1
|
Liu Q, Qin W, Hu G, Qin Y, Chen B, Huang J, Xing Z, Xu Q. A cyanobiphenol-based fluorescent probe for simultaneous recognition of Al 3+ and Zn 2+ and its application. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 329:125538. [PMID: 39637567 DOI: 10.1016/j.saa.2024.125538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/15/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
A facile cyanobiphenol based probe MHB was synthesized and determined by FT-IR, 1H NMR, 13C NMR, HRMS and X-ray Crystallography. The probe MHB exhibited fluorescence turn-on phenomenon accompany with obvious color change from dark bule to azure and blue-green upon the addition of Al3+ and Zn2+, respectively. The binding ratios between probe MHB and Al3+/Zn2+ was 1:1 determined by Job's plot, which was deeply investigated by FT-IR spectrum, 1HNMR titration and HRMS analysis. The detection limits of MHB to Al3+ and Zn2+ were 2.52 × 10-7 M and 1.74 × 10-7 M for Zn2+, respectively. The application experiments referring to real water samples, test paper and cell image showed that MHB was capable of qualitative and quantitative sensing Al3+/Zn2+ in environmental and biological system.
Collapse
Affiliation(s)
- Qi Liu
- Department of Chemistry, College of Arts and Sciences, Northeast Agricultural University, Harbin 150030, China
| | - Wen Qin
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China; Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Guiying Hu
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Yanfei Qin
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China; Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Bolin Chen
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China; Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Jiawen Huang
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China; Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Zhiyong Xing
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China; Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China.
| | - Qijiang Xu
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China; Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China.
| |
Collapse
|
2
|
Ocampo D, Damon LJ, Sanford L, Holtzen SE, Jones T, Allen MA, Dowell RD, Palmer AE. Cellular zinc status alters chromatin accessibility and binding of p53 to DNA. Life Sci Alliance 2024; 7:e202402638. [PMID: 38969365 PMCID: PMC11231577 DOI: 10.26508/lsa.202402638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/07/2024] Open
Abstract
Zn2+ is an essential metal required by approximately 850 human transcription factors. How these proteins acquire their essential Zn2+ cofactor and whether they are sensitive to changes in the labile Zn2+ pool in cells remain open questions. Using ATAC-seq to profile regions of accessible chromatin coupled with transcription factor enrichment analysis, we examined how increases and decreases in the labile zinc pool affect chromatin accessibility and transcription factor enrichment. We found 685 transcription factor motifs were differentially enriched, corresponding to 507 unique transcription factors. The pattern of perturbation and the types of transcription factors were notably different at promoters versus intergenic regions, with zinc-finger transcription factors strongly enriched in intergenic regions in elevated Zn2+ To test whether ATAC-seq and transcription factor enrichment analysis predictions correlate with changes in transcription factor binding, we used ChIP-qPCR to profile six p53 binding sites. We found that for five of the six targets, p53 binding correlates with the local accessibility determined by ATAC-seq. These results demonstrate that changes in labile zinc alter chromatin accessibility and transcription factor binding to DNA.
Collapse
Affiliation(s)
- Daniel Ocampo
- Department of Biochemistry, University of Colorado, Boulder, CO, USA
| | - Leah J Damon
- Department of Biochemistry, University of Colorado, Boulder, CO, USA
| | - Lynn Sanford
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Samuel E Holtzen
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Taylor Jones
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Mary A Allen
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Robin D Dowell
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Amy E Palmer
- Department of Biochemistry, University of Colorado, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| |
Collapse
|
3
|
Holtzen SE, Rakshit A, Palmer AE. Protocol for measuring labile cytosolic Zn 2+ using an in situ calibration of a genetically encoded FRET sensor. STAR Protoc 2024; 5:103130. [PMID: 38870018 PMCID: PMC11225890 DOI: 10.1016/j.xpro.2024.103130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/15/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024] Open
Abstract
Zinc (Zn2+) plays roles in structure, catalysis, and signaling. The majority of cellular Zn2+ is bound by proteins, but a fraction of total Zn2+ exists in a labile form. Here, we present a protocol for measuring labile cytosolic Zn2+ using an in situ calibration of a genetically encoded Förster resonance energy transfer (FRET) sensor. We describe steps for producing buffered Zn2+ solutions for performing an imaging-based calibration and analyzing the imaging data generated to determine labile Zn2+ concentration in single cells. For complete details on the use and execution of this protocol, please refer to Rakshit and Holtzen et al.1.
Collapse
Affiliation(s)
- Samuel E Holtzen
- BioFrontiers Institute and Department of Biochemistry, 3415 Colorado Avenue, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Molecular Cellular Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Ananya Rakshit
- BioFrontiers Institute and Department of Biochemistry, 3415 Colorado Avenue, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Amy E Palmer
- BioFrontiers Institute and Department of Biochemistry, 3415 Colorado Avenue, University of Colorado Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
4
|
Holtzen SE, Navid E, Kainov JD, Palmer AE. Transient Zn 2+ deficiency induces replication stress and compromises daughter cell proliferation. Proc Natl Acad Sci U S A 2024; 121:e2321216121. [PMID: 38687796 PMCID: PMC11087780 DOI: 10.1073/pnas.2321216121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Cells must replicate their genome quickly and accurately, and they require metabolites and cofactors to do so. Ionic zinc (Zn2+) is an essential micronutrient that is required for hundreds of cellular processes, including DNA synthesis and adequate proliferation. Deficiency in this micronutrient impairs DNA synthesis and inhibits proliferation, but the mechanism is unknown. Using fluorescent reporters to track single cells via long-term live-cell imaging, we find that Zn2+ is required at the G1/S transition and during S phase for timely completion of S phase. A short pulse of Zn2+ deficiency impairs DNA synthesis and increases markers of replication stress. These markers of replication stress are reversed upon resupply of Zn2+. Finally, we find that if Zn2+ is chelated during the mother cell's S phase, daughter cells enter a transient quiescent state, maintained by sustained expression of p21, which disappears upon reentry into the cell cycle. In summary, short pulses of mild Zn2+ deficiency in S phase specifically induce replication stress, which causes downstream proliferation impairments in daughter cells.
Collapse
Affiliation(s)
- Samuel E. Holtzen
- Department of Molecular Cellular and Developmental Biology, University of Colorado, Boulder, CO80309
| | - Elnaz Navid
- Department of Biochemistry, University of Colorado, Boulder, CO80309
| | - Joseph D. Kainov
- Department of Biochemistry, University of Colorado, Boulder, CO80309
| | - Amy E. Palmer
- Department of Biochemistry, University of Colorado, Boulder, CO80309
- BioFrontiers Institute, University of Colorado, Boulder, CO80309
| |
Collapse
|
5
|
Jensen GC, Janis MK, Nguyen HN, David OW, Zastrow ML. Fluorescent Protein-Based Sensors for Detecting Essential Metal Ions across the Tree of Life. ACS Sens 2024; 9:1622-1643. [PMID: 38587931 PMCID: PMC11073808 DOI: 10.1021/acssensors.3c02695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Genetically encoded fluorescent metal ion sensors are powerful tools for elucidating metal dynamics in living systems. Over the last 25 years since the first examples of genetically encoded fluorescent protein-based calcium indicators, this toolbox of probes has expanded to include other essential and non-essential metal ions. Collectively, these tools have illuminated fundamental aspects of metal homeostasis and trafficking that are crucial to fields ranging from neurobiology to human nutrition. Despite these advances, much of the application of metal ion sensors remains limited to mammalian cells and tissues and a limited number of essential metals. Applications beyond mammalian systems and in vivo applications in living organisms have primarily used genetically encoded calcium ion sensors. The aim of this Perspective is to provide, with the support of historical and recent literature, an updated and critical view of the design and use of fluorescent protein-based sensors for detecting essential metal ions in various organisms. We highlight the historical progress and achievements with calcium sensors and discuss more recent advances and opportunities for the detection of other essential metal ions. We also discuss outstanding challenges in the field and directions for future studies, including detecting a wider variety of metal ions, developing and implementing a broader spectral range of sensors for multiplexing experiments, and applying sensors to a wider range of single- and multi-species biological systems.
