51
|
Zinc Signals and Immunity. Int J Mol Sci 2017; 18:ijms18102222. [PMID: 29064429 PMCID: PMC5666901 DOI: 10.3390/ijms18102222] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/13/2017] [Accepted: 10/19/2017] [Indexed: 01/11/2023] Open
Abstract
Zinc homeostasis is crucial for an adequate function of the immune system. Zinc deficiency as well as zinc excess result in severe disturbances in immune cell numbers and activities, which can result in increased susceptibility to infections and development of especially inflammatory diseases. This review focuses on the role of zinc in regulating intracellular signaling pathways in innate as well as adaptive immune cells. Main underlying molecular mechanisms and targets affected by altered zinc homeostasis, including kinases, caspases, phosphatases, and phosphodiesterases, will be highlighted in this article. In addition, the interplay of zinc homeostasis and the redox metabolism in affecting intracellular signaling will be emphasized. Key signaling pathways will be described in detail for the different cell types of the immune system. In this, effects of fast zinc flux, taking place within a few seconds to minutes will be distinguish from slower types of zinc signals, also designated as “zinc waves”, and late homeostatic zinc signals regarding prolonged changes in intracellular zinc.
Collapse
|
52
|
Giacconi R, Costarelli L, Piacenza F, Basso A, Bürkle A, Moreno-Villanueva M, Grune T, Weber D, Stuetz W, Gonos ES, Schön C, Grubeck-Loebenstein B, Sikora E, Toussaint O, Debacq-Chainiaux F, Franceschi C, Hervonen A, Slagboom E, Ciccarone F, Zampieri M, Caiafa P, Jansen E, Dollé MET, Breusing N, Mocchegiani E, Malavolta M. Zinc-Induced Metallothionein in Centenarian Offspring From a Large European Population: The MARK-AGE Project. J Gerontol A Biol Sci Med Sci 2017; 73:745-753. [DOI: 10.1093/gerona/glx192] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/10/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
- Robertina Giacconi
- Translational Research Center of Nutrition and Ageing, IRCCS-INRCA, Ancona, Italy
| | - Laura Costarelli
- Translational Research Center of Nutrition and Ageing, IRCCS-INRCA, Ancona, Italy
| | - Francesco Piacenza
- Translational Research Center of Nutrition and Ageing, IRCCS-INRCA, Ancona, Italy
| | - Andrea Basso
- Translational Research Center of Nutrition and Ageing, IRCCS-INRCA, Ancona, Italy
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Germany
| | | | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, Germany
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Nuthetal, Germany
| | - Wolfgang Stuetz
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Stuttgart, Germany
| | - Efstathios S Gonos
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | | | | | - Ewa Sikora
- Laboratory of the Molecular Bases of Ageing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Claudio Franceschi
- CIG-Interdepartmental Center “L. Galvani”, Alma Mater Studiorum, University of Bologna, Italy
| | | | - Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Centre, The Netherlands
| | - Fabio Ciccarone
- Department of Biology, University of Rome “Tor Vergata”, Italy
| | - Michele Zampieri
- Department of Cellular Biotechnologies and Hematology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Italy
- Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Paola Caiafa
- Department of Cellular Biotechnologies and Hematology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Italy
- Pasteur Institute-Fondazione Cenci Bolognetti, Rome, Italy
| | - Eugène Jansen
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Martijn E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Nicolle Breusing
- Department of Applied Nutritional Science/Dietetics, Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Eugenio Mocchegiani
- Translational Research Center of Nutrition and Ageing, IRCCS-INRCA, Ancona, Italy
| | - Marco Malavolta
- Translational Research Center of Nutrition and Ageing, IRCCS-INRCA, Ancona, Italy
| |
Collapse
|
53
|
Intracellular zinc activates KCNQ channels by reducing their dependence on phosphatidylinositol 4,5-bisphosphate. Proc Natl Acad Sci U S A 2017; 114:E6410-E6419. [PMID: 28716904 DOI: 10.1073/pnas.1620598114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
M-type (Kv7, KCNQ) potassium channels are proteins that control the excitability of neurons and muscle cells. Many physiological and pathological mechanisms of excitation operate via the suppression of M channel activity or expression. Conversely, pharmacological augmentation of M channel activity is a recognized strategy for the treatment of hyperexcitability disorders such as pain and epilepsy. However, physiological mechanisms resulting in M channel potentiation are rare. Here we report that intracellular free zinc directly and reversibly augments the activity of recombinant and native M channels. This effect is mechanistically distinct from the known redox-dependent KCNQ channel potentiation. Interestingly, the effect of zinc cannot be attributed to a single histidine- or cysteine-containing zinc-binding site within KCNQ channels. Instead, zinc dramatically reduces KCNQ channel dependence on its obligatory physiological activator, phosphatidylinositol 4,5-bisphosphate (PIP2). We hypothesize that zinc facilitates interactions of the lipid-facing interface of a KCNQ protein with the inner leaflet of the plasma membrane in a way similar to that promoted by PIP2 Because zinc is increasingly recognized as a ubiquitous intracellular second messenger, this discovery might represent a hitherto unknown native pathway of M channel modulation and provide a fresh strategy for the design of M channel activators for therapeutic purposes.
Collapse
|
54
|
Abstract
The cellular constitution of Zn-proteins and Zn-dependent signaling depend on the capacity of Zn2+ to find specific binding sites in the face of a plethora of other high affinity ligands. The most prominent of these is metallothionein (MT). It serves as a storage site for Zn2+ under various conditions, and has chemical properties that support a dynamic role for MT in zinc trafficking. Consistent with these characteristics, changing the availability of zinc for cells and tissues causes rapid alteration of zinc bound to MT. Nevertheless, zinc trafficking occurs in metallothionein-null animals and cells, hypothetically making use of proteomic binding sites to mediate the intracellular movements of zinc. Like metallothionein, the proteome contains a large concentration of proteins that strongly coordinate zinc. In this environment, free Zn2+ may be of little significance. Instead, this review sets forth the basis for the hypothesis that components of the proteome and MT jointly provide the platform for zinc trafficking.
Collapse
Affiliation(s)
- David H Petering
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53217, USA.
| | - Afsana Mahim
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53217, USA.
| |
Collapse
|
55
|
The Functions of Metamorphic Metallothioneins in Zinc and Copper Metabolism. Int J Mol Sci 2017; 18:ijms18061237. [PMID: 28598392 PMCID: PMC5486060 DOI: 10.3390/ijms18061237] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/02/2017] [Accepted: 06/03/2017] [Indexed: 12/15/2022] Open
Abstract
Recent discoveries in zinc biology provide a new platform for discussing the primary physiological functions of mammalian metallothioneins (MTs) and their exquisite zinc-dependent regulation. It is now understood that the control of cellular zinc homeostasis includes buffering of Zn2+ ions at picomolar concentrations, extensive subcellular re-distribution of Zn2+, the loading of exocytotic vesicles with zinc species, and the control of Zn2+ ion signalling. In parallel, characteristic features of human MTs became known: their graded affinities for Zn2+ and the redox activity of their thiolate coordination environments. Unlike the single species that structural models of mammalian MTs describe with a set of seven divalent or eight to twelve monovalent metal ions, MTs are metamorphic. In vivo, they exist as many species differing in redox state and load with different metal ions. The functions of mammalian MTs should no longer be considered elusive or enigmatic because it is now evident that the reactivity and coordination dynamics of MTs with Zn2+ and Cu+ match the biological requirements for controlling—binding and delivering—these cellular metal ions, thus completing a 60-year search for their functions. MT represents a unique biological principle for buffering the most competitive essential metal ions Zn2+ and Cu+. How this knowledge translates to the function of other families of MTs awaits further insights into the specifics of how their properties relate to zinc and copper metabolism in other organisms.
Collapse
|
56
|
Mammalian Metallothionein-3: New Functional and Structural Insights. Int J Mol Sci 2017; 18:ijms18061117. [PMID: 28538697 PMCID: PMC5485941 DOI: 10.3390/ijms18061117] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/25/2022] Open
Abstract
Metallothionein-3 (MT-3), a member of the mammalian metallothionein (MT) family, is mainly expressed in the central nervous system (CNS). MT-3 possesses a unique neuronal growth inhibitory activity, and the levels of this intra- and extracellularly occurring metalloprotein are markedly diminished in the brain of patients affected by a number of metal-linked neurodegenerative disorders, including Alzheimer’s disease (AD). In these pathologies, the redox cycling of copper, accompanied by the production of reactive oxygen species (ROS), plays a key role in the neuronal toxicity. Although MT-3 shares the metal-thiolate clusters with the well-characterized MT-1 and MT-2, it shows distinct biological, structural and chemical properties. Owing to its anti-oxidant properties and modulator function not only for Zn, but also for Cu in the extra- and intracellular space, MT-3, but not MT-1/MT-2, protects neuronal cells from the toxicity of various Cu(II)-bound amyloids. In recent years, the roles of zinc dynamics and MT-3 function in neurodegeneration are slowly emerging. This short review focuses on the recent developments regarding the chemistry and biology of MT-3.
