1
|
Zhang Q, Dou S, Leng H, Shu Y. A small molecule modified UiO series MOFs for simultaneous detection of Fe 3+ and Zn 2. Talanta 2025; 286:127483. [PMID: 39733522 DOI: 10.1016/j.talanta.2024.127483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Iron and zinc are two metal ions with important roles in biology, industry and the environment, however, the excess or deficiency of both Fe3+ and Zn2+ can have negative effects on organisms and environment. Therefore, the development of efficient method for simultaneous detection of Fe3+ and Zn2+ provides timely information on metal content, simplifies operations and improves efficiency. In this work, a small molecule (COOH-BPEA) of recognizing Zn2+ modified the four metal-organic-framework (MOF) (UiO-66-X(66, OH, NH2 and OH/NH2)) was developed for the simultaneous detection of Fe3+ and Zn2+. The fluorescence signal of the small molecule is enhanced by small molecule chelating Zn2+ to block the photoinduced electron transfer (PET) effect. The fluorescence signals of the UiO series MOFs were quenched through Fe3+ with electron transfer and static quenching effect (SQE). It's worth mentioning that the emission wavelengths of the small molecules and MOFs did not interfere with each other. The UiO-66-NH2@BPEA with optimal performance was selected by fluorescence spectra for the detection of Fe3+ and Zn2+ with detection limit of 0.175 μM and 0.021 μM, respectively. The nanoprobe provides a fast response (less than 1 min) for both Fe3+ and Zn2+. Finally, we applied it to the simultaneous detection of Fe3+ and Zn2+ in environmental water, human serum and cell lysates.
Collapse
Affiliation(s)
- Qikun Zhang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Shuaihua Dou
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Han Leng
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Yang Shu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China.
| |
Collapse
|
2
|
Li XF, Wu FG. Aggregation-induced emission-based fluorescent probes for cellular microenvironment detection. Biosens Bioelectron 2025; 274:117130. [PMID: 39904094 DOI: 10.1016/j.bios.2025.117130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/27/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025]
Abstract
The cellular microenvironment exerts a pivotal regulatory influence on cell survival, function, and behavior. Dynamic analysis and detection of the cellular microenvironment can promptly elucidate changes in cellular microenvironmental information, uncover the pathogenesis of diseases associated with aberrant microenvironments, and aid in predicting disease risk and monitoring disease progression. Aggregation-induced emission (AIE) fluorescent molecules possess unique AIE characteristics and offer significant advantages in imaging and sensing cellular microenvironments. In this review, we present a profile of the remarkable progress achieved in utilizing AIE fluorescent molecules for detecting cellular microenvironments in recent years. We particularly focus on AIE fluorescent probes applied in imaging key parameters of the cellular microenvironment, including pH, viscosity, polarity, and temperature, as well as in analyzing critical biological components of the microenvironment, such as gas signal molecules, metal ions, redox state, and proteins. We underscore the design principles, detection mechanisms, sensing performance, and biological applications of these fluorescent probes. Furthermore, we address the current challenges confronting this field and provide prospects for the future development of AIE probes used for microenvironment detection. We trust that this review will inspire researchers to develop more precise and sensitive AIE fluorescent probes for the detection of cellular microenvironments.
Collapse
Affiliation(s)
- Xiang-Fei Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, 211189, China.