Collapse
Affiliation(s)
- Gary C Jensen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Makena K Janis
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Hazel N Nguyen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Ogonna W David
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Melissa L Zastrow
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
6
|
Singh R, Sharma D, Kumar A, Singh C, Singh A. Understanding zebrafish sleep and wakefulness physiology as an experimental model for biomedical research. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:827-842. [PMID: 38150068 DOI: 10.1007/s10695-023-01288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/07/2023] [Indexed: 12/28/2023]
Abstract
Sleep is a globally observable fact, or period of reversible distracted rest, that can be distinguished from arousal by various behavioral criteria. Although the function of sleep is an evolutionarily conserved behavior, its mechanism is not yet clear. The zebrafish (Danio rerio) has become a valuable model for neurobehavioral studies such as studying learning, memory, anxiety, and depression. It is characterized by a sleep-like state and circadian rhythm, making it comparable to mammals. Zebrafish are a good model for behavioral studies because they share genetic similarities with humans. A number of neurotransmitters are involved in sleep and wakefulness. There is a binding between melatonin and the hypocretin system present in zebrafish. The full understanding of sleep and wakefulness physiology in zebrafish is still unclear among researchers. Therefore, to make a clear understanding of the sleep/wake cycle in zebrafish, this article covers the mechanism involved behind it, and the role of the neuromodulator system followed by the mechanism of the HPA axis.
Collapse
Affiliation(s)
- Rima Singh
- Department of Pharmacology, Delhi Pharmaceutical Sciences & Research University (DPSRU), New Delhi, 110017, India
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Deepali Sharma
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Anoop Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences & Research University (DPSRU), New Delhi, 110017, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Chauras Campus, Distt, Tehri Garhwal, Uttarakhand, 246174, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India.
| |
Collapse
|
7
|
Hettiarachchi P, Niyangoda S, Shigemoto A, Solowiej IJ, Burdette SC, Johnson MA. Caged Zn 2+ Photolysis in Zebrafish Whole Brains Reveals Subsecond Modulation of Dopamine Uptake. ACS Chem Neurosci 2024; 15:772-782. [PMID: 38301116 PMCID: PMC11036533 DOI: 10.1021/acschemneuro.3c00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
Free, ionic zinc (Zn2+) modulates neurotransmitter dynamics in the brain. However, the sub-s effects of transient concentration changes of Zn2+ on neurotransmitter release and uptake are not well understood. To address this lack of knowledge, we have combined the photolysis of the novel caged Zn2+ compound [Zn(DPAdeCageOMe)]+ with fast scan cyclic voltammetry (FSCV) at carbon fiber microelectrodes in live, whole brain preparations from zebrafish (Danio rerio). After treating the brain with [Zn(DPAdeCageOMe)]+, Zn2+ was released by application of light that was gated through a computer-controlled shutter synchronized with the FSCV measurements and delivered through a 1 mm fiber optic cable. We systematically optimized the photocage concentration and light application parameters, including the total duration and light-to-electrical stimulation delay time. While sub-s Zn2+ application with this method inhibited DA reuptake, assessed by the first-order rate constant (k) and half-life (t1/2), it had no effect on the electrically stimulated DA overflow ([DA]STIM). Increasing the photocage concentration and light duration progressively inhibited uptake, with maximal effects occurring at 100 μM and 800 ms, respectively. Furthermore, uptake was inhibited 200 ms after Zn2+ photorelease, but no measurable effect occurred after 800 ms. We expect that application of this method to the zebrafish whole brain and other preparations will help expand the current knowledge of how Zn2+ affects neurotransmitter release/uptake in select neurological disease states.
Collapse
Affiliation(s)
- Piyanka Hettiarachchi
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Sayuri Niyangoda
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Austin Shigemoto
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA 01609
| | - Isabel J. Solowiej
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Shawn C. Burdette
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA 01609
| | - Michael A. Johnson
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
8
|
Franco C, Canzoniero LMT. Zinc homeostasis and redox alterations in obesity. Front Endocrinol (Lausanne) 2024; 14:1273177. [PMID: 38260166 PMCID: PMC10800374 DOI: 10.3389/fendo.2023.1273177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Impairment of both cellular zinc and redox homeostasis is a feature of several chronic diseases, including obesity. A significant two-way interaction exists between redox metabolism and the relatively redox-inert zinc ion. Redox metabolism critically influences zinc homeostasis and controls its cellular availability for various cellular functions by regulating zinc exchange from/to zinc-binding proteins. Zinc can regulate redox metabolism and exhibits multiple pro-antioxidant properties. On the other hand, even minor disturbances in zinc status and zinc homeostasis affect systemic and cellular redox homeostasis. At the cellular level, zinc homeostasis is regulated by a multi-layered machinery consisting of zinc-binding molecules, zinc sensors, and two selective families of zinc transporters, the Zinc Transporter (ZnT) and Zrt, Irt-like protein (ZIP). In the present review, we summarize the current state of knowledge on the role of the mutual interaction between zinc and redox homeostasis in physiology and pathophysiology, pointing to the role of zinc in the alterations responsible for redox stress in obesity. Since zinc transporters primarily control zinc homeostasis, we describe how changes in the expression and activity of these zinc-regulating proteins are associated with obesity.
Collapse
|
9
|
Holtzen SE, Navid E, Kainov JD, Palmer AE. Transient Zn 2+ deficiency induces replication stress and compromises daughter cell proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.08.570860. [PMID: 38106081 PMCID: PMC10723434 DOI: 10.1101/2023.12.08.570860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Cells must replicate their genome quickly and accurately, and they require metabolites and cofactors to do so. Ionic zinc (Zn2+) is an essential micronutrient that is required for hundreds of cellular processes, including DNA synthesis and adequate proliferation. Deficiency in this micronutrient impairs DNA synthesis and inhibits proliferation, but the mechanism is unknown. Using fluorescent reporters to track single cells via long-term live-cell imaging, we find that Zn2+ is required at the G1/S transition and during S-phase for timely completion of S-phase. A short pulse of Zn2+ deficiency impairs DNA synthesis and increases markers of replication stress. These markers of replication stress are reversed upon resupply of Zn2+. Finally, we find that if Zn2+ is removed during the mother cell's S-phase, daughter cells enter a transient quiescent state, maintained by sustained expression of p21, which disappears upon reentry into the cell cycle. In summary, short pulses of mild Zn2+ deficiency in S-phase specifically induce replication stress, which causes downstream proliferation impairments in daughter cells.
Collapse
Affiliation(s)
- Samuel E. Holtzen
- Department of Molecular Cellular and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80309
- Department of Biochemistry, University of Colorado, Boulder, CO, 80309
| | - Elnaz Navid
- Department of Biochemistry, University of Colorado, Boulder, CO, 80309
| | - Joseph D. Kainov
- Department of Biochemistry, University of Colorado, Boulder, CO, 80309
| | - Amy E. Palmer
- Department of Biochemistry, University of Colorado, Boulder, CO, 80309
- Department of Biochemistry and BioFrontiers Institute, University of Colorado, Boulder, CO, 80309
| |
Collapse
|
10
|
Damon LJ, Ocampo D, Sanford L, Jones T, Allen MA, Dowell RD, Palmer AE. Cellular zinc status alters chromatin accessibility and binding of transcription factor p53 to genomic sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567954. [PMID: 38045276 PMCID: PMC10690171 DOI: 10.1101/2023.11.20.567954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Zinc (Zn2+) is an essential metal required by approximately 2500 proteins. Nearly half of these proteins act on DNA, including > 850 human transcription factors, polymerases, DNA damage response factors, and proteins involved in chromatin architecture. How these proteins acquire their essential Zn2+ cofactor and whether they are sensitive to changes in the labile Zn2+ pool in cells remain open questions. Here, we examine how changes in the labile Zn2+ pool affect chromatin accessibility and transcription factor binding to DNA. We observed both increases and decreases in accessibility in different chromatin regions via ATAC-seq upon treating MCF10A cells with elevated Zn2+ or the Zn2+-specific chelator tris(2-pyridylmethyl)amine (TPA). Transcription factor enrichment analysis was used to correlate changes in chromatin accessibility with transcription factor motifs, revealing 477 transcription factor motifs that were differentially enriched upon Zn2+ perturbation. 186 of these transcription factor motifs were enriched in Zn2+ and depleted in TPA, and the majority correspond to Zn2+ finger transcription factors. We selected TP53 as a candidate to examine how changes in motif enrichment correlate with changes in transcription factor occupancy by ChIP-qPCR. Using publicly available ChIP-seq and nascent transcription datasets, we narrowed the 50,000+ ATAC-seq peaks to 2164 TP53 targets and subsequently selected 6 high-probability TP53 binding sites for testing. ChIP-qPCR revealed that for 5 of the 6 targets, TP53 binding correlates with the local accessibility determined by ATAC-seq. These results demonstrate that changes in labile zinc directly alter chromatin accessibility and transcription factor binding to DNA.