Collapse
|
57
|
Irvine GW, Stillman MJ. Residue Modification and Mass Spectrometry for the Investigation of Structural and Metalation Properties of Metallothionein and Cysteine-Rich Proteins. Int J Mol Sci 2017; 18:ijms18050913. [PMID: 28445428 PMCID: PMC5454826 DOI: 10.3390/ijms18050913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/23/2022] Open
Abstract
Structural information regarding metallothioneins (MTs) has been hard to come by due to its highly dynamic nature in the absence of metal-thiolate cluster formation and crystallization difficulties. Thus, typical spectroscopic methods for structural determination are limited in their usefulness when applied to MTs. Mass spectrometric methods have revolutionized our understanding of protein dynamics, structure, and folding. Recently, advances have been made in residue modification mass spectrometry in order to probe the hard-to-characterize structure of apo- and partially metalated MTs. By using different cysteine specific alkylation reagents, time dependent electrospray ionization mass spectrometry (ESI-MS), and step-wise “snapshot” ESI-MS, we are beginning to understand the dynamics of the conformers of apo-MT and related species. In this review we highlight recent papers that use these and similar techniques for structure elucidation and attempt to explain in a concise manner the data interpretations of these complex methods. We expect increasing resolution in our picture of the structural conformations of metal-free MTs as these techniques are more widely adopted and combined with other promising tools for structural elucidation.
Collapse
Affiliation(s)
- Gordon W Irvine
- Department of Chemistry, The University of Western Ontario, London, ON N6A 3K7, Canada.
| | - Martin J Stillman
- Department of Chemistry, The University of Western Ontario, London, ON N6A 3K7, Canada.
| |
Collapse
|
58
|
Rahman MT, Haque N, Abu Kasim NH, De Ley M. Origin, Function, and Fate of Metallothionein in Human Blood. Rev Physiol Biochem Pharmacol 2017; 173:41-62. [PMID: 28417197 DOI: 10.1007/112_2017_1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Toxic heavy metals, toxic organic compounds, reactive oxygen species (ROS), infections, and temperature are well-known metallothionein (MT) inducers in human blood. The current review aims to summarize synthesis, function, and fate of human blood MT in response to the known MT inducers. Part of the MTs that are synthesized in different organs such as the liver, kidney, and spleen is transported and stored in different blood cells and in plasma. Cells of the circulatory system also synthesize MT. From the circulation, MT returns to the kidney where the metal-bound MTs are degraded to release the metal ion that in turn induces MT expression therein. The blood MTs play important roles in metal detoxification, transportation, and storage. By neutralizing ROS, MTs protect blood cells from oxidative stress-induced cytotoxicity and genotoxicity. Arguably, MTs are also involved in immune suppression. Given the permeating distribution of blood MT throughout the body as well as its diverse role in the protection against harmful environmental factors and in metal homeostasis, MT could be better recognized as a major public health protein.
Collapse
Affiliation(s)
| | - Nazmul Haque
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
- Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
- Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Marc De Ley
- Laboratorium voor Biochemie, KU Leuven, Celestijnenlaan 200G, Postbus 2413, Heverlee, 3001, Leuven, Belgium
| |
Collapse
|
59
|
Silvestri S, Orlando P, Brugè F, Falcioni G, Tiano L. Effect of different metals on oxidative state and mitochondrial membrane potential in trout erythrocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2016; 134P1:280-285. [PMID: 27566895 DOI: 10.1016/j.ecoenv.2016.07.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 06/06/2023]
Abstract
Homeostasis of metal ions is critical for life and excessive exposure can promote cellular damage that could be due to oxidative damage. In this context we evaluated the effects of three different elements (copper, zinc and aluminum) on oxidative stress and mitochondrial functionality in nucleated trout erythrocytes (Oncorhynchus mykiss). Flowcytometric measurements using MitoProbe and DCFDA-H2 as fluorescent probes, indicated that redox active copper was able to influence all the biological parameters considered while redox inert, zinc and aluminum, show no significant effects. Toxicity of Al and Zn represent a debated argument and their ability to interact with other endogenous metal ions/metal binding proteins could play a role modulating their cellular toxicity.
Collapse
Affiliation(s)
- Sonia Silvestri
- Department of Clinical Dental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Patrick Orlando
- Department of Clinical Dental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Francesca Brugè
- Department of Clinical Dental Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Luca Tiano
- Department of Clinical Dental Sciences, Polytechnic University of Marche, Ancona, Italy.
| |
Collapse
|
60
|
Quinta-Ferreira ME, Sampaio Dos Aidos FDS, Matias CM, Mendes PJ, Dionísio JC, Santos RM, Rosário LM, Quinta-Ferreira RM. Modelling zinc changes at the hippocampal mossy fiber synaptic cleft. J Comput Neurosci 2016; 41:323-337. [PMID: 27696002 DOI: 10.1007/s10827-016-0620-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/11/2016] [Accepted: 08/15/2016] [Indexed: 01/18/2023]
Abstract
Zinc, a transition metal existing in very high concentrations in the hippocampal mossy fibers from CA3 area, is assumed to be co-released with glutamate and to have a neuromodulatory role at the corresponding synapses. The synaptic action of zinc is determined both by the spatiotemporal characteristics of the zinc release process and by the kinetics of zinc binding to sites located in the cleft area, as well as by their concentrations. This work addresses total, free and complexed zinc concentration changes, in an individual synaptic cleft, following single, short and long periods of evoked zinc release. The results estimate the magnitude and time course of the concentrations of zinc complexes, assuming that the dynamics of the release processes are similar to those of glutamate. It is also considered that, for the cleft zinc concentrations used in the model (≤ 1 μM), there is no postsynaptic zinc entry. For this reason, all released zinc ends up being reuptaken in a process that is several orders of magnitude slower than that of release and has thus a much smaller amplitude. The time derivative of the total zinc concentration in the cleft is represented by the difference between two alpha functions, corresponding to the released and uptaken components. These include specific parameters that were chosen assuming zinc and glutamate co-release, with similar time courses. The peak amplitudes of free zinc in the cleft were selected based on previously reported experimental cleft zinc concentration changes evoked by single and multiple stimulation protocols. The results suggest that following a low amount of zinc release, similar to that associated with one or a few stimuli, zinc clearance is mainly mediated by zinc binding to the high-affinity sites on the NMDA receptors and to the low-affinity sites on the highly abundant GLAST glutamate transporters. In the case of higher zinc release brought about by a larger group of stimuli, most zinc binding occurs essentially to the GLAST transporters, having the corresponding zinc complex a maximum concentration that is more than one order of magnitude larger than that for the high and low affinity NMDA sites. The other zinc complexes considered in the model, namely those formed with sites on the AMPA receptors, calcium and KATP channels and with ATP molecules, have much smaller contributions to the synaptic zinc clearance.
Collapse
Affiliation(s)
- M E Quinta-Ferreira
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, P-3004-504, Coimbra, Portugal.
- Department of Physics, University of Coimbra, P-3004-516, Coimbra, Portugal.
| | - F D S Sampaio Dos Aidos
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, P-3004-504, Coimbra, Portugal
- Department of Physics, University of Coimbra, P-3004-516, Coimbra, Portugal
- CFisUC, Department of Physics, University of Coimbra, P-3004-516, Coimbra, Portugal
| | - C M Matias
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, P-3004-504, Coimbra, Portugal
- UTAD- University of Trás-os-montes and Alto Douro, P-5000-801, Vila Real, Portugal
| | - P J Mendes
- Department of Physics, University of Coimbra, P-3004-516, Coimbra, Portugal
- LIP- Laboratory of Instrumentation and Experimental Particles Physics, P-3004-516, Coimbra, Portugal
| | - J C Dionísio
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, P-3004-504, Coimbra, Portugal
- Department of Animal Biology, University of Lisbon, P-1749-016, Lisbon, Portugal
| | - R M Santos
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, P-3004-504, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, P-3004-516, Coimbra, Portugal
| | - L M Rosário
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, P-3004-504, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, P-3004-516, Coimbra, Portugal
| | - R M Quinta-Ferreira
- CIEPQPF - Research Centre of Chemical Process Engineering and Forest Products, Department of Chemical Engineering, University of Coimbra, P-3030-790, Coimbra, Portugal
| |
Collapse
|
61
|
Rice JM, Zweifach A, Lynes MA. Metallothionein regulates intracellular zinc signaling during CD4(+) T cell activation. BMC Immunol 2016; 17:13. [PMID: 27251638 PMCID: PMC4890327 DOI: 10.1186/s12865-016-0151-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/23/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The ultra-low redox potential and zinc binding properties of the intracellular pool of mammalian metallothioneins (MT) suggest a role for MT in the transduction of redox signals into intracellular zinc signals. Increased expression of MT after exposure to heavy metals, oxidative stress, or inflammatory cytokines leads to an increased intracellular redox-mobilizable zinc pool that can affect downstream zinc-sensitive signaling pathways. CD4(+) T helper cells are poised to be influenced by MT transduced zinc signaling because they produce intracellular reactive oxygen species following activation through the T cell receptor and are sensitive to small changes in intracellular [Zn(2+)]. RESULTS MT expression and intracellular [Zn(2+)] are both increased during primary activation and expansion of naïve CD4(+) T cells into the Tr1 phenotype in vitro. When Tr1 cells from wildtype mice are compared with congenic mice lacking functional Mt1 and Mt2 genes, the expression of intracellular MT is associated with a greater increase in intracellular [Zn(2+)] immediately following exposure to reactive oxygen species or upon restimulation through the T cell receptor. The release of Zn(2+) from MT is associated with a greater increase in p38 MAPK activation following restimulation and decreased p38 MAPK activation in MT knockout Tr1 cells can be rescued by increasing intracellular [Zn(2+)]. Additionally, IL-10 secretion is increased in MT knockout Tr1 cells compared with wildtype controls and this increase is prevented when the intracellular [Zn(2+)] is increased experimentally. CONCLUSIONS Differences in zinc signaling associated with MT expression appear to be a result of preferential oxidation of MT and concomitant release of Zn(2+). Although zinc is released from many proteins following oxidation, release is greater when the cell contains an intracellular pool of MT. By expressing MT in response to certain environmental conditions, CD4(+) T cells are able to more efficiently release intracellular zinc and regulate signaling pathways following stimulation. The link between MT expression and increased zinc signaling following activation represents an important immunomodulatory mechanism of MT and illuminates the complex role MT plays in shaping immune responses.
