepartment of MedicineImperial College LondonLondonUK
| | - Alessandra Bordoni
- Department of Agri‐Food Sciences and TechnologiesUniversity of Bologna47521CesenaItaly
| | | | | | - Didier Dupont
- UMR 1253Science et Technologie du Lait et de l'ŒufINRA35000RennesFrance
| | - Francesca Danesi
- Department of Agri‐Food Sciences and TechnologiesUniversity of Bologna47521CesenaItaly
| | - Danit R. Shahar
- Department of Public HealthThe S. Daniel Abraham International Center for Health and NutritionBen‐Gurion University of the Negev84105Beer‐ShevaIsrael
| | - Ana Echaniz
- Cambridge Food Science LtdCB23 5ABCambridgeUK
| | - Roberta Re
- Cambridge Food Science LtdCB23 5ABCambridgeUK
| | | | - Amélie Deglaire
- UMR 1253Science et Technologie du Lait et de l'ŒufINRA35000RennesFrance
| | - Doreen Gille
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| | - Alexandra Schmid
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| | - Guy Vergères
- AgroscopeFederal Department of Economic AffairsEducation and Research EAER3003BerneSwitzerland
| |
Collapse
|
204
|
Cadmium-dependent expression of a new metallothionein identified in Trichomonas vaginalis. Biometals 2019; 32:887-899. [DOI: 10.1007/s10534-019-00220-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/28/2019] [Indexed: 10/25/2022]
|
205
|
Ohashi W, Hara T, Takagishi T, Hase K, Fukada T. Maintenance of Intestinal Epithelial Homeostasis by Zinc Transporters. Dig Dis Sci 2019; 64:2404-2415. [PMID: 30830525 DOI: 10.1007/s10620-019-05561-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
Zinc is an essential micronutrient for normal organ function, and dysregulation of zinc metabolism has been implicated in a wide range of diseases. Emerging evidence has revealed that zinc transporters play diverse roles in cellular homeostasis and function by regulating zinc trafficking via organelles or the plasma membrane. In the gastrointestinal tract, zinc deficiency leads to diarrhea and dysfunction of intestinal epithelial cells. Studies also showed that zinc transporters are very important in intestinal epithelial homeostasis. In this review, we describe the physiological roles of zinc transporters in intestinal epithelial functions and relevance of zinc transporters in gastrointestinal diseases.
Collapse
Affiliation(s)
- Wakana Ohashi
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takafumi Hara
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamabouji, Yamashiro, Tokushima, 770-8055, Japan
| | - Teruhisa Takagishi
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamabouji, Yamashiro, Tokushima, 770-8055, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Toshiyuki Fukada
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamabouji, Yamashiro, Tokushima, 770-8055, Japan.
- Division of Pathology, Department of Oral Diagnostic Sciences, School of dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
- RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0042, Japan.
| |
Collapse
|
206
|
Mezzaroba L, Alfieri DF, Colado Simão AN, Vissoci Reiche EM. The role of zinc, copper, manganese and iron in neurodegenerative diseases. Neurotoxicology 2019; 74:230-241. [PMID: 31377220 DOI: 10.1016/j.neuro.2019.07.007] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 01/16/2023]
Abstract
Metals are involved in different pathophysiological mechanisms associated with neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD) and multiple sclerosis (MS). The aim of this study was to review the effects of the essential metals zinc (Zn), copper (Cu), manganese (Mn) and iron (Fe) on the central nervous system (CNS), as well as the mechanisms involved in their neurotoxicity. Low levels of Zn as well as high levels of Cu, Mn, and Fe participate in the activation of signaling pathways of the inflammatory, oxidative and nitrosative stress (IO&NS) response, including nuclear factor kappa B and activator protein-1. The imbalance of these metals impairs the structural, regulatory, and catalytic functions of different enzymes, proteins, receptors, and transporters. Neurodegeneration occurs via association of metals with proteins and subsequent induction of aggregate formation creating a vicious cycle by disrupting mitochondrial function, which depletes adenosine triphosphate and induces IO&NS, cell death by apoptotic and/or necrotic mechanisms. In AD, at low levels, Zn suppresses β-amyloid-induced neurotoxicity by selectively precipitating aggregation intermediates; however, at high levels, the binding of Zn to β-amyloid may enhance formation of fibrillar β-amyloid aggregation, leading to neurodegeneration. High levels of Cu, Mn and Fe participate in the formation α-synuclein aggregates in intracellular inclusions, called Lewy Body, that result in synaptic dysfunction and interruption of axonal transport. In PD, there is focal accumulation of Fe in the substantia nigra, while in AD a diffuse accumulation of Fe occurs in various regions, such as cortex and hippocampus, with Fe marginally increased in the senile plaques. Zn deficiency induces an imbalance between T helper (Th)1 and Th2 cell functions and a failure of Th17 down-regulation, contributing to the pathogenesis of MS. In MS, elevated levels of Fe occur in certain brain regions, such as thalamus and striatum, which may be due to inflammatory processes disrupting the blood-brain barrier and attracting Fe-rich macrophages. Delineating the specific mechanisms by which metals alter redox homeostasis is essential to understand the pathophysiology of AD, PD, and MS and may provide possible new targets for their prevention and treatment of the patients affected by these NDDs.
Collapse
Affiliation(s)
- Leda Mezzaroba
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil; Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil
| | - Daniela Frizon Alfieri
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil
| | - Andrea Name Colado Simão
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil; Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil
| | - Edna Maria Vissoci Reiche
- Laboratory of Applied Immunology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil; Department of Pathology, Clinical Analysis and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Zip Code 86.038-440 Brazil.
| |
Collapse
|
207
|
The zinc transporter Zip14 (SLC39a14) affects Beta-cell Function: Proteomics, Gene expression, and Insulin secretion studies in INS-1E cells. Sci Rep 2019; 9:8589. [PMID: 31197210 PMCID: PMC6565745 DOI: 10.1038/s41598-019-44954-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/24/2019] [Indexed: 12/18/2022] Open
Abstract
Insulin secretion from pancreatic beta-cells is dependent on zinc ions as essential components of insulin crystals, zinc transporters are thus involved in the insulin secretory process. Zip14 (SLC39a14) is a zinc importing protein that has an important role in glucose homeostasis. Zip14 knockout mice display hyperinsulinemia and impaired insulin secretion in high glucose conditions. Endocrine roles for Zip14 have been established in adipocytes and hepatocytes, but not yet confirmed in beta-cells. In this study, we investigated the role of Zip14 in the INS-1E beta-cell line. Zip14 mRNA was upregulated during high glucose stimulation and Zip14 silencing led to increased intracellular insulin content. Large-scale proteomics showed that Zip14 silencing down-regulated ribosomal mitochondrial proteins, many metal-binding proteins, and others involved in oxidative phosphorylation and insulin secretion. Furthermore, proliferation marker Mki67 was down-regulated in Zip14 siRNA-treated cells. In conclusion, Zip14 gene expression is glucose sensitive and silencing of Zip14 directly affects insulin processing in INS-1E beta-cells. A link between Zip14 and ribosomal mitochondrial proteins suggests altered mitochondrial RNA translation, which could disturb mitochondrial function and thereby insulin secretion. This highlights a role for Zip14 in beta-cell functioning and suggests Zip14 as a future pharmacological target in the treatment of beta-cell dysfunction.
Collapse
|
208
|
Elitt CM, Fahrni CJ, Rosenberg PA. Zinc homeostasis and zinc signaling in white matter development and injury. Neurosci Lett 2019; 707:134247. [PMID: 31059767 DOI: 10.1016/j.neulet.2019.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 02/08/2023]
Abstract
Zinc is an essential dietary micronutrient that is abundant in the brain with diverse roles in development, injury, and neurological diseases. With new imaging tools and chelators selectively targeting zinc, the field of zinc biology is rapidly expanding. The importance of zinc homeostasis is now well recognized in neurodegeneration, but there is emerging data that zinc may be equally important in white matter disorders. This review provides an overview of zinc biology, including a discussion of clinical disorders of zinc deficiency, different zinc pools, zinc biomarkers, and methods for measuring zinc. It emphasizes our limited understanding of how zinc is regulated in oligodendrocytes and white matter. Gaps in knowledge about zinc transporters and zinc signaling are discussed. Zinc-induced oligodendrocyte injury pathways relevant to white matter stroke, multiple sclerosis, and white matter injury of prematurity are reviewed and examples of zinc-dependent proteins relevant to myelination highlighted. Finally, a novel ratiometric zinc sensor is reviewed, revealing new information about mobile zinc during oligodendrocyte differentiation. With a better understanding of zinc biology in oligodendrocytes, new therapeutic targets for white matter disorders may be possible and the necessary tools to appropriately study zinc are finally available.
Collapse
Affiliation(s)
- Christopher M Elitt
- Boston Children's Hospital, Department of Neurology and the F.M. Kirby Neurobiology Center, 300 Longwood Avenue, Boston, MA, United States; Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.
| | - Christoph J Fahrni
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Paul A Rosenberg
- Boston Children's Hospital, Department of Neurology and the F.M. Kirby Neurobiology Center, 300 Longwood Avenue, Boston, MA, United States; Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
209
|
Cotrim CA, Jarrott RJ, Martin JL, Drew D. A structural overview of the zinc transporters in the cation diffusion facilitator family. Acta Crystallogr D Struct Biol 2019; 75:357-367. [PMID: 30988253 PMCID: PMC6465983 DOI: 10.1107/s2059798319003814] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/19/2019] [Indexed: 01/07/2023] Open
Abstract
The cation diffusion facilitators (CDFs) are a family of membrane-bound proteins that maintain cellular homeostasis of essential metal ions. In humans, the zinc-transporter CDF family members (ZnTs) play important roles in zinc homeostasis. They do this by facilitating zinc efflux from the cytoplasm to the extracellular space across the plasma membrane or into intracellular organelles. Several ZnTs have been implicated in human health owing to their association with type 2 diabetes and neurodegenerative diseases. Although the structure determination of CDF family members is not trivial, recent advances in membrane-protein structural biology have resulted in two structures of bacterial YiiPs and several structures of their soluble C-terminal domains. These data reveal new insights into the molecular mechanism of ZnT proteins, suggesting a unique rocking-bundle mechanism that provides alternating access to the metal-binding site.