Collapse
Affiliation(s)
- Leah J. Damon
- Department of Biochemistry, University of Colorado, Boulder, CO 80303
| | - Daniel Ocampo
- Department of Biochemistry, University of Colorado, Boulder, CO 80303
| | - Lynn Sanford
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, 80309
| | - Taylor Jones
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, 80309
| | - Mary A. Allen
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, 80309
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303
| | - Robin D. Dowell
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, 80309
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303
| | - Amy E. Palmer
- Department of Biochemistry, University of Colorado, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303
| |
Collapse
|
11
|
Tseng HC, Pan CY. Dopamine Activates the D1R-Zn 2+ Signaling Pathway to Trigger Inflammatory Response in Primary-Cultured Rat Embryonic Cortical Neurons. Cell Mol Neurobiol 2023; 43:3593-3604. [PMID: 37289255 DOI: 10.1007/s10571-023-01367-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
Neuroinflammation is an early event during the pathogenesis of neurodegenerative disorders. Most studies focus on how the factors derived from pathogens or tissue damage activate the inflammation-pyroptosis cell death pathway. It is unclear whether endogenous neurotransmitters could induce inflammatory responses in neurons. Our previous reports have shown that dopamine-induced elevation of intracellular Zn2+ concentration via the D1-like receptor (D1R) is a prerequisite for autophagy and cell death in primary cultured rat embryonic neurons. Here we further examined that this D1R-Zn2+ signaling initiates the transient inflammatory response leading to cell death in cultured cortical neurons. Pretreating the cultured neurons with Zn2+ chelator and inhibitors against inflammation could enhance the cell viability in neurons treated with dopamine and dihydrexidine, an agonist of D1R. Both dopamine and dihydrexidine greatly enhanced inflammasome formation; a Zn2+ chelator, N,N,N',N'-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine, suppressed this increment. Dopamine and dihydrexidine increased the expression levels of NOD-like receptor pyrin domain-containing protein 3 and enhanced the maturation of caspase-1, gasdermin D, and IL-1β; these changes were all Zn2+-dependent. Dopamine treatment did not recruit the N-terminal of the gasdermin D to the plasma membrane but enhanced its localization to the autophagosomes. Pretreating the neurons with IL-1β could increase the viability of neurons challenged with dopamine. These results demonstrate a novel D1R-Zn2+ signaling cascade activating neuroinflammation and cell death. Therefore, maintaining a balance between dopamine homeostasis and inflammatory responses is an important therapeutic target for neurodegeneration. Dopamine elicits transient inflammatory responses in cultured cortical neurons via the D1R-Zn2+ signaling pathway. Dopamine elevates [Zn2+]i to induce the formation of inflammasomes, which activates caspase-1, resulting in the maturation of IL-1β and gasdermin D (GSDMD). Therefore, the homeostasis of dopamine and Zn2+ are critical therapeutic targets for inflammation-derived neurodegeneration.
Collapse
Affiliation(s)
- Hui-Chiun Tseng
- Department of Life Science, National Taiwan University, 1 Roosevelt Rd. Sec 4, Taipei, 106, Taiwan
| | - Chien-Yuan Pan
- Department of Life Science, National Taiwan University, 1 Roosevelt Rd. Sec 4, Taipei, 106, Taiwan.
| |
Collapse
|
12
|
Minckley TF, Salvagio LA, Fudge DH, Verhey K, Markus SM, Qin Y. Zn2+ decoration of microtubules arrests axonal transport and displaces tau, doublecortin, and MAP2C. J Cell Biol 2023; 222:e202208121. [PMID: 37326602 PMCID: PMC10276529 DOI: 10.1083/jcb.202208121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/31/2023] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
Intracellular Zn2+ concentrations increase via depolarization-mediated influx or intracellular release, but the immediate effects of Zn2+ signals on neuron function are not fully understood. By simultaneous recording of cytosolic Zn2+ and organelle motility, we find that elevated Zn2+ (IC50 ≈ 5-10 nM) reduces both lysosomal and mitochondrial motility in primary rat hippocampal neurons and HeLa cells. Using live-cell confocal microscopy and in vitro single-molecule TIRF imaging, we reveal that Zn2+ inhibits activity of motor proteins (kinesin and dynein) without disrupting their microtubule binding. Instead, Zn2+ directly binds to microtubules and selectively promotes detachment of tau, DCX, and MAP2C, but not MAP1B, MAP4, MAP7, MAP9, or p150glued. Bioinformatic predictions and structural modeling show that the Zn2+ binding sites on microtubules partially overlap with the microtubule binding sites of tau, DCX, dynein, and kinesin. Our results reveal that intraneuronal Zn2+ regulates axonal transport and microtubule-based processes by interacting with microtubules.
Collapse
Affiliation(s)
- Taylor F. Minckley
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | | | - Dylan H. Fudge
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kristen Verhey
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Steven M. Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Yan Qin
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| |
Collapse
|
13
|
Martínez-Parra L, Piñol-Cancer M, Sanchez-Cano C, Miguel-Coello AB, Di Silvio D, Gomez AM, Uriel C, Plaza-García S, Gallego M, Pazos R, Groult H, Jeannin M, Geraki K, Fernández-Méndez L, Urkola-Arsuaga A, Sánchez-Guisado MJ, Carrillo-Romero J, Parak WJ, Prato M, Herranz F, Ruiz-Cabello J, Carregal-Romero S. A Comparative Study of Ultrasmall Calcium Carbonate Nanoparticles for Targeting and Imaging Atherosclerotic Plaque. ACS NANO 2023; 17:13811-13825. [PMID: 37399106 PMCID: PMC10900527 DOI: 10.1021/acsnano.3c03523] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Atherosclerosis is a complex disease that can lead to life-threatening events, such as myocardial infarction and ischemic stroke. Despite the severity of this disease, diagnosing plaque vulnerability remains challenging due to the lack of effective diagnostic tools. Conventional diagnostic protocols lack specificity and fail to predict the type of atherosclerotic lesion and the risk of plaque rupture. To address this issue, technologies are emerging, such as noninvasive medical imaging of atherosclerotic plaque with customized nanotechnological solutions. Modulating the biological interactions and contrast of nanoparticles in various imaging techniques, including magnetic resonance imaging, is possible through the careful design of their physicochemical properties. However, few examples of comparative studies between nanoparticles targeting different hallmarks of atherosclerosis exist to provide information about the plaque development stage. Our work demonstrates that Gd (III)-doped amorphous calcium carbonate nanoparticles are an effective tool for these comparative studies due to their high magnetic resonance contrast and physicochemical properties. In an animal model of atherosclerosis, we compare the imaging performance of three types of nanoparticles: bare amorphous calcium carbonate and those functionalized with the ligands alendronate (for microcalcification targeting) and trimannose (for inflammation targeting). Our study provides useful insights into ligand-mediated targeted imaging of atherosclerosis through a combination of in vivo imaging, ex vivo tissue analysis, and in vitro targeting experiments.