Collapse
Affiliation(s)
- James M Rice
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, Unit 3125, Storrs, CT, 06269, USA. .,Present address: Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave., Boston, 02115, MA, USA.
| | - Adam Zweifach
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, Unit 3125, Storrs, CT, 06269, USA
| | - Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, Unit 3125, Storrs, CT, 06269, USA
| |
Collapse
|
62
|
Bajor M, Zaręba-Kozioł M, Zhukova L, Goryca K, Poznański J, Wysłouch-Cieszyńska A. An Interplay of S-Nitrosylation and Metal Ion Binding for Astrocytic S100B Protein. PLoS One 2016; 11:e0154822. [PMID: 27159591 PMCID: PMC4861259 DOI: 10.1371/journal.pone.0154822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/19/2016] [Indexed: 02/07/2023] Open
Abstract
Mammalian S100B protein plays multiple important roles in cellular brain processes. The protein is a clinically used marker for several pathologies including brain injury, neurodegeneration and cancer. High levels of S100B released by astrocytes in Down syndrome patients are responsible for reduced neurogenesis of neural progenitor cells and induction of cell death in neurons. Despite increasing understanding of S100B biology, there are still many questions concerning the detailed molecular mechanisms that determine specific activities of S100B. Elevated overexpression of S100B protein is often synchronized with increased nitric oxide-related activity. In this work we show S100B is a target of exogenous S-nitrosylation in rat brain protein lysate and identify endogenous S-nitrosylation of S100B in a cellular model of astrocytes. Biochemical studies are presented indicating S-nitrosylation tunes the conformation of S100B and modulates its Ca2+ and Zn2+ binding properties. Our in vitro results suggest that the possibility of endogenous S-nitrosylation should be taken into account in the further studies of in vivo S100B protein activity, especially under conditions of increased NO-related activity.
Collapse
Affiliation(s)
- Małgorzata Bajor
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Centre for Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Monika Zaręba-Kozioł
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Liliya Zhukova
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof Goryca
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Jarosław Poznański
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
63
|
Chalcogen bonding interactions between reducible sulfur and selenium compounds and models of zinc finger proteins. J Inorg Biochem 2016; 157:94-103. [DOI: 10.1016/j.jinorgbio.2016.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 01/07/2016] [Accepted: 01/09/2016] [Indexed: 02/04/2023]
|
64
|
Abstract
Zinc is an important micronutrient, essential in the diet to avoid a variety of conditions associated with malnutrition such as diarrhoea and alopecia. Lowered circulating levels of zinc are also found in diabetes mellitus, a condition which affects one in twelve of the adult population and whose treatments consume approximately 10 % of healthcare budgets. Zn2+ ions are essential for a huge range of cellular functions and, in the specialised pancreatic β-cell, for the storage of insulin within the secretory granule. Correspondingly, genetic variants in the SLC30A8 gene, which encodes the diabetes-associated granule-resident Zn2+ transporter ZnT8, are associated with an altered risk of type 2 diabetes. Here, we focus on (i) recent advances in measuring free zinc concentrations dynamically in subcellular compartments, and (ii) studies dissecting the role of intracellular zinc in the control of glucose homeostasis in vitro and in vivo. We discuss the effects on insulin secretion and action of deleting or over-expressing Slc30a8 highly selectively in the pancreatic β-cell, and the role of zinc in insulin signalling. While modulated by genetic variability, healthy levels of dietary zinc, and hence normal cellular zinc homeostasis, are likely to play an important role in the proper release and action of insulin to maintain glucose homeostasis and lower diabetes risk.
Collapse
|
65
|
Watson CY, Molina RM, Louzada A, Murdaugh KM, Donaghey TC, Brain JD. Effects of zinc oxide nanoparticles on Kupffer cell phagosomal motility, bacterial clearance, and liver function. Int J Nanomedicine 2015; 10:4173-84. [PMID: 26170657 PMCID: PMC4492628 DOI: 10.2147/ijn.s82807] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Zinc oxide engineered nanoparticles (ZnO ENPs) have potential as nanomedicines due to their inherent properties. Studies have described their pulmonary impact, but less is known about the consequences of ZnO ENP interactions with the liver. This study was designed to describe the effects of ZnO ENPs on the liver and Kupffer cells after intravenous (IV) administration. Materials and methods First, pharmacokinetic studies were conducted to determine the tissue distribution of neutron-activated 65ZnO ENPs post-IV injection in Wistar Han rats. Then, a noninvasive in vivo method to assess Kupffer cell phagosomal motility was employed using ferromagnetic iron particles and magnetometry. We also examined whether prior IV injection of ZnO ENPs altered Kupffer cell bactericidal activity on circulating Pseudomonas aeruginosa. Serum and liver tissues were collected to assess liver-injury biomarkers and histological changes, respectively. Results We found that the liver was the major site of initial uptake of 65ZnO ENPs. There was a time-dependent decrease in tissue levels of 65Zn in all organs examined, refecting particle dissolution. In vivo magnetometry showed a time-dependent and transient reduction in Kupffer cell phagosomal motility. Animals challenged with P. aeruginosa 24 hours post-ZnO ENP injection showed an initial (30 minutes) delay in vascular bacterial clearance. However, by 4 hours, IV-injected bacteria were cleared from the blood, liver, spleen, lungs, and kidneys. Seven days post-ZnO ENP injection, creatine phosphokinase and aspartate aminotransferase levels in serum were significantly increased. Histological evidence of hepatocyte damage and marginated neutrophils were observed in the liver. Conclusion Administration of ZnO ENPs transiently inhibited Kupffer cell phagosomal motility and later induced hepatocyte injury, but did not alter bacterial clearance from the blood or killing in the liver, spleen, lungs, or kidneys. Our data show that diminished Kupffer cell organelle motion correlated with ZnO ENP-induced liver injury.
Collapse
Affiliation(s)
- Christa Y Watson
- Center for Nanotechnology and Nanotoxicology, Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ramon M Molina
- Center for Nanotechnology and Nanotoxicology, Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andressa Louzada
- Center for Nanotechnology and Nanotoxicology, Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kimberly M Murdaugh
- Center for Nanotechnology and Nanotoxicology, Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Thomas C Donaghey
- Center for Nanotechnology and Nanotoxicology, Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph D Brain
- Center for Nanotechnology and Nanotoxicology, Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
66
|
Bellingham SA, Guo B, Hill AF. The secret life of extracellular vesicles in metal homeostasis and neurodegeneration. Biol Cell 2015; 107:389-418. [PMID: 26032945 DOI: 10.1111/boc.201500030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022]
Abstract
Biologically active metals such as copper, zinc and iron are fundamental for sustaining life in different organisms with the regulation of cellular metal homeostasis tightly controlled through proteins that coordinate metal uptake, efflux and detoxification. Many of the proteins involved in either uptake or efflux of metals are localised and function on the plasma membrane, traffic between intracellular compartments depending upon the cellular metal environment and can undergo recycling via the endosomal pathway. The biogenesis of exosomes also occurs within the endosomal system, with several major neurodegenerative disease proteins shown to be released in association with these vesicles, including the amyloid-β (Aβ) peptide in Alzheimer's disease and the infectious prion protein involved in Prion diseases. Aβ peptide and the prion protein also bind biologically active metals and are postulated to play important roles in metal homeostasis. In this review, we will discuss the role of extracellular vesicles in Alzheimer's and Prion diseases and explore their potential contribution to metal homeostasis.
Collapse
Affiliation(s)
- Shayne A Bellingham
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Belinda Guo
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
67
|
Relationship between Zinc (Zn (2+) ) and Glutamate Receptors in the Processes Underlying Neurodegeneration. Neural Plast 2015; 2015:591563. [PMID: 26106488 PMCID: PMC4461779 DOI: 10.1155/2015/591563] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/13/2015] [Indexed: 12/25/2022] Open
Abstract
The results from numerous studies have shown that an imbalance between particular neurotransmitters may lead to brain circuit dysfunction and development of many pathological states. The significance of glutamate pathways for the functioning of the nervous system is equivocal. On the one hand, glutamate transmission is necessary for neuroplasticity, synaptogenesis, or cell survival, but on the other hand an excessive and long-lasting increased level of glutamate in the synapse may lead to cell death. Under clinical conditions, hyperactivity of the glutamate system is associated with ischemia, epilepsy, and neurodegenerative diseases such as Alzheimer's, Huntington's, and many others. The achievement of glutamate activity in the physiological range requires efficient control by endogenous regulatory factors. Due to the fact that the free pool of ion Zn(2+) is a cotransmitter in some glutamate neurons; the role of this element in the pathophysiology of a neurodegenerative diseases has been intensively studied. There is a lot of evidence for Zn(2+) dyshomeostasis and glutamate system abnormalities in ischemic and neurodegenerative disorders. However, the precise interaction between Zn(2+) regulative function and the glutamate system is still not fully understood. This review describes the relationship between Zn(2+) and glutamate dependent signaling pathways under selected pathological central nervous system (CNS) conditions.