Collapse
Affiliation(s)
- Camila A. Cotrim
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Russell J. Jarrott
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Jennifer L. Martin
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - David Drew
- Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
210
|
Masiulionytė B, Valiulytė I, Tamašauskas A, Skiriutė D. Metallothionein Genes are Highly Expressed in Malignant Astrocytomas and Associated with Patient Survival. Sci Rep 2019; 9:5406. [PMID: 30932010 PMCID: PMC6443939 DOI: 10.1038/s41598-019-41974-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/18/2019] [Indexed: 02/06/2023] Open
Abstract
Gliomas are heterogeneous, primary brain tumours that originate from glial cells. The main type of gliomas is astrocytomas. There are four grades (I-IV) of astrocytoma malignancy. Astrocytoma grade IV known as glioblastoma multiforme (GBM) is the most common and aggressive type of astrocytic gliomas. Metallothioneins (MT) are low molecular weight, cysteine rich proteins encoded by a family of metallothionein (MT) genes. MT genes play a crucial role in carcinogenesis of diverse malignancies. We proposed MT genes as prognostic markers for malignant astrocytoma. MT1A, MT1E, MT1X, MT2, MT3 gene expression was elevated in grade IV astrocytomas (glioblastomas) as compared to astrocytomas grade I-III. Statistically significant differences were reached for MT1A and MT2 genes (Mann-Whitney test, p < 0.05). High MT1A, MT1X, MT2, MT3 genes expression was associated with shorter patient survival (Log-rank test, p < 0.05). MT1A gene promoter methylation was decreased in glioblastoma (57.6%) while the gene was highly methylated in grade II-III astrocytoma (from 66.7% to 83.3%) and associated with better patient survival (p < 0.05). MT1A gene methylation showed a trend of being associated with higher mRNA expression level in astrocytomas. Increased MT genes expression in grade IV astrocytomas as compared to I-III grade astrocytomas could be associated with malignant tumour behaviour and progression.
Collapse
Affiliation(s)
- Bernadeta Masiulionytė
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania
| | - Indrė Valiulytė
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania
| | - Arimantas Tamašauskas
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania
| | - Daina Skiriutė
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania.
| |
Collapse
|
211
|
Siddiqui S, Singh A, Ali S, Yadav M, Pandey V, Sharma D. Metallothionein: Potential therapeutic target for osteosarcoma. JOURNAL OF ONCOLOGICAL SCIENCES 2019. [DOI: 10.1016/j.jons.2019.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
212
|
McNeill RV, Mason AS, Hodson ME, Catto JWF, Southgate J. Specificity of the Metallothionein-1 Response by Cadmium-Exposed Normal Human Urothelial Cells. Int J Mol Sci 2019; 20:E1344. [PMID: 30884885 PMCID: PMC6471910 DOI: 10.3390/ijms20061344] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 01/21/2023] Open
Abstract
Occupational and environmental exposure to cadmium is associated with the development of urothelial cancer. The metallothionein (MT) family of genes encodes proteins that sequester metal ions and modulate physiological processes, including zinc homeostasis. Little is known about the selectivity of expression of the different MT isoforms. Here, we examined the effect of cadmium exposure on MT gene and isoform expression by normal human urothelial (NHU) cell cultures. Baseline and cadmium-induced MT gene expression was characterized by next-generation sequencing and RT-PCR; protein expression was assessed by Western blotting using isoform-specific antibodies. Expression of the zinc transporter-1 (SLC30A1) gene was also assessed. NHU cells displayed transcription of MT-2A, but neither MT-3 nor MT-4 genes. Most striking was a highly inducer-specific expression of MT-1 genes, with cadmium inducing transcription of MT-1A, MT-1G, MT-1H, and MT-1M. Whereas MT-1G was also induced by zinc and nickel ions and MT-1H by iron, both MT-1A and MT-1M were highly cadmium-specific, which was confirmed for protein using isoform-specific antibodies. Protein but not transcript endured post-exposure, probably reflecting sequestration. SLC30A1 transcription was also affected by cadmium ion exposure, potentially reflecting perturbation of intracellular zinc homeostasis. We conclude that human urothelium displays a highly inductive profile of MT-1 gene expression, with two isoforms identified as highly specific to cadmium, providing candidate transcript and long-lived protein biomarkers of cadmium exposure.
Collapse
Affiliation(s)
- Rhiannon V McNeill
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, York Biomedical Research Institute, University of York, York YO10 5DD, UK.
| | - Andrew S Mason
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, York Biomedical Research Institute, University of York, York YO10 5DD, UK.
| | - Mark E Hodson
- Department of Environment and Geography, University of York, York YO10 5DD, UK.
| | - James W F Catto
- Academic Urology Unit, University of Sheffield, Sheffield S10 2TN, UK.
| | - Jennifer Southgate
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, York Biomedical Research Institute, University of York, York YO10 5DD, UK.
| |
Collapse
|
213
|
van der Putten C, Veth J, Sukurova L, Zuiderwijk-Sick EA, Simonetti E, Koenen HJPM, Burm SM, van Noort JM, IJzerman AP, van Hijum SAFT, Diavatopoulos D, Bajramovic JJ. TLR-Induced IL-12 and CCL2 Production by Myeloid Cells Is Dependent on Adenosine A 3 Receptor-Mediated Signaling. THE JOURNAL OF IMMUNOLOGY 2019; 202:2421-2430. [PMID: 30804043 DOI: 10.4049/jimmunol.1800618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 01/16/2019] [Indexed: 11/19/2022]
Abstract
TLR-induced signaling potently activates cells of the innate immune system and is subject to regulation at different levels. Inflammatory conditions are associated with increased levels of extracellular adenosine, which can modulate TLR-induced production of cytokines through adenosine receptor-mediated signaling. There are four adenosine receptor subtypes that induce different signaling cascades. In this study, we demonstrate a pivotal contribution of adenosine A3 receptor (A3R)-mediated signaling to the TLR4-induced expression of IL-12 in different types of human myeloid APC. In dendritic cells, IL-12 and CCL2 responses as evoked by TLR2, 3, 4, 5, and 8, as well as IL-12 responses evoked by whole pathogens, were all reduced when A3R-mediated signaling was blocked. As a result, concomitant production of IFN-γ and IL-17 by T cells was significantly inhibited. We further show that selective inhibition of A3R-mediated signaling reduced TLR-induced phosphorylation of the transcription factor STAT1 at tyrosine 701. Next-generation sequencing revealed that A3R-mediated signaling controls the expression of metallothioneins, known inhibitors of STAT1 phosphorylation. Together our results reveal a novel regulatory layer of innate immune responses, with a central role for metallothioneins and autocrine/paracrine signaling via A3Rs.
Collapse
Affiliation(s)
- Céline van der Putten
- Alternatives Unit, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands
| | - Jennifer Veth
- Alternatives Unit, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands
| | - Lejla Sukurova
- Alternatives Unit, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands
| | - Ella A Zuiderwijk-Sick
- Alternatives Unit, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands
| | - Elles Simonetti
- Laboratory of Pediatric Infectious Diseases, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Hans J P M Koenen
- Laboratory of Pediatric Infectious Diseases, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Saskia M Burm
- Alternatives Unit, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands
| | | | - Ad P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, 2333 CC Leiden, the Netherlands
| | - Sacha A F T van Hijum
- Bacterial (Meta)genomics, Centre for Molecular and Biomolecular Informatics, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands; and.,NIZO, 6718 ZB Ede, the Netherlands
| | - Dimitri Diavatopoulos
- Laboratory of Pediatric Infectious Diseases, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Jeffrey J Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, 2288 GJ Rijswijk, the Netherlands;
| |
Collapse
|
214
|
Tsui KH, Hou CP, Chang KS, Lin YH, Feng TH, Chen CC, Shin YS, Juang HH. Metallothionein 3 Is a Hypoxia-Upregulated Oncogene Enhancing Cell Invasion and Tumorigenesis in Human Bladder Carcinoma Cells. Int J Mol Sci 2019; 20:ijms20040980. [PMID: 30813460 PMCID: PMC6413184 DOI: 10.3390/ijms20040980] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/18/2022] Open
Abstract
Metallothioneins have been viewed as modulators in a number of biological regulations regarding cancerous development; however, the function of metallothionein 3 (MT3) in bladder cancer is unexplored. We determined the regulatory mechanisms and potential function of MT3 in bladder carcinoma cells. Real-Time Reverse Transcriptase-Polymerase Chain Reaction (RT-qPCR) assays revealed that TSGH-8301 cells expressed more MT3 levels than RT-4, HT1376, and T24 cells. Immunoblot and RT-qPCR assays showed that arsenic (AS₂O₃) treatments enhanced the gene expression of MT3. Hypoxia induced HIF-1α, HIF-2α, and MT3 expression; furthermore, HIF-2α-knockdown attenuated hypoxic activation on MT3 expression. Ectopic overexpression of MT3 increased cell proliferation, invasion, and tumorigenesis significantly in T24 and HT1376 cells in vitro and in vivo; however, MT3-knockdown in TSGH-8301 cells had the reverse effect. Moreover, knockdown of MT3 enhanced arsenic-induced apoptosis determined by the Annexin V-FITC apoptosis assay. MT3-overexpression downregulated the gene expressions of N-myc downstream regulated gene 1 (NDRG1), N-myc downstream regulated gene 2 (NDRG2), and the mammary serine protease inhibitor (MASPIN) in HT1376 and T24 cells, whereas MT3-knockdown in TSGH-8301 cells had the opposite effect. The experiments indicated that MT3 is an arsenic- and hypoxia-upregulated oncogene that promotes cell growth and invasion of bladder carcinoma cells via downregulation of NDRG1, NDRG2, and MASPIN expressions.
Collapse
Affiliation(s)
- Ke-Hung Tsui
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| | - Chen-Pang Hou
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan.
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| | - Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan.
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| | - Chiu-Chun Chen
- Department of Medicine, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| | - Yi-Syuan Shin
- Department of Medicine, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan.