Collapse
Affiliation(s)
- Lydia Martínez-Parra
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), 20018 Donostia, Spain
| | - Marina Piñol-Cancer
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), 20018 Donostia, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Carlos Sanchez-Cano
- Donostia International Physics Center, Paseo Manuel de Lardizabal 4, Donostia, 20018, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea (UPV/EHU), 20018 Donostia, Spain
| | - Ana B Miguel-Coello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
| | - Desirè Di Silvio
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
| | - Ana M Gomez
- Instituto de Química Orgánica General, IQOG-CSIC, 28006 Madrid, Spain
| | - Clara Uriel
- Instituto de Química Orgánica General, IQOG-CSIC, 28006 Madrid, Spain
| | - Sandra Plaza-García
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
| | - Marta Gallego
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
| | - Raquel Pazos
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
| | - Hugo Groult
- Biotechnologies et Chimie des Bioressources pour la Santé, Littoral Environment et Sociétés (LIENSs Laboratory), UMR CNRS 7266, 17000 La Rochelle, France
| | - Marc Jeannin
- Laboratoire des Sciences de l'Ingénieur pour l'Environnement (LaSIE), UMR-CNRS 7536, La Rochelle Université, 7356 La Rochelle, France
| | - Kalotina Geraki
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| | - Laura Fernández-Méndez
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), 20018 Donostia, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Ainhize Urkola-Arsuaga
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
| | - María Jesús Sánchez-Guisado
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- Euskal Herriko Unibertsitatea (UPV/EHU), 20018 Donostia, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Juliana Carrillo-Romero
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- Basque Res & Technol Alliance BRTA, GAIKER, Technol Ctr, 48170 Zamudio, Spain
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, 22607 Hamburg, Germany
| | - Maurizio Prato
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Fernando Herranz
- NanoMedMol, Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid 28006, Spain
| | - Jesús Ruiz-Cabello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Susana Carregal-Romero
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
14
|
Dorward AM, Stewart AJ, Pitt SJ. The role of Zn2+ in shaping intracellular Ca2+ dynamics in the heart. J Gen Physiol 2023; 155:e202213206. [PMID: 37326614 PMCID: PMC10276528 DOI: 10.1085/jgp.202213206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Increasing evidence suggests that Zn2+ acts as a second messenger capable of transducing extracellular stimuli into intracellular signaling events. The importance of Zn2+ as a signaling molecule in cardiovascular functioning is gaining traction. In the heart, Zn2+ plays important roles in excitation-contraction (EC) coupling, excitation-transcription coupling, and cardiac ventricular morphogenesis. Zn2+ homeostasis in cardiac tissue is tightly regulated through the action of a combination of transporters, buffers, and sensors. Zn2+ mishandling is a common feature of various cardiovascular diseases. However, the precise mechanisms controlling the intracellular distribution of Zn2+ and its variations during normal cardiac function and during pathological conditions are not fully understood. In this review, we consider the major pathways by which the concentration of intracellular Zn2+ is regulated in the heart, the role of Zn2+ in EC coupling, and discuss how Zn2+ dyshomeostasis resulting from altered expression levels and efficacy of Zn2+ regulatory proteins are key drivers in the progression of cardiac dysfunction.
Collapse
Affiliation(s)
- Amy M. Dorward
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Alan J. Stewart
- School of Medicine, University of St Andrews, St Andrews, UK
| | | |
Collapse
|
15
|
Rakshit A, Holtzen SE, Lo MN, Conway KA, Palmer AE. Human cells experience a Zn 2+ pulse in early G1. Cell Rep 2023; 42:112656. [PMID: 37330912 PMCID: PMC10592493 DOI: 10.1016/j.celrep.2023.112656] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/07/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
Zinc is an essential micronutrient required for all domains of life. Cells maintain zinc homeostasis using a network of transporters, buffers, and transcription factors. Zinc is required for mammalian cell proliferation, and zinc homeostasis is remodeled during the cell cycle, but whether labile zinc changes in naturally cycling cells has not been established. We use genetically encoded fluorescent reporters, long-term time-lapse imaging, and computational tools to track labile zinc over the cell cycle in response to changes in growth media zinc and knockdown of the zinc-regulatory transcription factor MTF-1. Cells experience a pulse of labile zinc in early G1, whose magnitude varies with zinc in growth media. Knockdown of MTF-1 increases labile zinc and the zinc pulse. Our results suggest that cells need a minimum zinc pulse to proliferate and that if labile zinc levels are too high, cells pause proliferation until labile cellular zinc is lowered.
Collapse
Affiliation(s)
- Ananya Rakshit
- Department of Biochemistry and BioFrontiers Institute, 3415 Colorado Avenue, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Samuel E Holtzen
- Department of Molecular Cellular Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Maria N Lo
- Department of Biochemistry and BioFrontiers Institute, 3415 Colorado Avenue, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Kylie A Conway
- Department of Biochemistry and BioFrontiers Institute, 3415 Colorado Avenue, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Amy E Palmer
- Department of Biochemistry and BioFrontiers Institute, 3415 Colorado Avenue, University of Colorado Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
16
|
Schneider SE, Scott AK, Seelbinder B, Elzen CVD, Wilson RL, Miller EY, Beato QI, Ghosh S, Barthold JE, Bilyeu J, Emery NC, Pierce DM, Neu CP. Dynamic biophysical responses of neuronal cell nuclei and cytoskeletal structure following high impulse loading. Acta Biomater 2023; 163:339-350. [PMID: 35811070 PMCID: PMC10019187 DOI: 10.1016/j.actbio.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/11/2022] [Accepted: 07/01/2022] [Indexed: 12/28/2022]
Abstract
Cells are continuously exposed to dynamic environmental cues that influence their behavior. Mechanical cues can influence cellular and genomic architecture, gene expression, and intranuclear mechanics, providing evidence of mechanosensing by the nucleus, and a mechanoreciprocity between the nucleus and environment. Force disruption at the tissue level through aging, disease, or trauma, propagates to the nucleus and can have lasting consequences on proper functioning of the cell and nucleus. While the influence of mechanical cues leading to axonal damage has been well studied in neuronal cells, the mechanics of the nucleus following high impulse loading is still largely unexplored. Using an in vitro model of traumatic neural injury, we show a dynamic nuclear behavioral response to impulse stretch (up to 170% strain per second) through quantitative measures of nuclear movement, including tracking of rotation and internal motion. Differences in nuclear movement were observed between low and high strain magnitudes. Increased exposure to impulse stretch exaggerated the decrease in internal motion, assessed by particle tracking microrheology, and intranuclear displacements, assessed through high-resolution deformable image registration. An increase in F-actin puncta surrounding nuclei exposed to impulse stretch additionally demonstrated a corresponding disruption of the cytoskeletal network. Our results show direct biophysical nuclear responsiveness in neuronal cells through force propagation from the substrate to the nucleus. Understanding how mechanical forces perturb the morphological and behavioral response can lead to a greater understanding of how mechanical strain drives changes within the cell and nucleus, and may inform fundamental nuclear behavior after traumatic axonal injury. STATEMENT OF SIGNIFICANCE: The nucleus of the cell has been implicated as a mechano-sensitive organelle, courting molecular sensors and transmitting physical cues in order to maintain cellular and tissue homeostasis. Disruption of this network due to disease or high velocity forces (e.g., trauma) can not only result in orchestrated biochemical cascades, but also biophysical perturbations. Using an in vitro model of traumatic neural injury, we aimed to provide insight into the neuronal nuclear mechanics and biophysical responses at a continuum of strain magnitudes and after repetitive loads. Our image-based methods demonstrate mechanically-induced changes in cellular and nuclear behavior after high intensity loading and have the potential to further define mechanical thresholds of neuronal cell injury.