Collapse
|
68
|
Sreenath K, Yuan Z, Allen JR, Davidson MW, Zhu L. A fluorescent indicator for imaging lysosomal zinc(II) with Förster resonance energy transfer (FRET)-enhanced photostability and a narrow band of emission. Chemistry 2015; 21:867-74. [PMID: 25382395 PMCID: PMC4294628 DOI: 10.1002/chem.201403479] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Indexed: 12/29/2022]
Abstract
We demonstrate a strategy to transfer the zinc(II) sensitivity of a fluoroionophore with low photostability and a broad emission band to a bright and photostable fluorophore with a narrow emission band. The two fluorophores are covalently connected to afford an intramolecular Förster resonance energy transfer (FRET) conjugate. The FRET donor in the conjugate is a zinc(II)-sensitive arylvinylbipyridyl fluoroionophore, the absorption and emission of which undergo bathochromic shifts upon zinc(II) coordination. When the FRET donor is excited, efficient intramolecular energy transfer occurs to result in the emission of the acceptor boron dipyrromethene (4,4-difluoro-4-bora-3a,4a-diaza-s-indacene or BODIPY) as a function of zinc(II) concentration. The broad emission band of the donor/zinc(II) complex is transformed into the strong, narrow emission band of the BODIPY acceptor in the FRET conjugates, which can be captured within the narrow emission window that is preferred for multicolor imaging experiments. In addition to competing with other nonradiative decay processes of the FRET donor, the rapid intramolecular FRET of the excited FRET-conjugate molecule protects the donor fluorophore from photobleaching, thus enhancing the photostability of the indicator. FRET conjugates 3 and 4 contain aliphatic amino groups, which selectively target lysosomes in mammalian cells. This subcellular localization preference was verified by using confocal fluorescence microscopy, which also shows the zinc(II)-enhanced emission of 3 and 4 in lysosomes. It was further shown using two-color structured illumination microscopy (SIM), which is capable of extending the lateral resolution over the Abbe diffraction limit by a factor of two, that the morpholino-functionalized compound 4 localizes in the interior of lysosomes, rather than anchoring on the lysosomal membranes, of live HeLa cells.
Collapse
Affiliation(s)
- Kesavapillai Sreenath
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390 (USA)
| | - Zhao Yuan
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390 (USA)
| | - John R. Allen
- National High Magnetic Field Laboratory and Department of Biological Sciences, Florida State University, 1800 East Paul Dirac Drive, Tallahassee, FL 32310 (USA)
| | - Michael W. Davidson
- National High Magnetic Field Laboratory and Department of Biological Sciences, Florida State University, 1800 East Paul Dirac Drive, Tallahassee, FL 32310 (USA)
| | - Lei Zhu
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390 (USA)
| |
Collapse
|
69
|
Kochańczyk T, Drozd A, Krężel A. Relationship between the architecture of zinc coordination and zinc binding affinity in proteins – insights into zinc regulation. Metallomics 2015; 7:244-57. [DOI: 10.1039/c4mt00094c] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Relationship between the architecture and stability of zinc proteins.
Collapse
Affiliation(s)
- Tomasz Kochańczyk
- Laboratory of Chemical Biology
- Faculty of Biotechnology
- University of Wrocław
- 50-383 Wrocław, Poland
| | - Agnieszka Drozd
- Laboratory of Chemical Biology
- Faculty of Biotechnology
- University of Wrocław
- 50-383 Wrocław, Poland
| | - Artur Krężel
- Laboratory of Chemical Biology
- Faculty of Biotechnology
- University of Wrocław
- 50-383 Wrocław, Poland
| |
Collapse
|
70
|
Hanagata N, Morita H. Calcium ions rescue human lung epithelial cells from the toxicity of zinc oxide nanoparticles. J Toxicol Sci 2015; 40:625-35. [DOI: 10.2131/jts.40.625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Nobutaka Hanagata
- Nanotechnology Innovation Station, National Institute for Materials Science
- Graduate School of Life Science, Hokkaido University
| | - Hiromi Morita
- Nanotechnology Innovation Station, National Institute for Materials Science
| |
Collapse
|
71
|
Chen Y, Bai Y, Han Z, He W, Guo Z. Photoluminescence imaging of Zn2+in living systems. Chem Soc Rev 2015; 44:4517-46. [DOI: 10.1039/c5cs00005j] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Advances in PL imaging techniques, such as confocal microscopy, two photon microscopy, lifetime and optical imaging techniques, have made remarkable contributions in Zn2+tracking.
Collapse
Affiliation(s)
- Yuncong Chen
- State Key Laboratory of Coordination Chemistry
- Coordination Chemistry Institute
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
| | - Yang Bai
- State Key Laboratory of Coordination Chemistry
- Coordination Chemistry Institute
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
| | - Zhong Han
- State Key Laboratory of Coordination Chemistry
- Coordination Chemistry Institute
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry
- Coordination Chemistry Institute
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry
- Coordination Chemistry Institute
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210093
| |
Collapse
|
72
|
Sreenath K, Yuan Z, Allen JR, Davidson MW, Zhu L. A Fluorescent Indicator for Imaging Lysosomal Zinc(II) with Förster Resonance Energy Transfer (FRET)-Enhanced Photostability and a Narrow Band of Emission. Chemistry 2014; 21:4163-4163. [PMID: 25378058 DOI: 10.1002/chem.403479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Indexed: 01/05/2023]
Abstract
We demonstrate a strategy to transfer the zinc(II) sensitivity of a fluoroionophore with low photostability and a broad emission band to a bright and photostable fluorophore with a narrow emission band. The two fluorophores are covalently connected to afford an intramolecular Förster resonance energy transfer (FRET) conjugate. The FRET donor in the conjugate is a zinc(II)-sensitive arylvinylbipyridyl fluoroionophore, the absorption and emission of which undergo bathochromic shifts upon zinc(II) coordination. When the FRET donor is excited, efficient intramolecular energy transfer occurs to result in the emission of the acceptor boron dipyrromethene (4,4-difluoro-4-bora-3a,4a-diaza-s-indacene or BODIPY) as a function of zinc(II) concentration. The broad emission band of the donor/zinc(II) complex is transformed into the strong, narrow emission band of the BODIPY acceptor in the FRET conjugates, which can be captured within the narrow emission window that is preferred for multicolor imaging experiments. In addition to competing with other nonradiative decay processes of the FRET donor, the rapid intramolecular FRET of the excited FRET-conjugate molecule protects the donor fluorophore from photobleaching, thus enhancing the photostability of the indicator. FRET conjugates 3 and 4 contain aliphatic amino groups, which selectively target lysosomes in mammalian cells. This subcellular localization preference was verified by using confocal fluorescence microscopy, which also shows the zinc(II)-enhanced emission of 3 and 4 in lysosomes. It was further shown using two-color structured illumination microscopy (SIM), which is capable of extending the lateral resolution over the Abbe diffraction limit by a factor of two, that the morpholino-functionalized compound 4 localizes in the interior of lysosomes, rather than anchoring on the lysosomal membranes, of live HeLa cells.
Collapse
Affiliation(s)
- Kesavapillai Sreenath
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390 (USA); Present Address: Department of Chemistry, VTM NSS College, Dhanuvachapuram, Kerala, 695 503 (India)
| | | | | | | | | |
Collapse
|
73
|
|
74
|
Gonzalez-Iglesias H, Alvarez L, García M, Petrash C, Sanz-Medel A, Coca-Prados M. Metallothioneins (MTs) in the human eye: a perspective article on the zinc-MT redox cycle. Metallomics 2014; 6:201-8. [PMID: 24419560 DOI: 10.1039/c3mt00298e] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metallothioneins (MTs) are zinc-ion-binding proteins with a wide range of functions, among which are neuroprotection, maintenance of cellular zinc homeostasis, and defense against oxidative damage and inflammation. The human eye is enriched in MTs, and multiple isoforms may contribute to distinct antioxidant defense mechanisms in various ocular tissues. Zinc is a main regulator of MT gene and protein expression, and we recently applied bioanalytical techniques to address key questions on its relationship with MTs, including the stoichiometry of zinc-MT, the fate of zinc tracers ((nat)Zn and (68)Zn) in MTs during activation by exogenous zinc and cytokines, and the concentration of MTs in human ocular cells. We found that exogenously introduced zinc induced a potent de novo synthesis of MTs as well as a strong inhibition of pro-inflammatory cytokines. Zinc and cytokines also promote a stoichiometric transition of the MT complex from Zn6Cu1-MT to Zn7-MT, suggesting that MTs may interact more effectively with reactive oxygen species to decrease potential oxidative damage. Levels of MTs decrease with aging and disease, which may result in zinc release that is potentially cytotoxic. This state is also observed with increased oxidative stress and inflammation, suggesting that the antioxidant function of MTs has been impaired. In this review we propose a working model of the "zinc-metallothionein redox cycle" to regenerate and enhance the antioxidant function of MTs with the aim of combating the progression of these disease states.