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan.
| |
Collapse
|
215
|
Adulcikas J, Sonda S, Norouzi S, Sohal SS, Myers S. Targeting the Zinc Transporter ZIP7 in the Treatment of Insulin Resistance and Type 2 Diabetes. Nutrients 2019; 11:nu11020408. [PMID: 30781350 PMCID: PMC6412268 DOI: 10.3390/nu11020408] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/13/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a disease associated with dysfunctional metabolic processes that lead to abnormally high levels of blood glucose. Preceding the development of T2DM is insulin resistance (IR), a disorder associated with suppressed or delayed responses to insulin. The effects of this response are predominately mediated through aberrant cell signalling processes and compromised glucose uptake into peripheral tissue including adipose, liver and skeletal muscle. Moreover, a major factor considered to be the cause of IR is endoplasmic reticulum (ER) stress. This subcellular organelle plays a pivotal role in protein folding and processes that increase ER stress, leads to maladaptive responses that result in cell death. Recently, zinc and the proteins that transport this metal ion have been implicated in the ER stress response. Specifically, the ER-specific zinc transporter ZIP7, coined the "gate-keeper" of zinc release from the ER into the cytosol, was shown to be essential for maintaining ER homeostasis in intestinal epithelium and myeloid leukaemia cells. Moreover, ZIP7 controls essential cell signalling pathways similar to insulin and activates glucose uptake in skeletal muscle. Accordingly, ZIP7 may be essential for the control of ER localized zinc and mechanisms that disrupt this process may lead to ER-stress and contribute to IR. Accordingly, understanding the mechanisms of ZIP7 action in the context of IR may provide opportunities to develop novel therapeutic options to target this transporter in the treatment of IR and subsequent T2DM.
Collapse
Affiliation(s)
- John Adulcikas
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Sabrina Sonda
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Shaghayegh Norouzi
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Sukhwinder Singh Sohal
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| | - Stephen Myers
- College of Health and Medicine, School of Health Sciences, University of Tasmania, TAS 7005, Australia.
| |
Collapse
|
216
|
Zinc regulates ERp44-dependent protein quality control in the early secretory pathway. Nat Commun 2019; 10:603. [PMID: 30723194 PMCID: PMC6363758 DOI: 10.1038/s41467-019-08429-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/09/2019] [Indexed: 01/14/2023] Open
Abstract
Zinc ions (Zn2+) are imported into the early secretory pathway by Golgi-resident transporters, but their handling and functions are not fully understood. Here, we show that Zn2+ binds with high affinity to the pH-sensitive chaperone ERp44, modulating its localization and ability to retrieve clients like Ero1α and ERAP1 to the endoplasmic reticulum (ER). Silencing the Zn2+ transporters that uptake Zn2+ into the Golgi led to ERp44 dysfunction and increased secretion of Ero1α and ERAP1. High-resolution crystal structures of Zn2+-bound ERp44 reveal that Zn2+ binds to a conserved histidine-cluster. The consequent large displacements of the regulatory C-terminal tail expose the substrate-binding surface and RDEL motif, ensuring client capture and retrieval. ERp44 also forms Zn2+-bridged homodimers, which dissociate upon client binding. Histidine mutations in the Zn2+-binding sites compromise ERp44 activity and localization. Our findings reveal a role of Zn2+ as a key regulator of protein quality control at the ER-Golgi interface. Zinc ions (Zn2+) are imported by Golgi-resident transporters but the function of zinc in the early secretory pathway has remained unknown. Here the authors find that Zn2+ regulates protein quality control in the early secretory pathway by demonstrating that the pH-sensitive chaperone ERp44 binds Zn2+ and solving the Zn2+-bound ERp44 structure.
Collapse
|
217
|
Chabosseau P, Woodier J, Cheung R, Rutter GA. Sensors for measuring subcellular zinc pools. Metallomics 2019; 10:229-239. [PMID: 29431830 DOI: 10.1039/c7mt00336f] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Zinc homeostasis is essential for normal cellular function, and defects in this process are associated with a number of diseases including type 2 diabetes (T2D), neurological disorders and cardiovascular disease. Thus, variants in the SLC30A8 gene, encoding the vesicular/granular zinc transporter ZnT8, are associated with altered insulin release and increased T2D risk while the zinc importer ZIP12 is implicated in pulmonary hypertension. In light of these, and findings in other diseases, recent efforts have focused on the development of refined sensors for intracellular free zinc ions that can be targeted to subcellular regions including the cytosol, endoplasmic reticulum (ER), secretory granules, Golgi apparatus, nucleus and the mitochondria. Here, we discuss recent advances in Zn2+ probe engineering and their applications to the measurement of labile subcellular zinc pools in different cell types.
Collapse
Affiliation(s)
- Pauline Chabosseau
- Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| | | | | | | |
Collapse
|
218
|
Vaden RM, Guillen KP, Salvant JM, Santiago CB, Gibbons JB, Pathi SS, Arunachalam S, Sigman MS, Looper RE, Welm BE. A Cancer-Selective Zinc Ionophore Inspired by the Natural Product Naamidine A. ACS Chem Biol 2019; 14:106-117. [PMID: 30571086 DOI: 10.1021/acschembio.8b00977] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We present data demonstrating the natural product mimic, zinaamidole A (ZNA), is a modulator of metal ion homeostasis causing cancer-selective cell death by specifically inducing cellular Zn2+-uptake in transformed cells. ZNA's cancer selectivity was evaluated using metastatic, patient-derived breast cancer cells, established human breast cancer cell lines, and three-dimensional organoid models derived from normal and transformed mouse mammary glands. Structural analysis of ZNA demonstrated that the compound interacts with zinc through the N2-acyl-2-aminoimidazole core. Combination treatment with ZnSO4 strongly potentiated ZNA's cancer-specific cell death mechanism, an effect that was not observed with other transition metals. We show that Zn2+-dyshomeostasis induced by ZNA is unique and markedly more selective than other known Zn2+-interacting compounds such as clioquinol. The in vivo bioactivity of ZNA was also assessed and revealed that tumor-bearing mice treated with ZNA had improved survival outcomes. Collectively, these data demonstrate that the N2-acyl-2-aminoimidazole core of ZNA represents a powerful chemotype to induce cell death in cancer cells concurrently with a disruption in zinc homeostasis.
Collapse
Affiliation(s)
- Rachel M. Vaden
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | | | - Justin M. Salvant
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | - Celine B. Santiago
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | - Joseph B. Gibbons
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | | | | | - Matthew S. Sigman
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | - Ryan E. Looper
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | | |
Collapse
|
219
|
Polykretis P, Cencetti F, Donati C, Luchinat E, Banci L. Cadmium effects on superoxide dismutase 1 in human cells revealed by NMR. Redox Biol 2019; 21:101102. [PMID: 30654299 PMCID: PMC6348768 DOI: 10.1016/j.redox.2019.101102] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/29/2018] [Accepted: 01/07/2019] [Indexed: 11/24/2022] Open
Abstract
Cadmium is a toxic pollutant that in recent decades has become more widespread in the environment due to anthropogenic activities, significantly increasing the risk of exposure. Concurrently, a continually growing body of research has begun to enumerate the harmful effects that this heavy metal has on human health. Consequently, additional research is required to better understand the mechanism and effects of cadmium at the molecular level. The main mechanism of cadmium toxicity is based on the indirect induction of severe oxidative stress, through several processes that unbalance the anti-oxidant cellular defence system, including the displacement of metals such as zinc from its native binding sites. Such mechanism was thought to alter the in vivo enzymatic activity of SOD1, one of the main antioxidant proteins of many tissues, including the central nervous system. SOD1 misfolding and aggregation is correlated with cytotoxicity in neurodegenerative diseases such as amyotrophic lateral sclerosis. We assessed the effect of cadmium on SOD1 folding and maturation pathway directly in human cells through in-cell NMR. Cadmium does not directly bind intracellular SOD1, instead causes the formation of its intramolecular disulfide bond in the zinc-bound form. Metallothionein overexpression is strongly induced by cadmium, reaching NMR-detectable levels. The intracellular availability of zinc modulates both SOD1 oxidation and metallothionein overexpression, strengthening the notion that zinc-loaded metallothioneins help maintaining the redox balance under cadmium-induced acute stress. Cadmium does not bind to superoxide dismutase 1 (SOD1) in human cells. In defect of zinc, cadmium causes the premature oxidation of SOD1. Cadmium induces the overexpression of metallothioneins to levels detectable by NMR. Zinc modulates metallothionein expression and attenuates SOD1 oxidation.
Collapse
Affiliation(s)
- Panagis Polykretis
- Magnetic Resonance Center - CERM, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Florence, Italy
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Enrico Luchinat
- Magnetic Resonance Center - CERM, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| | - Lucia Banci
- Magnetic Resonance Center - CERM, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Florence, Italy; Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
220
|
Chun H, Korolnek T, Lee CJ, Coyne HJ, Winge DR, Kim BE, Petris MJ. An extracellular histidine-containing motif in the zinc transporter ZIP4 plays a role in zinc sensing and zinc-induced endocytosis in mammalian cells. J Biol Chem 2018; 294:2815-2826. [PMID: 30593504 DOI: 10.1074/jbc.ra118.005203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/21/2018] [Indexed: 01/27/2023] Open
Abstract
Zinc is an essential trace element that serves as a cofactor for enzymes in critical biochemical processes and also plays a structural role in numerous proteins. Zinc transporter ZIP4 (ZIP4) is a zinc importer required for dietary zinc uptake in the intestine and other cell types. Studies in cultured cells have reported that zinc stimulates the endocytosis of plasma membrane-localized ZIP4 protein, resulting in reduced cellular zinc uptake. Thus, zinc-regulated trafficking of ZIP4 is a key means for regulating cellular zinc homeostasis, but the underlying mechanisms are not well understood. In this study, we used mutational analysis, immunoblotting, HEK293 cells, and immunofluorescence microscopy to identify a histidine-containing motif (398HTH) in the first extracellular loop that is required for high sensitivity to low zinc concentrations in a zinc-induced endocytic response of mouse ZIP4 (mZIP4). Moreover, using synthetic peptides with selective substitutions and truncated mZIP4 variants, we provide evidence that histidine residues in this motif coordinate a zinc ion in mZIP4 homodimers at the plasma membrane. These findings suggest that 398HTH is an important zinc-sensing motif for eliciting high-affinity zinc-stimulated endocytosis of mZIP4 and provide insight into cellular mechanisms for regulating cellular zinc homeostasis in mammalian cells.