Collapse
Affiliation(s)
- Stephanie E Schneider
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Adrienne K Scott
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Benjamin Seelbinder
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Courtney Van Den Elzen
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Robert L Wilson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Emily Y Miller
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| | - Quinn I Beato
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Soham Ghosh
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA; Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA; School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Jeanne E Barthold
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Jason Bilyeu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Nancy C Emery
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, CO, USA
| | - David M Pierce
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Corey P Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA; Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
17
|
Swartchick CB, Chan J. Leveraging coordination chemistry to visualize metal ions via photoacoustic imaging. Curr Opin Chem Biol 2023; 74:102312. [PMID: 37146434 DOI: 10.1016/j.cbpa.2023.102312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 05/07/2023]
Abstract
Metal ions are indispensable to all living systems owing to their diverse roles. Perturbation of metal homeostasis have been linked to many pathological conditions. As such, visualizing metal ions in these complex environments are of utmost importance. Photoacoustic imaging is a promising modality that combines the sensitivity of fluorescence to the superior resolution of ultrasound, through a light-in sound-out process, making it an appealing modality for metal ion detection in vivo. In this review, we highlight recent advances in the development of photoacoustic imaging probes for in vivo detection of metal ions, such as potassium, copper, zinc, and palladium. In addition, we provide our perspective and outlook on the exciting field.
Collapse
Affiliation(s)
- Chelsea B Swartchick
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Jefferson Chan
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States.
| |
Collapse
|
18
|
Alshawaf AJ, Alnassar SA, Al-Mohanna FA. The interplay of intracellular calcium and zinc ions in response to electric field stimulation in primary rat cortical neurons in vitro. Front Cell Neurosci 2023; 17:1118335. [PMID: 37180947 PMCID: PMC10174245 DOI: 10.3389/fncel.2023.1118335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Recent pharmacological studies demonstrate a role for zinc (Zn2+) in shaping intracellular calcium (Ca2+) dynamics and vice versa in excitable cells including neurons and cardiomyocytes. Herein, we sought to examine the dynamic of intracellular release of Ca2+ and Zn2+ upon modifying excitability of primary rat cortical neurons using electric field stimulation (EFS) in vitro. We show that exposure to EFS with an intensity of 7.69 V/cm induces transient membrane hyperpolarization together with transient elevations in the cytosolic levels of Ca2+ and Zn2+ ions. The EFS-induced hyperpolarization was inhibited by prior treatment of cells with the K+ channel opener diazoxide. Chemical hyperpolarization had no apparent effect on either Ca2+ or Zn2+. The source of EFS-induced rise in Ca2+ and Zn2+ seemed to be intracellular, and that the dynamic inferred of an interplay between Ca2+ and Zn2+ ions, whereby the removal of extracellular Ca2+ augmented the release of intracellular Ca2+ and Zn2+ and caused a stronger and more sustained hyperpolarization. We demonstrate that Zn2+ is released from intracellular vesicles located in the soma, with major co-localizations in the lysosomes and endoplasmic reticulum. These studies further support the use of EFS as a tool to interrogate the kinetics of intracellular ions in response to changing membrane potential in vitro.
Collapse
Affiliation(s)
- Abdullah J. Alshawaf
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sarah A. Alnassar
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Futwan A. Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- *Correspondence: Futwan A. Al-Mohanna,
| |
Collapse
|
19
|
Goldberg JM, Lippard SJ. Mobile zinc as a modulator of sensory perception. FEBS Lett 2023; 597:151-165. [PMID: 36416529 PMCID: PMC10108044 DOI: 10.1002/1873-3468.14544] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
Mobile zinc is an abundant transition metal ion in the central nervous system, with pools of divalent zinc accumulating in regions of the brain engaged in sensory perception and memory formation. Here, we present essential tools that we developed to interrogate the role(s) of mobile zinc in these processes. Most important are (a) fluorescent sensors that report the presence of mobile zinc and (b) fast, Zn-selective chelating agents for measuring zinc flux in animal tissue and live animals. The results of our studies, conducted in collaboration with neuroscientist experts, are presented for sensory organs involved in hearing, smell, vision, and learning and memory. A general principle emerging from these studies is that the function of mobile zinc in all cases appears to be downregulation of the amplitude of the response following overstimulation of the respective sensory organs. Possible consequences affecting human behavior are presented for future investigations in collaboration with interested behavioral scientists.
Collapse
Affiliation(s)
| | - Stephen J Lippard
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
20
|
Krall R, Gale JR, Ross MM, Tzounopoulos T, Aizenman E. Intracellular zinc signaling influences NMDA receptor function by enhancing the interaction of ZnT1 with GluN2A. Neurosci Lett 2022; 790:136896. [PMID: 36202195 PMCID: PMC10153101 DOI: 10.1016/j.neulet.2022.136896] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022]
Abstract
Zinc, loaded into glutamate-containing presynaptic vesicles and released into the synapse in an activity-dependent manner, modulates neurotransmission through its actions on postsynaptic targets, prominently via high-affinity inhibition of GluN2A-containing NMDA receptors. Recently, we identified a postsynaptic transport mechanism that regulates endogenous zinc inhibition of NMDARs. In this new model of zinc regulation, the postsynaptic transporter ZnT1 mediates zinc inhibition of NMDARs by binding to GluN2A. Through this interaction, ZnT1, a transporter that moves zinc from the cytoplasm to the extracellular domain, generates a zinc microdomain that modulates NMDAR-mediated neurotransmission. As ZnT1 expression is transcriptionally driven by the metal-responsive transcription factor 1 (MTF-1), we found that intracellular zinc strongly drives MTF-1 in cortical neurons in vitro and increases the number of GluN2A-ZnT1 interactions, thereby enhancing tonic zinc inhibition of NMDAR-mediated currents. Importantly, this effect is absent when the interaction between GluN2A and ZnT1 is disrupted by a cell-permeable peptide. These results suggest that zinc-regulated gene expression can dynamically regulate NMDAR-mediated synaptic processes.
Collapse
Affiliation(s)
- Rebecca Krall
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Hearing Research Center, University of Pittsburgh, PA, USA
| | - Jenna R Gale
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline M Ross
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, University of Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Hearing Research Center, University of Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Damon LJ, Aaron J, Palmer AE. Single molecule microscopy to profile the effect of zinc status on transcription factor dynamics. Sci Rep 2022; 12:17789. [PMID: 36273101 PMCID: PMC9588069 DOI: 10.1038/s41598-022-22634-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 10/18/2022] [Indexed: 01/19/2023] Open
Abstract
The regulation of transcription is a complex process that involves binding of transcription factors (TFs) to specific sequences, recruitment of cofactors and chromatin remodelers, assembly of the pre-initiation complex and recruitment of RNA polymerase II. Increasing evidence suggests that TFs are highly dynamic and interact only transiently with DNA. Single molecule microscopy techniques are powerful approaches for tracking individual TF molecules as they diffuse in the nucleus and interact with DNA. Here we employ multifocus microscopy and highly inclined laminated optical sheet microscopy to track TF dynamics in response to perturbations in labile zinc inside cells. We sought to define whether zinc-dependent TFs sense changes in the labile zinc pool by determining whether their dynamics and DNA binding can be modulated by zinc. We used fluorescently tagged versions of the glucocorticoid receptor (GR), with two C4 zinc finger domains, and CCCTC-binding factor (CTCF), with eleven C2H2 zinc finger domains. We found that GR was largely insensitive to perturbations of zinc, whereas CTCF was significantly affected by zinc depletion and its dwell time was affected by zinc elevation. These results indicate that at least some transcription factors are sensitive to zinc dynamics, revealing a potential new layer of transcriptional regulation.