Collapse
Affiliation(s)
- Héctor Gonzalez-Iglesias
- Fundación de Investigación Oftalmológica, Instituto Oftalmológico Fernández-Vega, Avda. Dres. Fernandez-Vega, 34, 33012, Oviedo, Spain.
| | | | | | | | | | | |
Collapse
|
75
|
Chatel A, Faucet-Marquis V, Pfohl-Leszkowicz A, Gourlay-France C, Vincent-Hubert F. DNA adduct formation and induction of detoxification mechanisms in Dreissena polymorpha exposed to nitro-PAHs. Mutagenesis 2014; 29:457-65. [DOI: 10.1093/mutage/geu040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
76
|
Abstract
Mammalian metallothioneins (MTs) comprise a Zn3Cys9 cluster in the β domain and a Zn4Cys11 cluster in the α domain. They play a crucial role in storing and donating Zn(2+) ions to target metalloproteins and have been implicated in several diseases, thus understanding how MTs release Zn(2+) is of widespread interest. In this work, we present a strategy to compute the free energy for releasing Zn(2+) from MTs using a combination of classical molecular dynamics (MD) simulations, quantum-mechanics/molecular-mechanics (QM/MM) minimizations, and continuum dielectric calculations. The methodology is shown to reproduce the experimental observations that (1) the Zn-binding sites do not have equal Zn(2+) affinity and (2) the isolated β domain is thermodynamically less stable and releases Zn(2+) faster with oxidizing agents than the isolated α domain. It was used to compute the free energies for Zn(2+) release from the metal cluster in the absence and presence of the protein matrix (protein architecture and coupled protein-water interactions) to yield the respective disulfide-bonded product. The results show the importance of the protein matrix as well as protein dynamics and coupled conformational changes in accounting for the differential Zn(2+)-releasing propensity of the two domains with oxidizing agents.
Collapse
Affiliation(s)
- C Satheesan Babu
- Institute of Biomedical Sciences, Academia Sinica , Taipei 115, Taiwan , R.O.C
| | | | | | | |
Collapse
|
77
|
Cortese-Krott MM, Kulakov L, Opländer C, Kolb-Bachofen V, Kröncke KD, Suschek CV. Zinc regulates iNOS-derived nitric oxide formation in endothelial cells. Redox Biol 2014; 2:945-54. [PMID: 25180171 PMCID: PMC4143817 DOI: 10.1016/j.redox.2014.06.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 06/26/2014] [Indexed: 01/07/2023] Open
Abstract
Aberrant production of nitric oxide (NO) by inducible NO synthase (iNOS) has been implicated in the pathogenesis of endothelial dysfunction and vascular disease. Mechanisms responsible for the fine-tuning of iNOS activity in inflammation are still not fully understood. Zinc is an important structural element of NOS enzymes and is known to inhibit its catalytical activity. In this study we aimed to investigate the effects of zinc on iNOS activity and expression in endothelial cells. We found that zinc down-regulated the expression of iNOS (mRNA+protein) and decreased cytokine-mediated activation of the iNOS promoter. Zinc-mediated regulation of iNOS expression was due to inhibition of NF-κB transactivation activity, as determined by a decrease in both NF-κB-driven luciferase reporter activity and expression of NF-κB target genes, including cyclooxygenase 2 and IL-1β. However, zinc did not affect NF-κB translocation into the nucleus, as assessed by Western blot analysis of nuclear and cytoplasmic fractions. Taken together our results demonstrate that zinc limits iNOS-derived high output NO production in endothelial cells by inhibiting NF-κB-dependent iNOS expression, pointing to a role of zinc as a regulator of iNOS activity in inflammation. Zinc inhibits iNOS-dependent nitrite accumulation in endothelial cells. Zinc decreases cytokine-induced iNOS expression in endothelial cells. Zinc inhibits iNOS promoter activity. NF-kB silencing abolishes cytokine-induced iNOS expression. Zinc inhibits the transactivation activity of NF-κB.
Collapse
Affiliation(s)
- Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology, and Angiology, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany ; Research Group Immunobiology, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany
| | - Larissa Kulakov
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology, and Angiology, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany ; Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany
| | - Christian Opländer
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany
| | - Victoria Kolb-Bachofen
- Research Group Immunobiology, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany
| | - Klaus-D Kröncke
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany
| | - Christoph V Suschek
- Research Group Immunobiology, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany ; Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Universitätsstr. 1, Düsseldorf D-40225, Germany
| |
Collapse
|
78
|
Dagbay K, Eron SJ, Serrano BP, Velázquez-Delgado EM, Zhao Y, Lin D, Vaidya S, Hardy JA. A multipronged approach for compiling a global map of allosteric regulation in the apoptotic caspases. Methods Enzymol 2014; 544:215-49. [PMID: 24974292 DOI: 10.1016/b978-0-12-417158-9.00009-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the most promising and as yet underutilized means of regulating protein function is exploitation of allosteric sites. All caspases catalyze the same overall reaction, but they perform different biological roles and are differentially regulated. It is our hypothesis that many allosteric sites exist on various caspases and that understanding both the distinct and overlapping mechanisms by which each caspase can be allosterically controlled should ultimately enable caspase-specific inhibition. Here we describe the ongoing work and methods for compiling a comprehensive map of apoptotic caspase allostery. Central to this approach are the use of (i) the embedded record of naturally evolved allosteric sites that are sensitive to zinc-mediated inhibition, phosphorylation, and other posttranslational modifications, (ii) structural and mutagenic approaches, and (iii) novel binding sites identified by both rationally-designed and screening-derived small-molecule inhibitors.
Collapse
Affiliation(s)
- Kevin Dagbay
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts, USA
| | - Scott J Eron
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts, USA
| | - Banyuhay P Serrano
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts, USA
| | | | - Yunlong Zhao
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts, USA
| | - Di Lin
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts, USA
| | - Sravanti Vaidya
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts, USA
| | - Jeanne A Hardy
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts, USA.
| |
Collapse
|
79
|
Zhu L, Yuan Z, Simmons JT, Sreenath K. Zn(II)-coordination modulated ligand photophysical processes - the development of fluorescent indicators for imaging biological Zn(II) ions. RSC Adv 2014; 4:20398-20440. [PMID: 25071933 PMCID: PMC4111279 DOI: 10.1039/c4ra00354c] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Molecular photophysics and metal coordination chemistry are the two fundamental pillars that support the development of fluorescent cation indicators. In this article, we describe how Zn(II)-coordination alters various ligand-centered photophysical processes that are pertinent to developing Zn(II) indicators. The main aim is to show how small organic Zn(II) indicators work under the constraints of specific requirements, including Zn(II) detection range, photophysical requirements such as excitation energy and emission color, temporal and spatial resolutions in a heterogeneous intracellular environment, and fluorescence response selectivity between similar cations such as Zn(II) and Cd(II). In the last section, the biological questions that fluorescent Zn(II) indicators help to answer are described, which have been motivating and challenging this field of research.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390, United States
| | - Zhao Yuan
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390, United States
| | - J. Tyler Simmons
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390, United States
| | - Kesavapillai Sreenath
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way, Tallahassee, FL 32306-4390, United States
| |
Collapse
|
80
|
James SA, Feltis BN, de Jonge MD, Sridhar M, Kimpton JA, Altissimo M, Mayo S, Zheng C, Hastings A, Howard DL, Paterson DJ, Wright PFA, Moorhead GF, Turney TW, Fu J. Quantification of ZnO nanoparticle uptake, distribution, and dissolution within individual human macrophages. ACS NANO 2013; 7:10621-35. [PMID: 24187959 DOI: 10.1021/nn403118u] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The usefulness of zinc oxide (ZnO) nanoparticles has led to their wide distribution in consumer products, despite only a limited understanding of how this nanomaterial behaves within biological systems. From a nanotoxicological viewpoint the interaction(s) of ZnO nanoparticles with cells of the immune system is of specific interest, as these nanostructures are readily phagocytosed. In this study, rapid scanning X-ray fluorescence microscopy was used to assay the number ZnO nanoparticles associated with ∼1000 individual THP-1 monocyte-derived human macrophages. These data showed that nanoparticle-treated cells endured a 400% elevation in total Zn levels, 13-fold greater than the increase observed when incubated in the presence of an equitoxic concentration of ZnCl2. Even after excluding the contribution of internalized nanoparticles, Zn levels in nanoparticle treated cells were raised ∼200% above basal levels. As dissolution of ZnO nanoparticles is critical to their cytotoxic response, we utilized a strategy combining ion beam milling, X-ray fluorescence and scanning electron microscopy to directly probe the distribution and composition of ZnO nanoparticles throughout the cellular interior. This study demonstrated that correlative photon and ion beam imaging techniques can provide both high-resolution and statistically powerful information on the biology of metal oxide nanoparticles at the single-cell level. Our approach promises ready application to broader studies of phenomena at the interface of nanotechnology and biology.
Collapse
Affiliation(s)
- Simon A James
- Australian Synchrotron , Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Wu W, Bromberg PA, Samet JM. Zinc ions as effectors of environmental oxidative lung injury. Free Radic Biol Med 2013; 65:57-69. [PMID: 23747928 DOI: 10.1016/j.freeradbiomed.2013.05.048] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 04/29/2013] [Accepted: 05/31/2013] [Indexed: 12/30/2022]
Abstract
The redox-inert transition metal Zn is a micronutrient that plays essential roles in protein structure, catalysis, and regulation of function. Inhalational exposure to ZnO or to soluble Zn salts in occupational and environmental settings leads to adverse health effects, the severity of which appears dependent on the flux of Zn(2+) presented to the airway and alveolar cells. The cellular toxicity of exogenous Zn(2+) exposure is characterized by cellular responses that include mitochondrial dysfunction, elevated production of reactive oxygen species, and loss of signaling quiescence leading to cell death and increased expression of adaptive and inflammatory genes. Central to the molecular effects of Zn(2+) are its interactions with cysteinyl thiols, which alters their functionality by modulating their reactivity and participation in redox reactions. Ongoing studies aimed at elucidating the molecular toxicology of Zn(2+) in the lung are contributing valuable information about its role in redox biology and cellular homeostasis in normal and pathophysiology.