Collapse
Affiliation(s)
- Haarin Chun
- From the Department of Animal and Avian Sciences, and
| | | | - Chul-Jin Lee
- the Unit on Structural and Chemical Biology of Membrane Proteins, NICHD, National Institutes of Health, Bethesda, Maryland 20892
| | - H Jerome Coyne
- the Departments of Medicine and Biochemistry, University of Utah Health Sciences Center, Salt Lake City, Utah 84132, and
| | - Dennis R Winge
- the Departments of Medicine and Biochemistry, University of Utah Health Sciences Center, Salt Lake City, Utah 84132, and
| | - Byung-Eun Kim
- From the Department of Animal and Avian Sciences, and .,Biological Sciences Graduate Program, University of Maryland, College Park, Maryland 20742
| | - Michael J Petris
- the Departments of Biochemistry and .,Nutrition and Exercise Physiology, and.,Christopher S. Bond Life Science Center, University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
221
|
Pigors M, Common JE, Wong XFCC, Malik S, Scott CA, Tabarra N, Liany H, Liu J, Limviphuvadh V, Maurer-Stroh S, Tang MB, Lench N, Margolis DJ, van Heel DA, Mein CA, Novak N, Baurecht H, Weidinger S, McLean WI, Irvine AD, O’Toole EA, Simpson MA, Kelsell DP. Exome Sequencing and Rare Variant Analysis Reveals Multiple Filaggrin Mutations in Bangladeshi Families with Atopic Eczema and Additional Risk Genes. J Invest Dermatol 2018; 138:2674-2677. [DOI: 10.1016/j.jid.2018.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 01/26/2023]
|
222
|
Wong DL, Stillman MJ. Metallothionein: An Aggressive Scavenger-The Metabolism of Rhodium(II) Tetraacetate (Rh 2(CH 3CO 2) 4). ACS OMEGA 2018; 3:16314-16327. [PMID: 31458267 PMCID: PMC6643557 DOI: 10.1021/acsomega.8b02161] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/13/2018] [Indexed: 06/10/2023]
Abstract
Anthropogenic sources of xenobiotic metals with no physiological benefit are increasingly prevalent in the environment. The platinum group metals (Pd, Pt, Rh, Ru, Os, and Ir) are found in marine and plant species near urban sources, and are known to bioaccumulate, introducing these metals into the human food chain. Many of these metals are also being used in innovative cancer therapy, which leads to a direct source of exposure for humans. This paper aims to further our understanding of nontraditional metal metabolism via metallothionein, a protein involved in physiologically important metal homeostasis. The aggressive reaction of metallothionein and dirhodium(II) tetraacetate, a common synthetic catalyst known for its cytotoxicity, was studied in detail in vitro. Optical spectroscopic and equilibrium and time-dependent mass spectral data were used to define binding constants for this robust reaction, and molecular dynamics calculations were conducted to explain the observed results.
Collapse
Affiliation(s)
- Daisy L. Wong
- Department of Chemistry, The
University of Western Ontario, 1151 Richmond Street, N6A 5B7 London, Ontario, Canada
| | - Martin J. Stillman
- Department of Chemistry, The
University of Western Ontario, 1151 Richmond Street, N6A 5B7 London, Ontario, Canada
| |
Collapse
|
223
|
Role of Zinc Signaling in the Regulation of Mast Cell-, Basophil-, and T Cell-Mediated Allergic Responses. J Immunol Res 2018; 2018:5749120. [PMID: 30596108 PMCID: PMC6286780 DOI: 10.1155/2018/5749120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 10/23/2018] [Indexed: 01/26/2023] Open
Abstract
Zinc is essential for maintaining normal structure and physiological function of cells. Its deficiency causes growth retardation, immunodeficiency, and neuronal degeneration. Zinc homeostasis is tightly regulated by zinc transporters and metallothioneins that control zinc concentration and its distribution in individual cells and contributes to zinc signaling. The intracellular zinc signaling regulates immune reactions. Although many molecules involved in these processes have zinc-binding motifs, the molecular mechanisms and the role of zinc in immune responses have not been elucidated. We and others have demonstrated that zinc signaling plays diverse and specific roles in vivo and in vitro in studies using knockout mice lacking zinc transporter function and metallothionein function. In this review, we discuss the impact of zinc signaling focusing particularly on mast cell-, basophil-, and T cell-mediated inflammatory and allergic responses. We also describe zinc signaling dysregulation as a leading health problem in inflammatory disease and allergy.
Collapse
|
224
|
Golan Y, Lehvy A, Horev G, Assaraf YG. High proportion of transient neonatal zinc deficiency causing alleles in the general population. J Cell Mol Med 2018; 23:828-840. [PMID: 30450693 PMCID: PMC6349188 DOI: 10.1111/jcmm.13982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 01/01/2023] Open
Abstract
Loss of function (LoF) mutations in the zinc transporter SLC30A2/ZnT2 result in impaired zinc secretion into breast milk consequently causing transient neonatal zinc deficiency (TNZD) in exclusively breastfed infants. However, the frequency of TNZD causing alleles in the general population is yet unknown. Herein, we investigated 115 missense SLC30A2/ZnT2 mutations from the ExAC database, equally distributed in the entire coding region, harboured in 668 alleles in 60 706 healthy individuals of diverse ethnicity. To estimate the frequency of LoF SLC30A2/ZnT2 mutations in the general population, we used bioinformatics tools to predict the potential impact of these mutations on ZnT2 functionality, and corroborated these predictions by a zinc transport assay in human MCF-7 cells. We found 14 missense mutations that were markedly deleterious to zinc transport. Together with two conspicuous LoF mutations in the ExAC database, 26 SLC30A2/ZnT2 alleles harboured deleterious mutations, suggesting that at least 1 in 2334 newborn infants are at risk to develop TNZD. This high frequency of TNZD mutations combined with the World Health Organization-promoted increase in the rate of exclusive breastfeeding highlights the importance of genetic screening for inactivating SLC30A2/ZnT2 mutations in the general population for the early diagnosis and prevention of TNZD.
Collapse
Affiliation(s)
- Yarden Golan
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Adrian Lehvy
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Guy Horev
- Bioinformatics Knowledge Unit, The Lorry I. Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
225
|
Song G, Yuan S, Wen X, Xie Z, Lou L, Hu B, Cai Q, Xu B. Transcriptome analysis of Cd-treated switchgrass root revealed novel transcripts and the importance of HSF/HSP network in switchgrass Cd tolerance. PLANT CELL REPORTS 2018; 37:1485-1497. [PMID: 30003312 DOI: 10.1007/s00299-018-2318-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/19/2018] [Indexed: 05/25/2023]
Abstract
Transcriptome analysis of Cd-treated switchgrass roots not only revealed novel switchgrass transcripts and gene structures but also highlighted the indispensable role of HSF/HSP network in switchgrass Cd tolerance. Switchgrass (Panicum virgatum L.), a C4 perennial tall grass, can be used for revegetation of Cd-contaminated soil. In the present study, a comparative transcriptome analysis of Cd-treated switchgrass roots was conducted. The result revealed a total of 462 novel transcripts and refined gene structures of 2337 transcripts. KEGG pathway and Gene Ontology analyses of the differentially expressed genes (DEGs) suggested that activation of redox homeostasis and oxidation-related metabolic processes were the primary response to Cd stress in switchgrass roots. In particular, 21 out of 23 differentially expressed shock transcription factor genes (HSFs), and 22 out of 23 differentially expressed heat shock protein genes (HSPs) had increased expression levels after Cd treatment. Furthermore, over-expressing one HSP-encoding gene in Arabidopsis significantly improved plant Cd tolerance. The result highlighted the activation of the redox homeostasis and the involvement of the HSF/HSP network in re-establishing normal protein conformation and thus cellular homeostasis in switchgrass upon Cd stress. These DEGs, especially those of the HSF/HSP network, could be used as candidate genes for further functional studies toward improved plant Cd tolerance in switchgrass and related species.
Collapse
Affiliation(s)
- Gang Song
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
- Jiangsu Vocational College of Agriculture and Forestry, Jurong, 212400, People's Republic of China
| | - Shaoxun Yuan
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Xuehui Wen
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Zheni Xie
- College of Agro-grassland Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Laiqing Lou
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Bingyu Hu
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Qingsheng Cai
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - Bin Xu
- College of Agro-grassland Science, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
226
|
Ollig J, Kloubert V, Taylor KM, Rink L. B cell activation and proliferation increase intracellular zinc levels. J Nutr Biochem 2018; 64:72-79. [PMID: 30448545 PMCID: PMC6372723 DOI: 10.1016/j.jnutbio.2018.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/12/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022]
Abstract
Zinc ions serve as second messengers in major cellular pathways, including the regulation pathways of proliferation and their proper regulation is necessary for homeostasis and a healthy organism. Accordingly, expression of zinc transporters can be altered in various cancer cell lines and is often involved in producing elevated intracellular zinc levels. In this study, human B cells were infected with Epstein–Barr virus (EBV) to generate immortalized cells, which revealed traits of tumor cells, such as high proliferation rates and an extended lifespan. These cells showed differentially altered zinc transporter expression with ZIP7 RNA and protein expression being especially increased as well as a corresponding increased phosphorylation of ZIP7 in EBV-transformed B cells. Accordingly, free zinc levels were elevated within these cells. To prove whether the observed changes resulted from immortalization or rather high proliferation, free zinc levels in in vitro activated B cells and in freshly isolated B cells expressing the activation marker CD69 were determined. Here, comparatively increased zinc levels were found, suggesting that activation and proliferation, but not immortalization, act as crucial factors for the elevation of intracellular free zinc.
Collapse
Affiliation(s)
- Johanna Ollig
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Veronika Kloubert
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074 Aachen, Germany.
| | - Kathryn M Taylor
- Breast Cancer Molecular Pharmacology, Welsh School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, United Kingdom.
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074 Aachen, Germany.
| |
Collapse
|
227
|
Essential Role of Zinc and Zinc Transporters in Myeloid Cell Function and Host Defense against Infection. J Immunol Res 2018; 2018:4315140. [PMID: 30417019 PMCID: PMC6207864 DOI: 10.1155/2018/4315140] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022] Open
Abstract
Zinc is an essential micronutrient known to play a vital role in host defense against pathogens. Diets that are deficient in zinc lead to impaired immunity and delayed recovery from and worse outcomes following infection. Sustained insufficient zinc intake leads to dysregulation of the innate immune response and increases susceptibility to infection whereas zinc supplementation in at-risk populations has been shown to restore host defense and reduce pathogen-related morbidity and mortality. Upon infection, zinc deficiency leads to increased pathology due to imbalance in key signaling networks that result in excessive inflammation and collateral tissue damage. In particular, zinc impacts macrophage function, a critical front-line cell in host defense, in addition to other immune cells. Deficits in zinc adversely impact macrophage function resulting in dysregulation of phagocytosis, intracellular killing, and cytokine production. An additional work in this field has revealed a vital role for several zinc transporter proteins that are required for proper bioredistribution of zinc within mononuclear cells to achieve an optimal immune response against invading microorganisms. In this review, we will discuss the most recent developments regarding zinc's role in innate immunity and protection against pathogen invasion.