Collapse
Affiliation(s)
- Leah J. Damon
- grid.266190.a0000000096214564Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303 USA
| | - Jesse Aaron
- grid.443970.dAdvanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147 USA
| | - Amy E. Palmer
- grid.266190.a0000000096214564Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303 USA
| |
Collapse
|
22
|
Zhang SY, Liang JJ, Liu YQ. Excessive Zinc Ion Caused PC12 Cell Death Correlating with Inhibition of NOS and Increase of RAGE in Cells. Cell Biochem Biophys 2022; 80:755-761. [PMID: 36068383 DOI: 10.1007/s12013-022-01093-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 08/24/2022] [Indexed: 11/03/2022]
Abstract
Zinc ion (Zn2+) is an important functional factor; however, excessive Zn2+ can be toxic. To understand the neurotoxicity of excessive Zn2+ and the underlying mechanism, PC12 cells were treated with excessive Zn2+ and Zn2+ plus N, N, N', N'-Tetrakisethylenediamine (TPEN), a zinc ion chelator agent. Trypan blue and 3-(4,5-dimethyl-2- thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, thiazolyl blue tetrazolium bromide (MTT) assays were used to test cell viability; the relative kits were used to detect the activity of NOS synthase and the content of the receptor for advanced glycation end product (RAGE) in cells. We observed that excessive zinc caused PC12 cell damage and that TPEN partially reversed cell damage caused by excessive zinc. In addition, excessive zinc decreased total nitric oxide synthase (TNOS) activity in cells, in which constitutive nitric oxide synthase (cNOS) activity was significantly reduced; however, inducible nitric oxide synthase (iNOS) activity was extremely promoted. Moreover, excessive zinc upregulated the expression of RAGE, and TPEN effectively reversed the increase in RAGE induced by excessive zinc ions. Therefore, we concluded that excessive zinc caused PC12 cell damage, correlating with the inhibition of NOS and increase of RAGE induced in cells.
Collapse
Affiliation(s)
- Sai-Ya Zhang
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jing-Jing Liang
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yan-Qiang Liu
- College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
23
|
Zhang C, Dischler A, Glover K, Qin Y. Neuronal signalling of zinc: from detection and modulation to function. Open Biol 2022; 12:220188. [PMID: 36067793 PMCID: PMC9448499 DOI: 10.1098/rsob.220188] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Zinc is an essential trace element that stabilizes protein structures and allosterically modulates a plethora of enzymes, ion channels and neurotransmitter receptors. Labile zinc (Zn2+) acts as an intracellular and intercellular signalling molecule in response to various stimuli, which is especially important in the central nervous system. Zincergic neurons, characterized by Zn2+ deposits in synaptic vesicles and presynaptic Zn2+ release, are found in the cortex, hippocampus, amygdala, olfactory bulb and spinal cord. To provide an overview of synaptic Zn2+ and intracellular Zn2+ signalling in neurons, the present paper summarizes the fluorescent sensors used to detect Zn2+ signals, the cellular mechanisms regulating the generation and buffering of Zn2+ signals, as well as the current perspectives on their pleiotropic effects on phosphorylation signalling, synapse formation, synaptic plasticity, as well as sensory and cognitive function.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Anna Dischler
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Kaitlyn Glover
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Yan Qin
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| |
Collapse
|
24
|
Zinc in Cognitive Impairment and Aging. Biomolecules 2022; 12:biom12071000. [PMID: 35883555 PMCID: PMC9312494 DOI: 10.3390/biom12071000] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
Zinc, an essential micronutrient for life, was first discovered in 1869 and later found to be indispensable for the normal development of plants and for the normal growth of rats and birds. Zinc plays an important role in many physiological and pathological processes in normal mammalian brain development, especially in the development of the central nervous system. Zinc deficiency can lead to neurodegenerative diseases, mental abnormalities, sleep disorders, tumors, vascular diseases, and other pathological conditions, which can cause cognitive impairment and premature aging. This study aimed to review the important effects of zinc and zinc-associated proteins in cognitive impairment and aging, to reveal its molecular mechanism, and to highlight potential interventions for zinc-associated aging and cognitive impairments.
Collapse
|
25
|
Ha JH, Prela O, Carpizo DR, Loh SN. p53 and Zinc: A Malleable Relationship. Front Mol Biosci 2022; 9:895887. [PMID: 35495631 PMCID: PMC9043292 DOI: 10.3389/fmolb.2022.895887] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
A large percentage of transcription factors require zinc to bind DNA. In this review, we discuss what makes p53 unique among zinc-dependent transcription factors. The conformation of p53 is unusually malleable: p53 binds zinc extremely tightly when folded, but is intrinsically unstable in the absence of zinc at 37°C. Whether the wild-type protein folds in the cell is largely determined by the concentration of available zinc. Consequently, zinc dysregulation in the cell as well as a large percentage of tumorigenic p53 mutations can cause p53 to lose zinc, misfold, and forfeit its tumor suppressing activity. We highlight p53’s noteworthy biophysical properties that give rise to its malleability and how proper zinc binding can be restored by synthetic metallochaperones to reactivate mutant p53. The activity and mechanism of metallochaperones are compared to those of other mutant p53-targeted drugs with an emphasis on those that have reached the clinical trial stage.
Collapse
Affiliation(s)
- Jeung-Hoi Ha
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Orjola Prela
- Division of Surgical Oncology, Department of Surgery, Wilmot Cancer Center, University of Rochester, Rochester, NY, United States
| | - Darren R Carpizo
- Division of Surgical Oncology, Department of Surgery, Wilmot Cancer Center, University of Rochester, Rochester, NY, United States
| | - Stewart N Loh
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
26
|
Upmanyu N, Jin J, Emde HVD, Ganzella M, Bösche L, Malviya VN, Zhuleku E, Politi AZ, Ninov M, Silbern I, Leutenegger M, Urlaub H, Riedel D, Preobraschenski J, Milosevic I, Hell SW, Jahn R, Sambandan S. Colocalization of different neurotransmitter transporters on synaptic vesicles is sparse except for VGLUT1 and ZnT3. Neuron 2022; 110:1483-1497.e7. [PMID: 35263617 DOI: 10.1016/j.neuron.2022.02.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/08/2022] [Accepted: 02/10/2022] [Indexed: 12/26/2022]
Abstract
Vesicular transporters (VTs) define the type of neurotransmitter that synaptic vesicles (SVs) store and release. While certain mammalian neurons release multiple transmitters, it is not clear whether the release occurs from the same or distinct vesicle pools at the synapse. Using quantitative single-vesicle imaging, we show that a vast majority of SVs in the rodent brain contain only one type of VT, indicating specificity for a single neurotransmitter. Interestingly, SVs containing dual transporters are highly diverse (27 types) but small in proportion (2% of all SVs), excluding the largest pool that carries VGLUT1 and ZnT3 (34%). Using VGLUT1-ZnT3 SVs, we demonstrate that the transporter colocalization influences the SV content and synaptic quantal size. Thus, the presence of diverse transporters on the same vesicle is bona fide, and depending on the VT types, this may act to regulate neurotransmitter type, content, and release in space and time.
Collapse
Affiliation(s)
- Neha Upmanyu
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Jialin Jin
- European Neurosciences Institute, A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society, Göttingen 37077, Germany
| | - Henrik von der Emde
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Marcelo Ganzella
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Leon Bösche
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Viveka Nand Malviya
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Evi Zhuleku
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Antonio Zaccaria Politi
- Live-Cell Imaging Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Momchil Ninov
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Marcel Leutenegger
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Dietmar Riedel
- Department of Structural Dynamics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Julia Preobraschenski
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen 37075, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen 37075, Germany
| | - Ira Milosevic
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7BN, UK; Multidisciplinary Institute of Ageing, MIA-Portugal, University of Coimbra, Coimbra 3000-370, Portugal
| | - Stefan W Hell
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg 69028, Germany
| | - Reinhard Jahn
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Sivakumar Sambandan
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany.
| |
Collapse
|
27
|
Abstract
The functions, purposes, and roles of metallothioneins have been the subject of speculations since the discovery of the protein over 60 years ago. This article guides through the history of investigations and resolves multiple contentions by providing new interpretations of the structure-stability-function relationship. It challenges the dogma that the biologically relevant structure of the mammalian proteins is only the one determined by X-ray diffraction and NMR spectroscopy. The terms metallothionein and thionein are ambiguous and insufficient to understand biological function. The proteins need to be seen in their biological context, which limits and defines the chemistry possible. They exist in multiple forms with different degrees of metalation and types of metal ions. The homoleptic thiolate coordination of mammalian metallothioneins is important for their molecular mechanism. It endows the proteins with redox activity and a specific pH dependence of their metal affinities. The proteins, therefore, also exist in different redox states of the sulfur donor ligands. Their coordination dynamics allows a vast conformational landscape for interactions with other proteins and ligands. Many fundamental signal transduction pathways regulate the expression of the dozen of human metallothionein genes. Recent advances in understanding the control of cellular zinc and copper homeostasis are the foundation for suggesting that mammalian metallothioneins provide a highly dynamic, regulated, and uniquely biological metal buffer to control the availability, fluctuations, and signaling transients of the most competitive Zn(II) and Cu(I) ions in cellular space and time.