Collapse
Affiliation(s)
- Weidong Wu
- School of Public Health XinXiang Medical University XinXiang, China 453003; Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Philip A Bromberg
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - James M Samet
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, U.S. EPA, Chapel Hill, NC 27514, USA.
| |
Collapse
|
82
|
Antony S, Bayse CA. Density Functional Theory Study of the Attack of Ebselen on a Zinc-Finger Model. Inorg Chem 2013; 52:13803-5. [DOI: 10.1021/ic401429z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Sonia Antony
- Department of Chemistry and Biochemistry, Old Dominion University, Hampton Boulevard, Norfolk, Virginia 23529, United States
| | - Craig A. Bayse
- Department of Chemistry and Biochemistry, Old Dominion University, Hampton Boulevard, Norfolk, Virginia 23529, United States
| |
Collapse
|
83
|
Shen C, James SA, de Jonge MD, Turney TW, Wright PFA, Feltis BN. Relating Cytotoxicity, Zinc Ions, and Reactive Oxygen in ZnO Nanoparticle–Exposed Human Immune Cells. Toxicol Sci 2013; 136:120-30. [DOI: 10.1093/toxsci/kft187] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
84
|
Szewczyk B. Zinc homeostasis and neurodegenerative disorders. Front Aging Neurosci 2013; 5:33. [PMID: 23882214 PMCID: PMC3715721 DOI: 10.3389/fnagi.2013.00033] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/24/2013] [Indexed: 12/19/2022] Open
Abstract
Zinc is an essential trace element, whose importance to the function of the central nervous system (CNS) is increasingly being appreciated. Alterations in zinc dyshomeostasis has been suggested as a key factor in the development of several neuropsychiatric disorders. In the CNS, zinc occurs in two forms: the first being tightly bound to proteins and, secondly, the free, cytoplasmic, or extracellular form found in presynaptic vesicles. Under normal conditions, zinc released from the synaptic vesicles modulates both ionotropic and metabotropic post-synaptic receptors. While under clinical conditions such as traumatic brain injury, stroke or epilepsy, the excess influx of zinc into neurons has been found to result in neurotoxicity and damage to postsynaptic neurons. On the other hand, a growing body of evidence suggests that a deficiency, rather than an excess, of zinc leads to an increased risk for the development of neurological disorders. Indeed, zinc deficiency has been shown to affect neurogenesis and increase neuronal apoptosis, which can lead to learning and memory deficits. Altered zinc homeostasis is also suggested as a risk factor for depression, Alzheimer's disease (AD), aging, and other neurodegenerative disorders. Under normal CNS physiology, homeostatic controls are put in place to avoid the accumulation of excess zinc or its deficiency. This cellular zinc homeostasis results from the actions of a coordinated regulation effected by different proteins involved in the uptake, excretion and intracellular storage/trafficking of zinc. These proteins include membranous transporters (ZnT and Zip) and metallothioneins (MT) which control intracellular zinc levels. Interestingly, alterations in ZnT and MT have been recently reported in both aging and AD. This paper provides an overview of both clinical and experimental evidence that implicates a dysfunction in zinc homeostasis in the pathophysiology of depression, AD, and aging.
Collapse
Affiliation(s)
- Bernadeta Szewczyk
- Department of Neurobiology, Institute of Pharmacology Polish Academy of SciencesKrakow, Poland
| |
Collapse
|
85
|
Mocchegiani E, Romeo J, Malavolta M, Costarelli L, Giacconi R, Diaz LE, Marcos A. Zinc: dietary intake and impact of supplementation on immune function in elderly. AGE (DORDRECHT, NETHERLANDS) 2013; 35:839-60. [PMID: 22222917 PMCID: PMC3636409 DOI: 10.1007/s11357-011-9377-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 12/21/2011] [Indexed: 05/07/2023]
Abstract
The diet in the elderly does not provide a sufficient level of nutrients needed to maintain an adequate healthy status leading to micronutrient deficiencies and impaired immune response with subsequent development of degenerative diseases. Nutrient "zinc" is a relevant micronutrient involved in maintaining a good integrity of many body homeostatic mechanisms, including immune efficiency, owing to its requirement for the biological activity of many enzymes, proteins and for cellular proliferation and genomic stability. Old people aged 60-65 years and older have zinc intakes below 50% of the recommended daily allowance on a given day. Many causes can be involved: among them, altered intestinal absorption, inadequate mastication, psychosocial factors, drugs interactions, altered subcellular processes (zinc transporters (Zip and ZnT family), metallothioneins, divalent metal transporter-1). Zinc supplementation may remodel the immune alterations in elderly leading to healthy ageing. Several zinc trials have been carried out with contradictory data, perhaps due to incorrect choice of an effective zinc supplementation in old subjects showing subsequent zinc toxic effects on immunity. Old subjects with specific IL-6 polymorphism (GG allele carriers; named C-) are more prone for zinc supplementation than the entire old population, in whom correct dietary habits with foods containing zinc (Mediterranean diet) may be sufficient in restoring zinc deficiency and impaired immune response. We summarise the main causes of low zinc dietary intake in elderly reporting an update on the impact of zinc supplementation upon the immune response also on the basis of individual IL-6 polymorphism.
Collapse
Affiliation(s)
- Eugenio Mocchegiani
- Ctr. Nutrition and Ageing, Italian National Research Centres on Ageing (INRCA), Via Birarelli 8, 60121, Ancona, Italy.
| | | | | | | | | | | | | |
Collapse
|
86
|
Görg B, Schliess F, Häussinger D. Osmotic and oxidative/nitrosative stress in ammonia toxicity and hepatic encephalopathy. Arch Biochem Biophys 2013; 536:158-63. [PMID: 23567841 DOI: 10.1016/j.abb.2013.03.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/14/2013] [Accepted: 03/16/2013] [Indexed: 02/06/2023]
Abstract
Hepatic encephalopathy (HE) is a neuropsychiatric complication of acute or chronic liver failure. Currently, HE in cirrhotic patients is seen as a clinical manifestation of a low grade cerebral edema which exacerbates in response to a variety of precipitating factors after an ammonia-induced exhaustion of the volume-regulatory capacity of the astrocyte. Astrocyte swelling triggers a complex signaling cascade which relies on NMDA receptor activation, elevation of intracellular Ca(2+) concentration and prostanoid-driven glutamate exocytosis, which result in increased formation of reactive nitrogen and oxygen species (RNOS) through activation of NADPH oxidase and nitric oxide synthase. Since RNOS in turn promote astrocyte swelling, a self-amplifying signaling loop between osmotic- and oxidative stress ensues, which triggers a variety of downstream consequences. These include protein tyrosine nitration (PTN), oxidation of RNA, mobilization of zinc, alterations in intra- and intercellular signaling and multiple effects on gene transcription. Whereas PTN can affect the function of a variety of proteins, such as glutamine synthetase, oxidized RNA may affect local protein synthesis at synapses, thereby potentially interfering with protein synthesis-dependent memory formation. PTN and RNA oxidation are also found in post mortem human cerebral cortex of cirrhotic patients with HE but not in those without HE, thereby confirming a role for oxidative stress in the pathophysiology of HE. Evidence derived from animal experiments and human post mortem brain tissue also indicates an up-regulation of microglia activation markers in the absence of increased synthesis of pro-inflammatory cytokines. However, the role of activated microglia in the pathophysiology of HE needs to be worked out in more detail. Most recent observations made in whole genome micro-array analyses of post mortem human brain tissue point to a hitherto unrecognized activation of multiple anti-inflammatory signaling pathways.
Collapse
Affiliation(s)
- Boris Görg
- Heinrich-Heine-University Düsseldorf, Clinic for Gastroenterology, Hepatology, and Infectious Diseases, Germany
| | | | | |
Collapse
|
87
|
Abstract
After iron, zinc is the most abundant essential trace metal. Intracellular zinc ([Zn]i) is maintained across a wide range of cells and species in a tight quota (100 to 500 μM) by a dynamic process of transport, intracellular vesicular storage, and binding to a large number of proteins (estimated at 3-10% of human proteome). As such, zinc is an integral component of numerous metalloenzymes, structural proteins, and transcription factors. It is generally assumed that a vanishingly small component of [Zn]i, referred to as free or labile zinc, and operationally defined as the pool sensitive to chelation (by agents such as N, N, N’, N’-tetrakis [2-pyridylmethyl] ethylenediamine [TPEN]) and capable of detection by a variety of chemical and genetic sensors, participates in signal transduction pathways. Zinc deficiencies, per se, can arise from acquired (malnutrition, alcoholism) or genetic (mutations in molecules affecting zinc homeostasis, the informative and first example being acrodermatitis enteropathica) factors or as a component of various diseases (e.g., sickle cell disease, cystic fibrosis, sepsis). Hypozincemia has profound effects on developing humans, and all facets of physiological function (neuronal, endocrine, immunological) are affected, although considerably less is known regarding cardiovascular pathophysiology. In this review, we provide an update on current knowledge of molecular and cellular aspects of zinc homeostasis and then focus on implications of zinc signaling in pulmonary endothelium as it relates to programmed cell death, altered contractility, and septic and aseptic injury to this segment of the lung.