Collapse
|
228
|
Function, Structure, and Transport Aspects of ZIP and ZnT Zinc Transporters in Immune Cells. J Immunol Res 2018; 2018:9365747. [PMID: 30370308 PMCID: PMC6189677 DOI: 10.1155/2018/9365747] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Abstract
Zinc is an important trace metal in immune systems, and zinc transporters are involved in many immune responses. Recent advances have revealed the structural and biochemical bases for zinc transport across the cell membrane, with clinical implications for the regulation of zinc homeostasis in immune cells like dendritic cells, T cells, B cells, and mast cells. In this review, we discuss the function, structure, and transport aspects of two major mammalian zinc transporter types, importers and exporters. First, Zrt-/Irt-like proteins (ZIPs) mediate the zinc influx from the extracellular or luminal side into the cytoplasm. There are 14 ZIP family members in humans. They form a homo- or heterodimer with 8 transmembrane domains and extra-/intracellular domains of various lengths. Several ZIP members show specific extracellular domains composed of two subdomains, a helix-rich domain and proline-alanine-leucine (PAL) motif-containing domain. Second, ZnT (zinc transporter) was initially identified in early studies of zinc biology; it mediates zinc efflux as a counterpart of ZIPs in zinc homeostasis. Ten family members have been identified. They show a unique architecture characterized by a Y-shaped conformation and a large cytoplasmic domain. A precise, comprehensive understanding of the structures and transport mechanisms of ZIP and ZnT in combination with mice experiments would provide promising drug targets as well as a basis for identifying other transporters with therapeutic potential.
Collapse
|
229
|
Coverdale JPC, Khazaipoul S, Arya S, Stewart AJ, Blindauer CA. Crosstalk between zinc and free fatty acids in plasma. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:532-542. [PMID: 30266430 PMCID: PMC6372834 DOI: 10.1016/j.bbalip.2018.09.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/23/2018] [Accepted: 09/23/2018] [Indexed: 12/13/2022]
Abstract
In mammalian blood plasma, serum albumin acts as a transport protein for free fatty acids, other lipids and hydrophobic molecules including neurodegenerative peptides, and essential metal ions such as zinc to allow their systemic distribution. Importantly, binding of these chemically extremely diverse entities is not independent, but linked allosterically. One particularly intriguing allosteric link exists between free fatty acid and zinc binding. Albumin thus mediates crosstalk between energy status/metabolism and organismal zinc handling. In recognition of the fact that even small changes in extracellular zinc concentration and speciation modulate the function of many cell types, the albumin-mediated impact of free fatty acid concentration on zinc distribution may be significant for both normal physiological processes including energy metabolism, insulin activity, heparin neutralisation, blood coagulation, and zinc signalling, and a range of disease states, including metabolic syndrome, cardiovascular disease, myocardial ischemia, diabetes, and thrombosis. Serum albumin binds and transports both free fatty acids and Zn2+ ions Elevated plasma free fatty acids impair Zn2+ binding by albumin through an allosteric mechanism The resulting changes in plasma zinc speciation are thought to impact blood coagulation and may promote thrombosis Increased free Zn2+ may lead to enhanced zinc export from plasma and dysregulation of zinc homeostasis in multiple tissues
Collapse
Affiliation(s)
| | | | - Swati Arya
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Alan J Stewart
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | | |
Collapse
|
230
|
Xiong X, Lan D, Li J, Lin Y, Zi X. Effects of Zinc Supplementation During In Vitro Maturation on Meiotic Maturation of Oocytes and Developmental Capacity in Yak. Biol Trace Elem Res 2018; 185:89-97. [PMID: 29247445 DOI: 10.1007/s12011-017-1217-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/04/2017] [Indexed: 12/26/2022]
Abstract
Zinc (Zn) is an essential trace element that is required during mammalian developmental processes. The objective of this study was to investigate the effects of Zn supplementation during in vitro maturation (IVM) on the developmental capacity of yak (Bos grunniens) oocytes. Cumulus expansion, nuclear maturation, intracellular glutathione (GSH), reactive oxygen species (ROS) levels, superoxide dismutase (SOD) activity, subsequent embryonic development, and the expression of Zn transporters (ZnTs) and Zrt and Irt-like proteins (ZiPs) were evaluated. The Zn concentrations in yak plasma and follicular fluid were 0.740 ± 0.012 and 0.382 ± 0.009 μg/mL, respectively. The cumulus expansion did not show significant differences in COCs after matured with or without Zn supplementation (P > 0.05). The intracellular GSH was higher in oocytes matured with 1 or 2 mg/L Zn than in control group (0 mg/L) (P < 0.05). However, ROS levels of oocytes matured with 1 or 2 mg/L Zn were reduced significantly compared with the control and 0.5 mg/L groups (P < 0.05). The SOD activity was increased significantly after Zn supplementation. The cleavage rate was not significantly different after Zn supplementation (P > 0.05). Percentages of matured oocytes that developed into the blastocyst stage after IVF were 47.9, 50.5, 60.4, and 58.9% for 0, 0.5, 1, and 2 mg/L Zn groups, respectively. Gene expression analysis revealed that the expression patterns associated with Zn were changed after Zn supplementation. In conclusion, Zn supplementation to IVM improved yak oocyte maturation and subsequent development by increasing GSH and SOD activity, decreasing ROS in oocytes.
Collapse
Affiliation(s)
- Xianrong Xiong
- College of Life Science and Technology, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Daoliang Lan
- College of Life Science and Technology, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Jian Li
- College of Life Science and Technology, Southwest Minzu University, Chengdu, Sichuan, 610041, China.
| | - Yaqiu Lin
- College of Life Science and Technology, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Xiangdong Zi
- College of Life Science and Technology, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
231
|
Ha E, Bae JH. Zinc transporter SLC39A11 polymorphisms are associated with chronic gastritis in the Korean population: the possible effect on spicy food intake. Nutr Res 2018; 57:78-85. [PMID: 30122198 DOI: 10.1016/j.nutres.2018.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/23/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022]
Abstract
Herein, we hypothesized that ZIP11 variants would be important risk factors for chronic gastritis and that there would be an interaction effect of the relationship between their variants and spicy food intake on the development of chronic gastritis. Participants in this cross-sectional study (n = 3882 + 252) were recruited from a cohort of the Korean Genome and Epidemiology Study in 2001. Age, sex, education, smoking and drinking status, exercise, stress, and income level of all participants were determined by a questionnaire. Demographic and anthropometric data were collected. Fasting blood samples were collected to determine the serum levels of glucose, insulin, total bilirubin, total cholesterol, high-density lipoprotein cholesterol, and triglycerides. The presence of chronic gastritis was defined as a confirmed diagnosis by a physician. Food consumption was determined using a semiquantitative food frequency questionnaire. We found 8 different single nucleotide polymorphisms (SNPs) that are significantly different between subjects without gastritis and those with gastritis. Of these 8 SNPs, 3 SNP (rs17183225 [C/T], rs17780814 [A/C], and rs17780820 [A/G]) are closely located in the intronic region of zinc transporter SLC39A11, commonly known as ZIP11, and show linkage disequilibrium (D' = 1.0). We also found that participants with (TCA + TCG) haplotype of ZIP11 at high levels of dietary intake of spicy foods show a significantly increasing tendency in the odds of having chronic gastritis when compared with those with CAA haplotype (odds ratio, 2.620; 95% confidence interval, 1.207-5.689). The data indicate positive associations between higher meal frequency and lower spicy food preference and gastritis. In conclusion, we found that zinc transporter gene ZIP11 is associated with chronic gastritis in the Korean population and it may interact with spicy food, which suggests ZIP11 as a therapeutic target for precision nutrition.
Collapse
Affiliation(s)
- Eunyoung Ha
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Ji-Hyun Bae
- Department of Food Science and Nutrition, College of Natural Sciences, Keimyung University, Daegu, Republic of Korea.
| |
Collapse
|
232
|
How cellular Zn 2+ signaling drives physiological functions. Cell Calcium 2018; 75:53-63. [PMID: 30145429 DOI: 10.1016/j.ceca.2018.08.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 01/10/2023]
Abstract
Zinc is an essential micronutrient affecting many aspects of human health. Cellular Zn2+ homeostasis is critical for cell function and survival. Zn2+, acting as a first or second messenger, triggers signaling pathways that mediate the physiological roles of Zn2+. Transient changes in Zn2+ concentrations within the cell or in the extracellular region occur following its release from Zn2+ binding metallothioneins, its transport across membranes by the ZnT or ZIP transporters, or release of vesicular Zn2+. These transients activate a distinct Zn2+ sensing receptor, ZnR/GPR39, or modulate numerous proteins and signaling pathways. Importantly, Zn2+ signaling regulates cellular physiological functions such as: proliferation, differentiation, ion transport and secretion. Indeed, novel therapeutic approaches aimed to maintain Zn2+ homeostasis and signaling are evolving. This review focuses on recent findings describing roles of Zn2+ and its transporters in regulating physiological or pathological processes.
Collapse
|
233
|
Park C, Jeong J. Synergistic cellular responses to heavy metal exposure: A minireview. Biochim Biophys Acta Gen Subj 2018; 1862:1584-1591. [PMID: 29631058 DOI: 10.1016/j.bbagen.2018.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 03/22/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Metal-responsive transcription factor 1 (MTF-1) induces the expression of metallothioneins (MTs) which bind and sequester labile metal ions. While MTF-1 primarily responds to excess metal exposure, additional stress response mechanisms are activated by excess metals. Evidence suggests potential crosstalk between responses mediated by MTF-1 and stress signaling enhances cellular tolerance to metal exposure. SCOPE OF REVIEW This review aims to summarize the current understanding of interaction between the stress response mediated by MTF-1 and other cellular mechanisms, notably the nuclear factor κB (NF-κB) and heat shock response (HSR). MAJOR CONCLUSIONS Crosstalk between MTF-1 mediated metal response and NF-κB signaling or HSR can modulate expression of stress proteins in response to metal exposure via effects on precursor signals or direct interaction of transcriptional activators. The interaction between stress signaling pathways can enhance cell survival and tolerance through a unified response system. GENERAL SIGNIFICANCE Elucidating the interactions between MTF-1 and cell stress response mechanisms is critical to a comprehensive understanding of metal-based cellular effects. Co-activation of HSR and NF-κB signaling allows the cell to detect metal contamination in the environment and improve survival outcomes.