Collapse
Affiliation(s)
- Artur Krężel
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Wrocław 50-383, Poland
| | - Wolfgang Maret
- Departments of Biochemistry and Nutritional Sciences, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9NH, U.K
| |
Collapse
|
28
|
Wang D, Jin MF, Li L, Liu Y, Sun Y, Ni H. PRG5 Knockout Precipitates Late-Onset Hypersusceptibility to Pilocarpine-Induced Juvenile Seizures by Exacerbating Hippocampal Zinc Signaling-Mediated Mitochondrial Damage. Front Neurosci 2021; 15:715555. [PMID: 34512249 PMCID: PMC8430038 DOI: 10.3389/fnins.2021.715555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Epileptogenesis is understood as the plastic process that produces a persistent reorganization of the brain’s neural network after a precipitating injury (recurrent neonatal seizures, for instance) with a latent period, finally leading to neuronal hyperexcitability. Plasticity-related genes (PRGs), also known as lipid phosphate phosphatase-related proteins (PLPPRs), are regulators of mitochondrial membrane integrity and energy metabolism. This study was undertaken to determine whether PRG5 gene knockout contributes to the delayed hypersensitivity induced by developmental seizures and the aberrant sprouting of hippocampal mossy fibers, and to determine whether it is achieved through the mitochondrial pathway. Here, we developed a “twist” seizure model by coupling pilocarpine-induced juvenile seizures with later exposure to penicillin to test the long-term effects of PRG5 knockout on seizure latency through comparison with wild-type (WT) mice. Hippocampal mossy fiber sprouting (MFS) was detected by Timm staining. In order to clarify the mechanism of the adverse reactions triggered by PRG5 knockout, hippocampal HT22 neuronal cultures were exposed to glutamate, with or without PRG5 interference. Mitochondrial function, oxidative stress indicators and zinc ion content were detected. Results PRG5 gene knockout significantly reduced the seizure latency, and aggravated the lowered seizure threshold induced by developmental seizures. Besides, knockout of the PRG5 gene reduced the MFS scores to a certain extent. Furthermore, PRG5 gene silencing significantly increases the zinc ion content in hippocampal neurons, impairs neuronal activity and mitochondrial function, and exacerbates glutamate-induced oxidative stress damage. Conclusion In summary, PRG5 KO is associated with significantly greater hypersusceptibility to juvenile seizures in PRG5(–/–) mice compared with WT mice. These effects may be related to the hippocampal zinc signaling. The effects do not appear to be related to changes in MFS because KO mice with juvenile seizures had the shortest seizure latencies but exhibited less MFS than WT mice with juvenile seizures.
Collapse
Affiliation(s)
- Dandan Wang
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.,Department of Pediatrics, First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Mei-Fang Jin
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Lili Li
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yueying Liu
- Department of Pediatrics, North Branch, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuxiao Sun
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Hong Ni
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
29
|
Abstract
Zinc (Zn2+) is an essential metal in biology, and its bioavailability is highly regulated. Many cell types exhibit fluctuations in Zn2+ that appear to play an important role in cellular function. However, the detailed molecular mechanisms by which Zn2+ dynamics influence cell physiology remain enigmatic. Here, we use a combination of fluorescent biosensors and cell perturbations to define how changes in intracellular Zn2+ impact kinase signaling pathways. By simultaneously monitoring Zn2+ dynamics and kinase activity in individual cells, we quantify changes in labile Zn2+ and directly correlate changes in Zn2+ with ERK and Akt activity. Under our experimental conditions, Zn2+ fluctuations are not toxic and do not activate stress-dependent kinase signaling. We demonstrate that while Zn2+ can nonspecifically inhibit phosphatases leading to sustained kinase activation, ERK and Akt are predominantly activated via upstream signaling and through a common node via Ras. We provide a framework for quantification of Zn2+ fluctuations and correlate these fluctuations with signaling events in single cells to shed light on the role that Zn2+ dynamics play in healthy cell signaling.
Collapse
|
30
|
Isaev NK, Stelmashook EV, Genrikhs EE. Role of zinc and copper ions in the pathogenetic mechanisms of traumatic brain injury and Alzheimer's disease. Rev Neurosci 2021; 31:233-243. [PMID: 31747384 DOI: 10.1515/revneuro-2019-0052] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/24/2019] [Indexed: 12/24/2022]
Abstract
The disruption of homeostasis of zinc (Zn2+) and copper (Cu2+) ions in the central nervous system is involved in the pathogenesis of many neurodegenerative diseases, such as amyotrophic lateral sclerosis, Wilson's, Creutzfeldt-Jakob, Parkinson's, and Alzheimer's diseases (AD), and traumatic brain injury (TBI). The last two pathological conditions of the brain are the most common; moreover, it is possible that TBI is a risk factor for the development of AD. Disruptions of Zn2+ and Cu2+ homeostasis play an important role in the mechanisms of pathogenesis of both TBI and AD. This review attempts to summarize and systematize the currently available research data on this issue. The neurocytotoxicity of Cu2+ and Zn2+, the synergism of the toxic effect of calcium and Zn2+ ions on the mitochondria of neurons, and the interaction of Zn2+ and Cu2+ with β-amyloid (Abeta) and tau protein are considered.
Collapse
Affiliation(s)
- Nickolay K Isaev
- M.V. Lomonosov Moscow State University, N.A. Belozersky Institute of Physico-Chemical Biology, Biological Faculty, Moscow 119991, Russia.,Research Center of Neurology, Moscow 125367, Russia
| | | | | |
Collapse
|
31
|
The Multifaceted Roles of Zinc in Neuronal Mitochondrial Dysfunction. Biomedicines 2021; 9:biomedicines9050489. [PMID: 33946782 PMCID: PMC8145363 DOI: 10.3390/biomedicines9050489] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/17/2022] Open
Abstract
Zinc is a highly abundant cation in the brain, essential for cellular functions, including transcription, enzymatic activity, and cell signaling. However, zinc can also trigger injurious cascades in neurons, contributing to the pathology of neurodegenerative diseases. Mitochondria, critical for meeting the high energy demands of the central nervous system (CNS), are a principal target of the deleterious actions of zinc. An increasing body of work suggests that intracellular zinc can, under certain circumstances, contribute to neuronal damage by inhibiting mitochondrial energy processes, including dissipation of the mitochondrial membrane potential (MMP), leading to ATP depletion. Additional consequences of zinc-mediated mitochondrial damage include reactive oxygen species (ROS) generation, mitochondrial permeability transition, and excitotoxic calcium deregulation. Zinc can also induce mitochondrial fission, resulting in mitochondrial fragmentation, as well as inhibition of mitochondrial motility. Here, we review the known mechanisms responsible for the deleterious actions of zinc on the organelle, within the context of neuronal injury associated with neurodegenerative processes. Elucidating the critical contributions of zinc-induced mitochondrial defects to neurotoxicity and neurodegeneration may provide insight into novel therapeutic targets in the clinical setting.