Collapse
Affiliation(s)
- Kalidasan Thambiayya
- Department of Bioengineering, University of Pittsburgh and University of Pittsburgh School of Medicine and Graduate School Public Health, Pittsburgh, Pennsylvania, USA
| | | | | | | |
Collapse
|
88
|
Oxidation reactivity of zinc–cysteine clusters in metallothionein. J Biol Inorg Chem 2013; 18:333-42. [DOI: 10.1007/s00775-013-0977-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 01/06/2013] [Indexed: 12/24/2022]
|
89
|
Blindauer CA. Lessons on the critical interplay between zinc binding and protein structure and dynamics. J Inorg Biochem 2013; 121:145-55. [PMID: 23376625 DOI: 10.1016/j.jinorgbio.2013.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 01/08/2013] [Accepted: 01/08/2013] [Indexed: 02/05/2023]
Abstract
Zinc is one of the most important micronutrients for virtually all living organisms, and hence, it is important to understand the molecular mechanisms for its homeostasis. Besides proteins involved in transmembrane transport, both extra- and intracellular zinc-binding proteins play important roles in the respective metabolic networks. Important examples for extracellular zinc transporters are mammalian serum albumins, and for intracellular zinc handling, certain metallothioneins are of relevance. The availability of protein structures including relevant metal binding sites is a fundamental prerequisite to decipher the mechanisms that govern zinc binding dynamics in these proteins, but their determination can prove to be surprisingly challenging. Due to the spectroscopic silence of Zn(2+), combinations of biophysical techniques including electrospray ionisation mass spectrometry (ESI-MS) and multinuclear NMR, isothermal titration calorimetry (ITC) and extended X-ray absorption fine structure (EXAFS) spectroscopy, coupled with site-directed mutagenesis and molecular modelling have proven to be valuable approaches to understand not only the zinc-binding properties of metallothioneins and albumins, but also the influence of other physiologically relevant competing agents. These studies have demonstrated why the bacterial metallothionein SmtA contains a site inert towards exchange with Cd(2+), why the plant metallothionein EC from wheat is partially unfolded in the presence of Cd(2+), and how fatty acids impact on the zinc-binding ability of mammalian serum albumins.
Collapse
|
90
|
Abstract
The nutritional essentiality of zinc for the growth of living organisms had been recognized long before zinc biochemistry began with the discovery of zinc in carbonic anhydrase in 1939. Painstaking analytical work then demonstrated the presence of zinc as a catalytic and structural cofactor in a few hundred enzymes. In the 1980s, the field again gained momentum with the new principle of "zinc finger" proteins, in which zinc has structural functions in domains that interact with other biomolecules. Advances in structural biology and a rapid increase in the availability of gene/protein databases now made it possible to predict zinc-binding sites from metal-binding motifs detected in sequences. This procedure resulted in the definition of zinc proteomes and the remarkable estimate that the human genome encodes ∼3000 zinc proteins. More recent developments focus on the regulatory functions of zinc(II) ions in intra- and intercellular information transfer and have tantalizing implications for yet additional functions of zinc in signal transduction and cellular control. At least three dozen proteins homeostatically control the vesicular storage and subcellular distribution of zinc and the concentrations of zinc(II) ions. Novel principles emerge from quantitative investigations on how strongly zinc interacts with proteins and how it is buffered to control the remarkably low cellular and subcellular concentrations of free zinc(II) ions. It is fair to conclude that the impact of zinc for health and disease will be at least as far-reaching as that of iron.
Collapse
Affiliation(s)
- Wolfgang Maret
- King's College London, Metal Metabolism Group, Division of Diabetes and Nutritional Sciences, School of Medicine, London, United Kingdom.
| |
Collapse
|
91
|
Abstract
Zinc(II) ions are catalytic, structural, and regulatory cofactors in proteins. In contrast to painstakingly collecting the pieces by isolating and characterizing zinc proteins, 'omics' approaches are now allowing us to tease out information about zinc proteins from genomes and to piece together the information to a broader knowledge and appreciation of the role of zinc in biology. Estimates for the number of zinc proteins in the human genome and in genomes of other organisms have been derived from a bioinformatics approach: mining sequence databases for homologies of known zinc-coordination motifs with characteristic ligand signatures for metal binding and combining this information with the knowledge about metal-binding domains of proteins. This approach resulted in an impressive number of almost 3000 human zinc proteins and made major contributions to our understanding of the composition of the zinc proteome and the functions of zinc proteins. However, the impact of zinc on protein science is even greater. Predictions do not include yet undiscovered ligand signatures, coordination environments that employ complex binding patterns with nonsequential binding of ligands and ligand bridges, zinc/protein interactions at protein interfaces, and transient interactions of zinc(II) ions with proteins that are not known to be zinc proteins. All this information and recent discoveries of how cellular zinc is controlled and how zinc(II) ions function as signaling ions add an hitherto unrecognized dimension to the zinc proteome of multicellular eukaryotic organisms. Zinc proteomics employs a combination of approaches from different disciplines, such as bioinformatics, biology, inorganic biochemistry, and significantly, analytical and structural chemistry. It provides crucial large-scale datasets for interpreting the roles of zinc in health and disease at both a molecular and a global, systems biology, level.
Collapse
Affiliation(s)
- Wolfgang Maret
- King's College London, School of Medicine, Diabetes and Nutritional Sciences Division, Metal Metabolism Group, London, SE1 9NH, UK,
| |
Collapse
|
92
|
|
93
|
Abstract
The vast knowledge of the physiologic functions of zinc in at least 3000 proteins and the recent recognition of fundamental regulatory functions of zinc(II) ions released from cells or within cells links this nutritionally essential metal ion to numerous diseases. However, this knowledge so far has had remarkably limited impact on diagnosing, preventing, and treating human diseases. One major roadblock is a lack of suitable biomarkers that would detect changes in cellular zinc metabolism and relate them to specific disease outcomes. It is not only the right amount of zinc in the diet that maintains health. At least as important is the proper functioning of the dozens of proteins that control cellular zinc homeostasis, regulate intracellular traffic of zinc between the cytosol and vesicles/organelles, and determine the fluctuations of signaling zinc(II) ions. Cellular zinc deficiencies or overloads, a term referring to zinc concentrations exceeding the cellular zinc buffering capacity, compromise the redox balance. Zinc supplementation may not readily remedy zinc deficiency if other factors limit the capability of a cell to control zinc. The role of zinc in human diseases requires a general understanding of the wide spectrum of functions of zinc, how zinc is controlled, how it interacts with the metabolism of other metal ions, in particular copper and iron, and how perturbation of specific zinc-dependent molecular processes causes disease and influences the progression of disease.
Collapse
|
94
|
Abstract
Cadmium is known for its toxicity in animals and man as it is not used in these species. Its only role in biology is as a zinc replacement at the catalytic site of a particular class of carbonic anhydrases in some marine diatoms. The toxicity of cadmium continues to be a significant public health concern as cadmium enters the food chain and it is taken up by tobacco smokers. The biochemical basis for its toxicity has been the objective of research for over 50 years. Cadmium damages the kidneys, the lungs upon inhalation, and interferes with bone metabolism. Evidence is accumulating that it affects the cardiovascular system. Cadmium is classified as a human carcinogen. It generates oxidative stress. This chapter discusses the chemistry and biochemistry of cadmium(II) ions, the only important state of cadmium in biology. This background is needed to interpret the countless effects of cadmium in laboratory experiments with cultured cells or with animals with regard to their significance for human health. Evaluation of the risks of cadmium exposure and the risk factors that affect cadmium's biological effects in tissues is an on-going process. It appears that the more we learn about the biochemistry of cadmium and the more sensitive assays we develop for determining exposure, the lower we need to set the upper limits for exposure to protect those at risk. But proper control of cadmium's presence and interactions with living species and the environment still needs to be based on improved knowledge about the mechanisms of cadmium toxicity; the gaps in our knowledge in this area are discussed herein.
Collapse
|
95
|
De Leon-Rodriguez L, Lubag AJM, Sherry AD. Imaging free zinc levels in vivo - what can be learned? Inorganica Chim Acta 2012; 393:12-23. [PMID: 23180883 PMCID: PMC3501686 DOI: 10.1016/j.ica.2012.06.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our ever-expanding knowledge about the role of zinc in biology includes its role in redox modulation, immune response, neurotransmission, reproduction, diabetes, cancer, and Alzheimers disease is galvanizing interest in detecting and monitoring the various forms of Zn(II) in biological systems. This paper reviews reported strategies for detecting and tracking of labile or "free" unchelated Zn(II) in tissues. While different bound structural forms of Zn(II) have been identified and studied in vitro by multiple techniques, very few molecular imaging methods have successfully tracked the ion in vivo. A number of MRI and optical strategies have now been reported for detection of free Zn(II) in cells and tissues but only a few have been applied successfully in vivo. A recent report of a MRI sensor for in vivo tracking of Zn(II) released from pancreatic β-cells during insulin secretion exemplifies the promise of rational design of new Zn(II) sensors for tracking this biologically important ion in vivo. Such studies promise to provide new insights into zinc trafficking in vivo and the critical role of this ion in many human diseases.