Collapse
Affiliation(s)
- Chanyoung Park
- Program in Biochemistry and Biophysics, Amherst College, Amherst, MA 01002, United States
| | - Jeeyon Jeong
- Program in Biochemistry and Biophysics, Amherst College, Amherst, MA 01002, United States; Department of Biology, Amherst College, Amherst, MA 01002, United States.
| |
Collapse
|
234
|
Bornhorst J, Kipp AP, Haase H, Meyer S, Schwerdtle T. The crux of inept biomarkers for risks and benefits of trace elements. Trends Analyt Chem 2018. [DOI: 10.1016/j.trac.2017.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
235
|
Sheng M, Zhang G, Wang J, Yang Q, Zhao H, Cheng X, Xu Z. Remifentanil Induces Cardio Protection Against Ischemia/Reperfusion Injury by Inhibiting Endoplasmic Reticulum Stress Through the Maintenance of Zinc Homeostasis. Anesth Analg 2018; 127:267-276. [PMID: 29771714 DOI: 10.1213/ane.0000000000003414] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although it is well known that remifentanil (Rem) elicits cardiac protection against ischemia/reperfusion (I/R) injury, the underlying mechanism remains unclear. This study tested if Rem can protect the heart from I/R injury by inhibiting endoplasmic reticulum (ER) stress through the maintenance of zinc (Zn) homeostasis. METHODS Isolated rat hearts were subjected to 30 minutes of regional ischemia followed by 2 hours of reperfusion. Rem was given by 3 consecutive 5-minute infusions, and each infusion was followed by a 5-minute drug-free perfusion before ischemia. Total Zn concentrations in cardiac tissue, cardiac function, infarct size, and apoptosis were assessed. H9c2 cells were subjected to 6 hours of hypoxia and 2 hours of reoxygenation (hypoxia/reoxygenation [H/R]), and Rem was given for 30 minutes before hypoxia. Metal-responsive transcription factor 1 (MTF1) overexpression plasmids were transfected into H9c2 cells 48 hours before hypoxia. Intracellular Zn level, cell viability, and mitochondrial injury parameters were evaluated. A Zn chelator N,N,N',N'-tetrakis-(2-pyridylmethyl) ethylenediamine (TPEN) or an ER stress activator thapsigargin was administrated during in vitro and ex vivo studies. The regulatory molecules related to Zn homeostasis and ER stress in cardiac tissue, and cardiomyocytes were analyzed by Western blotting. RESULTS Rem caused significant reversion of Zn loss from the heart (Rem + I/R versus I/R, 9.43 ± 0.55 vs 7.53 ± 1.18; P < .05) by suppressing the expression of MTF1 and Zn transporter 1 (ZnT1). The inhibited expression of ER stress markers after Rem preconditioning was abolished by TPEN. Rem preconditioning improved the cardiac function accompanied by the reduction of infarct size (Rem + I/R versus I/R, 21% ± 4% vs 40% ± 6%; P < .05). The protective effects of Rem could be reserved by TPEN and thapsigargin. Similar effects were observed in H9c2 cells exposed to H/R. In addition, MTF1 overexpression blocked the inhibitory effects of Rem on ZnT1 expression and ER stress at reoxygenation. Rem attenuated the collapse of mitochondrial membrane potential (ΔΨm) and the generation of mitochondrial reactive oxygen species by inhibiting ER stress via cardiac Zn restoration (Rem + H/R versus H/R, 79.57% ± 10.62% vs 58.27% ± 4.32%; P < .05). CONCLUSIONS Rem maintains Zn homeostasis at reperfusion by inhibiting MTF1 and ZnT1 expression, leading to the attenuation of ER stress and cardiac injury. Our findings provide a promising therapeutic approach for managing acute myocardial I/R injury.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cation Transport Proteins/genetics
- Cation Transport Proteins/metabolism
- Cell Line
- Cytoprotection
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Disease Models, Animal
- Endoplasmic Reticulum Stress/drug effects
- Homeostasis
- Isolated Heart Preparation
- Male
- Membrane Potential, Mitochondrial/drug effects
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/physiopathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Rats, Wistar
- Reactive Oxygen Species/metabolism
- Remifentanil/pharmacology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Ventricular Function, Left/drug effects
- Zinc/metabolism
- Transcription Factor MTF-1
Collapse
Affiliation(s)
- Mingwei Sheng
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
- Department of Anesthesiology, Tianjin First Center Hospital, Tianjin, China
| | - Ge Zhang
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jiannan Wang
- Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Qing Yang
- Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Huanhuan Zhao
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xinxin Cheng
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Zhelong Xu
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
236
|
Thompson ED, Hogstrand C, Glover CN. From sea squirts to squirrelfish: facultative trace element hyperaccumulation in animals. Metallomics 2018; 10:777-793. [PMID: 29850752 DOI: 10.1039/c8mt00078f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The hyperaccumulation of trace elements is a widely characterized phenomenon in plants, bacteria, and fungi, but has received little attention in animals. However, there are numerous examples of animals that specifically and facultatively accumulate trace elements in the absence of elevated environmental concentrations. Metal hyperaccumulating animals are usually marine invertebrates, likely owing to environmental (e.g. constant exposure via the water) and physiological (e.g. osmoconforming and reduced integument permeability) factors. However, there are examples of terrestrial animals (insect larvae) and marine vertebrates (e.g. squirrelfish) that accumulate high body and/or tissue metal burdens. This review examines examples of animal hyperaccumulation of the elements arsenic, copper, iron, titanium, vanadium and zinc, describing mechanisms by which accumulation occurs and, where possible, hypothesizing functional roles. Groups such as the ascidians (sea squirts), molluscs (gastropods, bivalves and cephalopods) and polychaete annelids feature prominently as animals with hyperaccumulating capacity. Many of these species are potential model organisms offering insight into fundamental processes underlying metal handling, with relevance to human disease and aquatic metal toxicity, and some offer promise in applied fields such as bioremediation.
Collapse
Affiliation(s)
- E David Thompson
- Department of Biological Sciences, Northern Kentucky University, SC 245 Nunn Dr Highland Heights, KY 41099, USA.
| | | | | |
Collapse
|
237
|
Yu X, Kogan S, Chen Y, Tsang AT, Withers T, Lin H, Gilleran J, Buckley B, Moore D, Bertino J, Chan C, Kimball SD, Loh SN, Carpizo DR. Zinc Metallochaperones Reactivate Mutant p53 Using an ON/OFF Switch Mechanism: A New Paradigm in Cancer Therapeutics. Clin Cancer Res 2018; 24:4505-4517. [PMID: 29914895 DOI: 10.1158/1078-0432.ccr-18-0822] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/17/2018] [Accepted: 06/04/2018] [Indexed: 01/04/2023]
Abstract
Purpose: Zinc metallochaperones (ZMC) are a new class of anticancer drugs that reactivate zinc-deficient mutant p53 by raising and buffering intracellular zinc levels sufficiently to restore zinc binding. In vitro pharmacodynamics of ZMCs indicate that p53-mutant activity is ON by 4-6 hours and is OFF by 24. We sought to understand the mechanism of this regulation and to translate these findings preclinically. We further sought to innovate the formulation of ZMCs to improve efficacy.Experimental Design: We performed in vitro mechanistic studies to determine the role of cellular zinc homeostatic mechanisms in the transient pharmacodynamics of ZMCs. We conducted preclinical pharmacokinetic, pharmacodynamic, and efficacy studies using a genetically engineered murine pancreatic cancer model (KPC) to translate these mechanistic findings and investigate a novel ZMC formulation.Results:In vitro, cellular zinc homeostatic mechanisms that restore zinc to its physiologic levels function as the OFF switch in ZMC pharmacodynamics. In vivo pharmacokinetic studies indicate that ZMCs have a short half-life (< 30 minutes), which is sufficient to significantly improve survival in mice expressing a zinc-deficient allele (p53R172H) while having no effect in mice expressing a non-zinc-deficient allele (p53R270H). We synthesized a novel formulation of the drug in complex with zinc and demonstrate this significantly improves survival over ZMC1.Conclusions: Cellular zinc homeostatic mechanisms function as an OFF switch in ZMC pharmacodynamics, indicating that a brief period of p53-mutant reactivation is sufficient for on-target efficacy. ZMCs synthesized in complex with zinc are an improved formulation. Clin Cancer Res; 24(18); 4505-17. ©2018 AACR.
Collapse
Affiliation(s)
- Xin Yu
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Samuel Kogan
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Pharmacology, Rutgers University, Piscataway, New Jersey
| | - Ying Chen
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Ashley T Tsang
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Mount Sinai St. Luke's Roosevelt General Surgery Residency Program, New York, New York
| | - Tracy Withers
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Hongxia Lin
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - John Gilleran
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - Brian Buckley
- Rutgers Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey
| | - Dirk Moore
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Biostatistics. Rutgers School of Public Health, Rutgers University, New Brunswick, New Jersey
| | - Joseph Bertino
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Chang Chan
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - S David Kimball
- Mount Sinai St. Luke's Roosevelt General Surgery Residency Program, New York, New York.,Rutgers Translational Sciences, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey.,Z53 Therapeutics, Inc, Holmdel, New Jersey
| | - Stewart N Loh
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| | - Darren R Carpizo
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. .,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Pharmacology, Rutgers University, Piscataway, New Jersey.,Z53 Therapeutics, Inc, Holmdel, New Jersey
| |
Collapse
|
238
|
Abstract
Ventricular myocardial development is a well-orchestrated process involving different cardiac structures, multiple signal pathways, and myriad proteins. Dysregulation of this important developmental event can result in cardiomyopathies, such as left ventricle non-compaction, which affect the pediatric population and the adults. Human and mouse studies have shed light upon the etiology of some cardiomyopathy cases and highlighted the contribution of both genetic and environmental factors. However, the regulation of ventricular myocardial development remains incompletely understood. Zinc is an essential trace metal with structural, enzymatic, and signaling function. Perturbation of zinc homeostasis has resulted in developmental and physiological defects including cardiomyopathy. In this review, we summarize several mechanisms by which zinc and zinc transporters can impact the regulation of ventricular myocardial development. Based on our review, we propose that zinc deficiency and mutations of zinc transporters may underlie some cardiomyopathy cases especially those involving ventricular myocardial development defects.