Collapse
|
32
|
Krall RF, Tzounopoulos T, Aizenman E. The Function and Regulation of Zinc in the Brain. Neuroscience 2021; 457:235-258. [PMID: 33460731 DOI: 10.1016/j.neuroscience.2021.01.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/31/2022]
Abstract
Nearly sixty years ago Fredrich Timm developed a histochemical technique that revealed a rich reserve of free zinc in distinct regions of the brain. Subsequent electron microscopy studies in Timm- stained brain tissue found that this "labile" pool of cellular zinc was highly concentrated at synaptic boutons, hinting a possible role for the metal in synaptic transmission. Although evidence for activity-dependent synaptic release of zinc would not be reported for another twenty years, these initial findings spurred decades of research into zinc's role in neuronal function and revealed a diverse array of signaling cascades triggered or regulated by the metal. Here, we delve into our current understanding of the many roles zinc plays in the brain, from influencing neurotransmission and sensory processing, to activating both pro-survival and pro-death neuronal signaling pathways. Moreover, we detail the many mechanisms that tightly regulate cellular zinc levels, including metal binding proteins and a large array of zinc transporters.
Collapse
Affiliation(s)
- Rebecca F Krall
- Department of Neurobiology, University of Pittsburgh School of Medicine, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, USA
| | - Thanos Tzounopoulos
- Department of Otolaryngology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA.
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, USA.
| |
Collapse
|
33
|
Pratt EPS, Damon LJ, Anson KJ, Palmer AE. Tools and techniques for illuminating the cell biology of zinc. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118865. [PMID: 32980354 DOI: 10.1016/j.bbamcr.2020.118865] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Zinc (Zn2+) is an essential micronutrient that is required for a wide variety of cellular processes. Tools and methods have been instrumental in revealing the myriad roles of Zn2+ in cells. This review highlights recent developments fluorescent sensors to measure the labile Zn2+ pool, chelators to manipulate Zn2+ availability, and fluorescent tools and proteomics approaches for monitoring Zn2+-binding proteins in cells. Finally, we close with some highlights on the role of Zn2+ in regulating cell function and in cell signaling.
Collapse
Affiliation(s)
- Evan P S Pratt
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America
| | - Leah J Damon
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America
| | - Kelsie J Anson
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America
| | - Amy E Palmer
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America.
| |
Collapse
|
34
|
Kocyła A, Tran JB, Krężel A. Galvanization of Protein-Protein Interactions in a Dynamic Zinc Interactome. Trends Biochem Sci 2020; 46:64-79. [PMID: 32958327 DOI: 10.1016/j.tibs.2020.08.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/10/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
The presence of Zn2+ at protein-protein interfaces modulates complex function, stability, and introduces structural flexibility/complexity, chemical selectivity, and reversibility driven in a Zn2+-dependent manner. Recent studies have demonstrated that dynamically changing Zn2+ affects numerous cellular processes, including protein-protein communication and protein complex assembly. How Zn2+-involved protein-protein interactions (ZPPIs) are formed and dissociate and how their stability and reactivity are driven in a zinc interactome remain poorly understood, mostly due to experimental obstacles. Here, we review recent research advances on the role of Zn2+ in the formation of interprotein sites, their architecture, function, and stability. Moreover, we underline the importance of zinc networks in intersystemic communication and highlight bioinformatic and experimental challenges required for the identification and investigation of ZPPIs.
Collapse
Affiliation(s)
- Anna Kocyła
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Józef Ba Tran
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Artur Krężel
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland.
| |
Collapse
|
35
|
Copley SD. The physical basis and practical consequences of biological promiscuity. Phys Biol 2020; 17:10.1088/1478-3975/ab8697. [PMID: 32244231 PMCID: PMC9291633 DOI: 10.1088/1478-3975/ab8697] [Citation(s) in RCA: 13] [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
Proteins interact with metabolites, nucleic acids, and other proteins to orchestrate the myriad catalytic, structural and regulatory functions that support life from the simplest microbes to the most complex multicellular organisms. These molecular interactions are often exquisitely specific, but never perfectly so. Adventitious "promiscuous" interactions are ubiquitous due to the thousands of macromolecules and small molecules crowded together in cells. Such interactions may perturb protein function at the molecular level, but as long as they do not compromise organismal fitness, they will not be removed by natural selection. Although promiscuous interactions are physiologically irrelevant, they are important because they can provide a vast reservoir of potential functions that can provide the starting point for evolution of new functions, both in nature and in the laboratory.
Collapse
Affiliation(s)
- Shelley D Copley
- Department of Molecular, Cellular and Developmental Biology and Cooperative Institute for Research in Environmental Sciences, University of Colorado Boulder, Boulder, Colorado, UNITED STATES
| |
Collapse
|
36
|
Bird AJ, Wilson S. Zinc homeostasis in the secretory pathway in yeast. Curr Opin Chem Biol 2020; 55:145-150. [PMID: 32114317 DOI: 10.1016/j.cbpa.2020.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/14/2019] [Accepted: 01/26/2020] [Indexed: 01/20/2023]
Abstract
It is estimated that up to 10% of proteins in eukaryotes require zinc for their function. Although the majority of these proteins are located in the nucleus and cytosol, a small subset is secreted from cells or is located within an intracellular compartment. As many of these compartmentalized metalloproteins fold to their native state and bind their zinc cofactor inside an organelle, cells require mechanisms to maintain supply of zinc to these compartments even under conditions of zinc deficiency. At the same time, intracellular compartments can also be the site for storing zinc ions, which then can be mobilized when needed. In this review, we highlight insight that has been obtained from yeast models about how zinc homeostasis is maintained in the secretory pathway and vacuole.
Collapse
Affiliation(s)
- Amanda J Bird
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, 43210, USA; Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA; Center for RNA Biology, The Ohio State University, Columbus, OH, 43210, USA.
| | - Stevin Wilson
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
37
|
Sanford L, Palmer AE. Dissociated Hippocampal Neurons Exhibit Distinct Zn 2+ Dynamics in a Stimulation-Method-Dependent Manner. ACS Chem Neurosci 2020; 11:508-514. [PMID: 32013397 PMCID: PMC7251562 DOI: 10.1021/acschemneuro.0c00006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Ionic Zn2+ has increasingly been recognized as an important neurotransmitter and signaling ion in glutamatergic neuron pathways. Intracellular Zn2+ transiently increases as a result of neuronal excitation, and this Zn2+ signal is essential for neuron plasticity, but the source and regulation of the signal is still unclear. In this study, we rigorously quantified Zn2+, Ca2+, and pH dynamics in dissociated mouse hippocampal neurons stimulated with bath application of high KCl or glutamate. While both stimulation methods yielded Zn2+ signals, Ca2+ influx, and acidification, glutamate stimulation induced more sustained high intracellular Ca2+ and a larger increase in intracellular Zn2+. However, the stimulation-induced pH change was similar between conditions, indicating that a different cellular change is responsible for the stimulation-dependent difference in Zn2+ signal. This work provides the first robust quantification of Zn2+ dynamics in neurons using different methods of stimulation.
Collapse
Affiliation(s)
- Lynn Sanford
- Department of Biochemistry, BioFrontiers Institute , University of Colorado Boulder , Boulder , Colorado 80309 , United States
| | - Amy E Palmer
- Department of Biochemistry, BioFrontiers Institute , University of Colorado Boulder , Boulder , Colorado 80309 , United States
| |
Collapse
|
38
|
Hanif M, Rafiq M, Yousuf M, Kotwica-Mojzych K, Saleem M, Mojzych M. Organic small molecular receptors as fluorimetric/bioimaging probe for extracellular/intracellular zinc sensation. Bioorg Chem 2019; 94:103398. [PMID: 31679837 DOI: 10.1016/j.bioorg.2019.103398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/01/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Muhammad Hanif
- Department of Chemistry, GC University Faisalabad, Sub Campus Layyah, Pakistan
| | - Muhammad Rafiq
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Muhammad Yousuf
- Department of Theriogenology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Katarzyna Kotwica-Mojzych
- Department of Histology and Embryology with Experimental Cytology Unit, Medical University of Lublin, Radziwiłłowska 11, (Collegium Medicum), 20-080 Lublin, Poland
| | - Muhammad Saleem
- Department of Chemistry, University of Sargodha, Sub-campus Bhakkar, Pakistan.
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, 3-go Maja 54, 08-110 Siedlce, Poland.
| |
Collapse
|