Collapse
Affiliation(s)
- Luis De Leon-Rodriguez
- Departamento de Quimica. Universidad de Guanajuato. Cerro de la Venada S.N. Col. Pueblito de Rocha., Guanajuato, Gto. Mexico, C.P, 36040
| | | | | |
Collapse
|
96
|
Alvarez L, Gonzalez-Iglesias H, Garcia M, Ghosh S, Sanz-Medel A, Coca-Prados M. The stoichiometric transition from Zn6Cu1-metallothionein to Zn7-metallothionein underlies the up-regulation of metallothionein (MT) expression: quantitative analysis of MT-metal load in eye cells. J Biol Chem 2012; 287:28456-69. [PMID: 22722935 DOI: 10.1074/jbc.m112.365015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We examined the profiling of gene expression of metallothioneins (MTs) in human tissues from cadaver eyes with microarray-based analysis. All MT1 isoforms, with the exception of MT1B, were abundantly expressed in lens and corneal tissue. Along with MT1B, MT4 was not detected in any tissues. Antibodies to MT1/2 labeled the corneal epithelial and endothelial cells, whereas MT3 label the retinal ganglion cells. We studied the effects of zinc and cytokines on the gene expression of MT isoforms in a corneal epithelial cell line (HCEsv). Zinc exerted an up-regulation of the expression of MT isoforms, and this effect was further potentiated in the presence of IL1α or TNFα. Zinc also elicited a strong down-regulation of the expression of inflammatory cytokines, and this effect was blocked in the presence of TNFα or IL1α. The concentration of MTs, bound zinc, and the metal stoichiometry of MTs in cultured HCEsv were determined by mass spectrometry. The total concentration of MTs was 0.24 ± 0.03 μM and, after 24 h of zinc exposure, increased to 0.96 ± 0.01 μM. The combination of zinc and IL1α further enhanced the level of MTs to 1.13 ± 0.03 μM. The average metal stoichiometry of MTs was Zn(6)Cu(1)-MT, and after exposure to the different treatments, it changed to Zn(7)-MT. Actinomycin D blocked transcription, and cycloheximide attenuated synthesis of MTs in the presence or absence of zinc, suggesting transcriptional regulation. Overall the data provide molecular and analytical evidence on the interplay between zinc, MTs, and proinflammatory cytokines in HCEsv cells, with potential implications on cell-based inflammatory eye diseases.
Collapse
Affiliation(s)
- Lydia Alvarez
- Fundación de Investigación Oftalmológica, Instituto Oftalmológico Fernández-Vega, 33012 Oviedo, Spain
| | | | | | | | | | | |
Collapse
|
97
|
Sikorska M, Krężel A, Otlewski J. Femtomolar Zn2+ affinity of LIM domain of PDLIM1 protein uncovers crucial contribution of protein-protein interactions to protein stability. J Inorg Biochem 2012; 115:28-35. [PMID: 22922308 DOI: 10.1016/j.jinorgbio.2012.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 05/24/2012] [Accepted: 05/28/2012] [Indexed: 10/28/2022]
Abstract
An individual LIM domain has approximately 55 amino acids with 8 highly conserved residues responsible for binding of two Zn(2+) into two distinct zinc finger motifs. We examined LIM domain stability of PDLIM1 protein (known also as Elfin protein), its C-terminally extended constructs as well as separate zinc fingers, and several full domain mutants in terms of Zn(2+) affinity and domain stability. Thermal denaturation, mass spectrometry, limited proteolysis, protein oxidation and circular dichroism techniques were used to determine a set of thermodynamic stability parameters. The results demonstrate unambiguously very high (femtomolar) affinity of both Zn(2+) to the conserved LIM domain (K(d)(av)=2.5×10(-14) M) and its additional elevation in the C-terminally extended domain construct (K(d)(av)=3.1×10(-15) M). We demonstrate in the example of PDLIM1 using a set of LIM protein constructs and its zinc finger peptides that stability of the entire zinc-containing domain is not only defined by the Zn(2+) coordination environment but significantly depends on the set of protein-protein interactions with the C-terminus of the protein. We discuss structural similarities of LIM domains and suggest the prolongation of the conserved LIM sequence to its C-terminal helix that has a significant impact on domain stability. We also discuss the functionality of LIM domains in terms of different physiological zinc and redox buffering capacity.
Collapse
Affiliation(s)
- Małgorzata Sikorska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wrocław, Tamka 2, 50-137 Wrocław, Poland
| | | | | |
Collapse
|
98
|
Huber KL, Hardy JA. Mechanism of zinc-mediated inhibition of caspase-9. Protein Sci 2012; 21:1056-65. [PMID: 22573662 DOI: 10.1002/pro.2090] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 04/25/2012] [Indexed: 11/05/2022]
Abstract
Zinc-mediated inhibition is implicated in global caspase regulation, with relief of zinc-mediated inhibition central to both small-molecule and natively induced caspase activation. As an initiator, caspase-9 regulates the upstream stages of the apoptotic caspase cascade, making it a critical control point. Here we identify two distinct zinc-binding sites on caspase-9. The first site, composed of H237, C239, and C287, includes the active site dyad and is primarily responsible for zinc-mediated inhibition. The second binding site at C272 is distal from the active site. Given the amino-acid conservation in both regions, these sites appear to be present across the caspase family underscoring the importance of zinc-mediated regulation of this class of enzymes.
Collapse
Affiliation(s)
- Kristen L Huber
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | | |
Collapse
|
99
|
Sharif R, Thomas P, Zalewski P, Fenech M. The role of zinc in genomic stability. Mutat Res 2012; 733:111-121. [PMID: 21939673 DOI: 10.1016/j.mrfmmm.2011.08.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 08/23/2011] [Accepted: 08/25/2011] [Indexed: 05/31/2023]
Abstract
Zinc (Zn) is an essential trace element required for maintaining both optimal human health and genomic stability. Zn plays a critical role in the regulation of DNA repair mechanisms, cell proliferation, differentiation and apoptosis involving the action of various transcriptional factors and DNA or RNA polymerases. Zn is an essential cofactor or structural component for important antioxidant defence proteins and DNA repair enzymes such as Cu/Zn SOD, OGG1, APE and PARP and may also affect activities of enzymes such as BHMT and MTR involved in methylation reactions in the folate-methionine cycle. This review focuses on the role of Zn in the maintenance of genome integrity and the effects of deficiency or excess on genomic stability events and cell death.
Collapse
Affiliation(s)
- Razinah Sharif
- CSIRO Food and Nutritional Sciences, Adelaide, South Australia, Australia
| | | | | | | |
Collapse
|
100
|
Thambiayya K, Wasserloos K, Kagan VE, Stoyanovsky D, Pitt BR. A critical role for increased labile zinc in reducing sensitivity of cultured sheep pulmonary artery endothelial cells to LPS-induced apoptosis. Am J Physiol Lung Cell Mol Physiol 2012; 302:L1287-95. [PMID: 22523284 DOI: 10.1152/ajplung.00385.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously noted an important signaling role for decreased labile intracellular zinc ([ Zn ] (i)) in LPS-induced apoptosis in cultured sheep pulmonary artery endothelial cells (SPAEC) (Tang ZL, Wasserloos KJ, Liu X, Stitt MS, Reynolds IJ, Pitt BR, St Croix CM. Mol Cell Biochem 234-235: 211-217, 2002; Thambiayya K, Wasserloos KJ, Huang Z, Kagan VE, St Croix CM, Pitt BR. Am J Physiol Lung Cell Mol Physiol 300: L624-632, 2011). In the present study, we used small interfering RNA (siRNA) to important contributors of zinc homeostasis [ SLC39A14 or Zrt/Irt-like protein 14 (ZIP14), a zinc importer; metallothionein (MT), a zinc binding protein ] to define molecular pathways by which extracellular zinc or nitric oxide (NO) increase labile [ Zn ] (i) [ e.g., zinc-sensitive fluorophore (FluoZin-3) detectable and/or chelatable by N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine ] and reduce the sensitivity of SPAEC to LPS. Addition of 10 μM zinc to serum-free medium of SPAEC increased [ Zn ] (i) and abolished LPS-induced apoptosis (e.g., increased annexin V binding). The increase in [ Zn ] (i) and the protective effect of extracellular zinc were sensitive to reduction in ZIP14 expression (by siRNA), but not affected by collectively knocking down major isoforms of sheep MT (sMT-Ia, -Ib, -Ic, and -II). Pretreatment of wild-type SPAEC with 250 μM of the NO donor S-nitroso-N-acetylpenicillamine (SNAP) increased labile zinc in a relatively similar fashion to addition of extracellular zinc and reduced sensitivity of SPAEC to LPS-induced apoptosis (e.g., caspase-3/7 activation) in a N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine-sensitive fashion. The antiapoptotic effects of SNAP were insensitive to siRNA knockdown of ZIP14, but were abolished (along with SNAP-induced increase in [ Zn ] (i)) when SPAEC were pretreated with siRNA to sheep MT. Zinc was able to directly inhibit recombinant caspase-3 activity in an in vitro assay. Collectively, these data show that increases in labile [ Zn ] (i) are an important component of ZIP14- or NO-mediated resistance to LPS-induced apoptosis. Cytoprotection via ZIP14 appeared to be secondary to transcellular movement of extracellular zinc, whereas NO-mediated protection was secondary to S-nitrosation of MT and redistribution of [ Zn ] (i).
Collapse
Affiliation(s)
- Kalidasan Thambiayya
- Dept. Environmental and Occupational Health, Univ. of Pittsburgh Graduate School Public Health, Bridgeside Point Bldg., 100 Technology Dr., Ste. 555, Pittsburgh, PA 15219, USA
| | | | | | | | | |
Collapse
|