Collapse
Affiliation(s)
- Wen Lin
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Deqiang Li
- Division of Cardiac Surgery, School of Medicine, University of Maryland, 800 West Baltimore ST, Rm 314, Baltimore, MD, 21201, USA.
| |
Collapse
|
239
|
Kogan S, Carpizo DR. Zinc Metallochaperones as Mutant p53 Reactivators: A New Paradigm in Cancer Therapeutics. Cancers (Basel) 2018; 10:E166. [PMID: 29843463 PMCID: PMC6025018 DOI: 10.3390/cancers10060166] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 12/22/2022] Open
Abstract
Restoration of wild-type structure and function to mutant p53 with a small molecule (hereafter referred to as "reactivating" mutant p53) is one of the holy grails in cancer therapeutics. The majority of TP53 mutations are missense which generate a defective protein that is targetable. We are currently developing a new class of mutant p53 reactivators called zinc metallochaperones (ZMCs) and, here, we review our current understanding of them. The p53 protein requires the binding of a single zinc ion, coordinated by four amino acids in the DNA binding domain, for proper structure and function. Loss of the wild-type structure by impairing zinc binding is a common mechanism of inactivating p53. ZMCs reactivate mutant p53 using a novel two-part mechanism that involves restoring the wild-type structure by reestablishing zinc binding and activating p53 through post-translational modifications induced by cellular reactive oxygen species (ROS). The former causes a wild-type conformation change, the later induces a p53-mediated apoptotic program to kill the cancer cell. ZMCs are small molecule metal ion chelators that bind zinc and other divalent metal ions strong enough to remove zinc from serum albumin, but weak enough to donate it to mutant p53. Recently we have extended our understanding of the mechanism of ZMCs to the role of cells' response to this zinc surge. We found that cellular zinc homeostatic mechanisms, which normally function to maintain free intracellular zinc levels in the picomolar range, are induced by ZMCs. By normalizing zinc levels, they function as an OFF switch to ZMCs because zinc levels are no longer sufficiently high to maintain a wild-type structure. This on/off switch leads to a transient nature to the mechanism of ZMCs in which mutant p53 activity comes on in a few hours and then is turned off. This finding has important implications for the translation of ZMCs to the clinic because it indicates that ZMC concentrations need not be maintained at high levels for their activity. Indeed, we found that short exposures (as little as 15 min) were adequate to observe the mutant p53 reactivating activity. This switch mechanism imparts an advantage over other targeted therapeutics in that efficacy can be accomplished with minimal exposure which minimizes toxicity and maximizes the therapeutic window. This on/off switch mechanism is unique in targeted cancer therapeutics and will impact the design of human clinical trials.
Collapse
Affiliation(s)
- Samuel Kogan
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
- Department of Pharmacology, Rutgers University, Piscataway, NJ 08854, USA.
| | - Darren R Carpizo
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
- Department of Pharmacology, Rutgers University, Piscataway, NJ 08854, USA.
- Z53 Therapeutics, Inc., Holmdel, NJ 07733, USA.
| |
Collapse
|
240
|
Eron SJ, MacPherson DJ, Dagbay KB, Hardy JA. Multiple Mechanisms of Zinc-Mediated Inhibition for the Apoptotic Caspases-3, -6, -7, and -8. ACS Chem Biol 2018; 13:1279-1290. [PMID: 29364645 PMCID: PMC5959779 DOI: 10.1021/acschembio.8b00064] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Zinc is emerging as a widely used and important biological regulatory signal. Cellular zinc levels are tightly regulated by a complex array of zinc importers and exporters to control processes such as apoptotic cell death. While caspase inhibition by zinc has been reported previously, the reported inhibition constants were too weak to suggest a critical biological role for zinc-mediated inhibition. In this work, we have adopted a method of assessing available zinc. This allowed assessment of accurate inhibition constants for apoptotic caspases, caspase-3, -6, -7, and -8. Each of these caspases are inhibited by zinc at intracellular levels but with widely differing inhibition constants and different zinc binding stoichiometries. Caspase-3, -6, and -8 appear to be constitutively inhibited by typical zinc levels, and this inhibition must be lifted to allow activation. The inhibition constant for caspase-7 (76 nM) is much weaker than for the other apoptotic caspases (2.6-6.9 nM) suggesting that caspase-7 is not inactivated by normal zinc concentrations but can be inhibited under conditions of zinc stress. Caspase-3, -7, and -8 were found to bind three, one, and two zincs, respectively. In each of these caspases, zinc was present in the active site, in contrast to caspase-6, which binds one zinc allosterically. The most notable new mechanism to emerge from this work is for zinc-mediated inhibition of caspase-8. Zinc binds caspase-8 directly at the active site and at a second site. Zinc binding inhibits formation of the caspase-8 dimer, the activated form of the enzyme. Together these findings suggest that zinc plays a critical role in regulation of apoptosis by direct inactivation of caspases, in a manner that is unique for each caspase.
Collapse
Affiliation(s)
- Scott J. Eron
- Department of Chemistry, 104 LGRT, 710 N. Pleasant St. University of Massachusetts Amherst, MA 01003, USA
| | - Derek J. MacPherson
- Department of Chemistry, 104 LGRT, 710 N. Pleasant St. University of Massachusetts Amherst, MA 01003, USA
| | - Kevin B. Dagbay
- Department of Chemistry, 104 LGRT, 710 N. Pleasant St. University of Massachusetts Amherst, MA 01003, USA
| | - Jeanne A. Hardy
- Department of Chemistry, 104 LGRT, 710 N. Pleasant St. University of Massachusetts Amherst, MA 01003, USA
| |
Collapse
|
241
|
Baltaci AK, Yuce K, Mogulkoc R. Zinc Metabolism and Metallothioneins. Biol Trace Elem Res 2018; 183:22-31. [PMID: 28812260 DOI: 10.1007/s12011-017-1119-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/02/2017] [Indexed: 12/20/2022]
Abstract
Among the trace elements, zinc is one of the most used elements in biological systems. Zinc is found in the structure of more than 2700 enzymes, including hydrolases, transferases, oxyreductases, ligases, isomerases, and lyases. Not surprisingly, it is present in almost all body cells. Preserving the stability and integrity of biological membranes and ion channels, zinc is also an intracellular regulator and provides structural support to proteins during molecular interactions. It acts as a structural element in nucleic acids or other gene-regulating proteins. Metallothioneins, the low molecular weight protein family rich in cysteine groups, are involved significantly in numerous physiological and pathological processes including particularly oxidative stress. A critical role of metallothioneins (MT) is to bind zinc with high affinity and to serve as an intracellular zinc reservoir. By releasing free intracellular zinc when needed, MTs mediate the unique physiological roles of zinc. MT expression is induced by zinc elevation, and thus, zinc homeostasis is maintained. That MT mediates the effects of zinc, besides having strong radical scavenging effects, points to the critical part it plays in oxidative stress. The present review aims to give information on metallothioneins, which have critical importance in the metabolism and molecular pathways of zinc.
Collapse
Affiliation(s)
| | - Kemal Yuce
- Department of Physiology, Medical Faculty, Selcuk University, Konya, Turkey
| | - Rasim Mogulkoc
- Department of Physiology, Medical Faculty, Selcuk University, Konya, Turkey
| |
Collapse
|
242
|
Determination of Metallothionein Isoforms in Fish by Cadmium Saturation Combined with Anion Exchange HPLC-ICP-MS. Chromatographia 2018. [DOI: 10.1007/s10337-018-3523-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
243
|
Intermittent hypoxia-generated ROS contributes to intracellular zinc regulation that limits ischemia/reperfusion injury in adult rat cardiomyocyte. J Mol Cell Cardiol 2018; 118:122-132. [PMID: 29577873 DOI: 10.1016/j.yjmcc.2018.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/05/2018] [Accepted: 03/21/2018] [Indexed: 12/20/2022]
Abstract
Intermittent hypoxia (IH) has been shown to exert cardioprotective effects against ischemia/reperfusion (I/R) injury through the preservation of ion homeostasis. I/R dramatically elevated cytosolic Zn2+ and caused cardiomyocyte death. However, the role of IH exposure in the relationship between Zn2+ regulation and cardioprotection is still unclear. The aim of the present study was to study whether IH exposure could help in intracellular Zn2+ regulation, hence contributing to cardioprotection against I/R injury. Adult rat cardiomyocytes were exposed to IH (5% O2, 5% CO2 and balanced N2) for 30 min followed by 30 min of normoxia (21% O2, 5% CO2 and balanced N2). Changes in intracellular Zn2+ concentration were determined using a Zn2+-specific fluorescent dye, FluoZin-3 or RhodZin-3. Fluorescence was monitored under an inverted fluorescent or confocal microscope. The results demonstrated that I/R or 2,2'-dithiodipyridine (DTDP), a reactive disulphide compound, induced Zn2+ release from metallothioneins (MTs), subsequently causing cytosolic Zn2+ overload, which in turn increased intracellular Zn2+ entry into the mitochondria via a Ca2+ uniporter, hence inducing mitochondrial membrane potential loss, and eventually led to cell death. However, the cytosolic Zn2+ overload and cell death caused by I/R or DTDP was significantly reduced by treatment of cardiomyocytes with IH. The findings from this study suggest that IH might exert its cardioprotective effect through reducing the I/R-induced cytosolic Zn2+ overload and cell death in cardiomyocytes.
Collapse
|
244
|
Critical Role of Zinc as Either an Antioxidant or a Prooxidant in Cellular Systems. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9156285. [PMID: 29743987 PMCID: PMC5884210 DOI: 10.1155/2018/9156285] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 01/11/2023]
Abstract
Zinc is recognized as an essential trace metal required for human health; its deficiency is strongly associated with neuronal and immune system defects. Although zinc is a redox-inert metal, it functions as an antioxidant through the catalytic action of copper/zinc-superoxide dismutase, stabilization of membrane structure, protection of the protein sulfhydryl groups, and upregulation of the expression of metallothionein, which possesses a metal-binding capacity and also exhibits antioxidant functions. In addition, zinc suppresses anti-inflammatory responses that would otherwise augment oxidative stress. The actions of zinc are not straightforward owing to its numerous roles in biological systems. It has been shown that zinc deficiency and zinc excess cause cellular oxidative stress. To gain insights into the dual action of zinc, as either an antioxidant or a prooxidant, and the conditions under which each role is performed, the oxidative stresses that occur in zinc deficiency and zinc overload in conjunction with the intracellular regulation of free zinc are summarized. Additionally, the regulatory role of zinc in mitochondrial homeostasis and its impact on oxidative stress are briefly addressed.
Collapse
|
245
|
Tejeda-Guzmán C, Rosas-Arellano A, Kroll T, Webb SM, Barajas-Aceves M, Osorio B, Missirlis F. Biogenesis of zinc storage granules in Drosophila melanogaster. J Exp Biol 2018; 221:jeb168419. [PMID: 29367274 PMCID: PMC5897703 DOI: 10.1242/jeb.168419] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/17/2018] [Indexed: 12/16/2022]
Abstract
Membrane transporters and sequestration mechanisms concentrate metal ions differentially into discrete subcellular microenvironments for use in protein cofactors, signalling, storage or excretion. Here we identify zinc storage granules as the insect's major zinc reservoir in principal Malpighian tubule epithelial cells of Drosophila melanogaster The concerted action of Adaptor Protein-3, Rab32, HOPS and BLOC complexes as well as of the white-scarlet (ABCG2-like) and ZnT35C (ZnT2/ZnT3/ZnT8-like) transporters is required for zinc storage granule biogenesis. Due to lysosome-related organelle defects caused by mutations in the homologous human genes, patients with Hermansky-Pudlak syndrome may lack zinc granules in beta pancreatic cells, intestinal paneth cells and presynaptic vesicles of hippocampal mossy fibers.
Collapse
Affiliation(s)
- Carlos Tejeda-Guzmán
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, C.P. 07360, México
| | - Abraham Rosas-Arellano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, C.P. 07360, México
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA 94025, USA
| | - Samuel M Webb
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA 94025, USA
| | - Martha Barajas-Aceves
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, C.P. 07360, México
| | - Beatriz Osorio
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, C.P. 07360, México
| | - Fanis Missirlis
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, C.P. 07360, México
| |
Collapse
|
246
|
Trace Elements and Healthcare: A Bioinformatics Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1005:63-98. [PMID: 28916929 DOI: 10.1007/978-981-10-5717-5_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Biological trace elements are essential for human health. Imbalance in trace element metabolism and homeostasis may play an important role in a variety of diseases and disorders. While the majority of previous researches focused on experimental verification of genes involved in trace element metabolism and those encoding trace element-dependent proteins, bioinformatics study on trace elements is relatively rare and still at the starting stage. This chapter offers an overview of recent progress in bioinformatics analyses of trace element utilization, metabolism, and function, especially comparative genomics of several important metals. The relationship between individual elements and several diseases based on recent large-scale systematic studies such as genome-wide association studies and case-control studies is discussed. Lastly, developments of ionomics and its recent application in human health are also introduced.
Collapse
|
247
|
Golan Y, Kambe T, Assaraf YG. The role of the zinc transporter SLC30A2/ZnT2 in transient neonatal zinc deficiency. Metallomics 2018; 9:1352-1366. [PMID: 28665435 DOI: 10.1039/c7mt00162b] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Breast milk is the optimal nutrient mix for infants until the age of 6 months. However, in some cases, due to genetic alterations as well as nutrient deficiencies in nursing mothers, infants may suffer from inadequate levels of micronutrients upon exclusive breastfeeding. In this respect, transient neonatal zinc deficiency (TNZD) is caused by loss-of-function mutations in the zinc transporter SLC30A2/ZnT2 gene, resulting in poor secretion of zinc into the breast milk. Consequently, infants exclusively breastfed with zinc-deficient breast milk develop severe zinc deficiency. The main initial symptoms of zinc deficiency are dermatitis, diarrhea, alopecia, and loss of appetite. Importantly, zinc supplementation of these zinc-deficient infants effectively and rapidly resolves these TNZD symptoms. In the current review, we present the major steps towards the identification of the molecular mechanisms underlying TNZD and propose novel approaches that could be implemented in order to achieve an early diagnosis of TNZD towards the prevention of TNZD morbidity. We also discuss the importance of assessing the prevalence of TNZD in the general population, while taking into consideration its autosomal dominant inheritance that was recently established, also supported by a large number of SLC30A2/ZnT2 variants recently identified in American lactating mothers. These findings indicating that TNZD is more frequent than initially thought, along with the increasing number of TNZD cases that were recently reported worldwide, prompted us here to highlight the importance of early diagnosis of SLC30A2/ZnT2 variants in order to supplement zinc-deficient infants in real-time, thus preventing TNZD morbidity and enhancing newborn health. This early genetic diagnosis of zinc deficiency could possibly prove to be a useful platform for the identification of other micronutrient deficiencies, which could be readily resolved by proper real-time supplementation of the infant's diet.
Collapse
Affiliation(s)
- Yarden Golan
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | | | | |
Collapse
|
248
|
Park Y, Zhang J, Cai L. Reappraisal of metallothionein: Clinical implications for patients with diabetes mellitus. J Diabetes 2018; 10:213-231. [PMID: 29072367 DOI: 10.1111/1753-0407.12620] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 08/29/2017] [Accepted: 10/20/2017] [Indexed: 12/22/2022] Open
Abstract
Reactive oxygen and nitrogen species (ROS and RNS, respectively) are byproducts of cellular physiological processes of the metabolism of intermediary nutrients. Although physiological defense mechanisms readily convert these species into water or urea, an improper balance between their production and removal leads to oxidative stress (OS), which is harmful to cellular components. This OS may result in uncontrolled growth or, ultimately, cell death. In addition, ROS and RNS are closely related to the development of diabetes and its complications. Therefore, numerous researchers have proposed the development of strategies for the removal of ROS/RNS to prevent or treat diabetes and its complications. Some molecules that are synthesized in the body or obtained from food participate in the removal and neutralization of ROS and RNS. Metallothionein, a cysteine-rich protein, is a metal-binding protein that has a wide range of functions in cellular homeostasis and immunity. Metallothionein can be induced by a variety of conditions, including zinc supplementation, and plays a crucial role in mediating anti-OS, anti-apoptotic, detoxification, and anti-inflammatory effects. Metallothionein can modulate various stress-induced signaling pathways (mitogen-activated protein kinase, Wnt, nuclear factor-κB, phosphatidylinositol 3-kinase, sirtuin 1/AMP-activated protein kinase and fibroblast growth factor 21) to alleviate diabetes and diabetic complications. However, a deeper understanding of the functional, biochemical, and molecular characteristics of metallothionein is needed to bring about new opportunities for OS therapy. This review focuses on newly proposed functions of a metallothionein and their implications relevant to diabetes and its complications.
Collapse
Affiliation(s)
- Yongsoo Park
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- Hanyang University, College of Medicine and Engineering, Seoul, South Korea
| | - Jian Zhang
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- The Center of Cardiovascular Disorders, The First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- Department of Radiation Oncology, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
249
|
Role of Zinc Homeostasis in the Pathogenesis of Diabetes and Obesity. Int J Mol Sci 2018; 19:ijms19020476. [PMID: 29415457 PMCID: PMC5855698 DOI: 10.3390/ijms19020476] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/11/2022] Open
Abstract
Zinc deficiency is a risk factor for obesity and diabetes. However, until recently, the underlying molecular mechanisms remained unclear. The breakthrough discovery that the common polymorphism in zinc transporter SLC30A8/ZnT8 may increase susceptibility to type 2 diabetes provided novel insights into the role of zinc in diabetes. Our group and others showed that altered ZnT8 function may be involved in the pathogenesis of type 2 diabetes, indicating that the precise control of zinc homeostasis is crucial for maintaining health and preventing various diseases, including lifestyle-associated diseases. Recently, the role of the zinc transporter ZIP13 in the regulation of beige adipocyte biogenesis was clarified, which indicated zinc homeostasis regulation as a possible therapeutic target for obesity and metabolic syndrome. Here we review advances in the role of zinc homeostasis in the pathophysiology of diabetes, and propose that inadequate zinc distribution may affect the onset of diabetes and metabolic diseases by regulating various critical biological events.
Collapse
|
250
|
Falfushynska HI, Gnatyshyna LL, Ivanina AV, Sokolova IM, Stoliar OB. Detoxification and cellular stress responses of unionid mussels Unio tumidus from two cooling ponds to combined nano-ZnO and temperature stress. CHEMOSPHERE 2018; 193:1127-1142. [PMID: 29874741 DOI: 10.1016/j.chemosphere.2017.11.079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 06/08/2023]
Abstract
Bivalve mollusks from the cooling reservoirs of fuel power plants (PP) are acclimated to the chronic heating and chemical pollution. We investigated stress responses of the mussels from these ponds to determine their tolerance to novel environmental pollutant, zinc oxide nanoparticles (nZnO). Male Unio tumidus from the reservoirs of Dobrotvir and Burschtyn PPs (DPP and BPP), Ukraine were exposed for 14 days to nZnO (3.1 μM), Zn2+ (3.1 μM) at 18 °C, elevated temperature (T, 25 °C), or nZnO at 25 °C (nZnO + T). Control groups were held at 18 °C. Zn-containing exposures resulted in the elevated concentrations of total and Zn-bound metallothionein (MT and Zn-MT) in the digestive gland, an increase in the levels of non-metalated MT (up to 5 times) and alkali-labile phosphates and lysosomal membrane destabilization in hemocytes. A common signature of nZnO exposures was modulation of the multixenobiotic-resistance protein activity (a decrease in the digestive gland and increase in the gills). The origin of population strongly affected the cellular stress responses of mussels. DPP-mussels showed depletion of caspase-3 in the digestive gland and up-regulation of HSP70, HSP72 and HSP60 levels in the gill during most exposures, whereas in the BPP-mussels caspase-3 was up-regulated and HSPs either downregulated or maintained stable. BPP-mussels were less adapted to heating shown by a glutathione depletion at elevated temperature (25 °C). Comparison with the earlier studies on mussels from pristine habitats show that an integrative 'eco-exposome'-based approach is useful for the forecast of the biological responses to novel adverse effects on aquatic organisms.
Collapse
Affiliation(s)
- Halina I Falfushynska
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil National Pedagogical University, Ternopil, Ukraine
| | - Lesya L Gnatyshyna
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil National Pedagogical University, Ternopil, Ukraine; Department of General Chemistry, Ternopil State Medical University, Ternopil, Ukraine
| | - Anna V Ivanina
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd., Charlotte, NC 28223, USA
| | - Inna M Sokolova
- Department of Marine Biology, Institute of Biological Sciences, University of Rostock, Rostock, Germany.
| | - Oksana B Stoliar
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil National Pedagogical University, Ternopil, Ukraine.
| |
Collapse
|