| |
Collapse
|
3
|
Kang J, Liao P, Xiang R, Liao W, Yang C, Wang S, Liu Q, Li G. Interfacial Asymmetrically Coordinated Zn-MOF for High-Efficiency Electrosynthetic Oxime. Angew Chem Int Ed Engl 2025; 64:e202419550. [PMID: 39981894 DOI: 10.1002/anie.202419550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Indexed: 02/22/2025]
Abstract
Oximes are important intermediates for various chemicals synthesis such as pharmaceuticals, among which one vital precursor for producing neurological disease, antimicrobial and anticancer agents is piperidone oxime (PDO). Compared with conventional thermocatalytic method, it's more attractive to synthesize PDO via green electrocatalytic technology especially utilizing waste nitrogen oxides gas as nitrogen source. However, there are great challenges in catalyst design for high-efficiency electrosynthetic oxime due to the low electron transport rate and multiple competing reactions. Herein, we propose an interfacial coordination strategy based on metal-organic frameworks (MOF) electrocatalyst for the first time to promote oxime electrosynthesis, by building Zn-O bridges between graphite felt (GF) and zeolitic imidazolate framework (ZIF-7/CGF). Specially, ZIF-7/CGF delivers a Faraday efficiency (FE) of 75.9 % with yield up to 73.1 % for 1-methyl-4-piperidone oxime, which is far superior to the catalyst without Zn-O bridges (a FE of 10.7 % and yield of 10.3 %). In-depth mechanism study shows that the introducing Zn-O bridges can promote the electron transfer and induce Zn sites transforming into distorted tetrahedron (Zn-N3O) coordination mode, which benefits for intermediates adsorption and conversion. The developed strategy presents wide universalities towards various oximes electrosynthesis and adapts to other MOF materials (ZIF-8, ZIF-4). This work provides new insights for electrosynthetic organic chemicals and upgrading nitrogen cycle through rational design surficial coordinated electrocatalysts.
Collapse
Affiliation(s)
- Jiawei Kang
- Key Laboratory of Bioinorganic and Synthetic Chemistry of Ministry of Education, LIFM, IGCME, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Peisen Liao
- Key Laboratory of Bioinorganic and Synthetic Chemistry of Ministry of Education, LIFM, IGCME, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Runan Xiang
- Key Laboratory of Bioinorganic and Synthetic Chemistry of Ministry of Education, LIFM, IGCME, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Wenpei Liao
- Key Laboratory of Bioinorganic and Synthetic Chemistry of Ministry of Education, LIFM, IGCME, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Chenyu Yang
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230029, China
| | - Shihan Wang
- Key Laboratory of Bioinorganic and Synthetic Chemistry of Ministry of Education, LIFM, IGCME, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Qinghua Liu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230029, China
| | - Guangqin Li
- Key Laboratory of Bioinorganic and Synthetic Chemistry of Ministry of Education, LIFM, IGCME, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
4
|
Ding M, Chen B, Wilson DA, Tu Y, Peng F. From Autonomous Chemical Micro-/Nanomotors to Rationally Engineered Bio-Interfaces. Angew Chem Int Ed Engl 2025; 64:e202423207. [PMID: 39905915 DOI: 10.1002/anie.202423207] [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: 11/27/2024] [Indexed: 02/06/2025]
Abstract
Developing micro-/nanomotors that convert a chemical energy input into a local gradient field and motion is an appealing but challenging task that holds particular promise for the intersection of materials and nanoengineering. Over the past two decades, remarkable advancements have refined these out-of-equilibrium chemically powered micro-/nanomotors, enabling them to orchestrate in situ chemical transformations that dynamically change local environments. The ionic products, radicals, gases, and electric fields from these active materials reshape the microenvironment, paving the way for ecofriendly disease interventions. This review discusses the state-of-the-art reactions that propel these energy-consuming micro-/nanomotors and elucidates the emerging implications of their products on biological systems. Particular emphasis has been placed on their potential for neural modulation, reactive oxygen species (ROS) regulation, synergistic tumor therapy, antibacterial strategies, and tissue regeneration. Collectively, these sketches provide a landscape of therapeutic modalities, heralding a new era of biomedicine. By harnessing the in situ product field of this active matter, we envision a paradigm shift toward active therapies that transcend conventional approaches, promising breakthroughs in disease diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Miaomiao Ding
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Bin Chen
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Daniela A Wilson
- Institute for Molecules and Materials, Radboud University, Nijmegen, 6525 AJ, The Netherland
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| |
Collapse
|
5
|
Manning A, Mendelson BZ, Bender PTR, Bainer K, Ruby R, Shifflett VR, Dariano DF, Webb BA, Geldenhuys WJ, Anderson CT. The Astrocytic Zinc Transporter ZIP12 Is a Synaptic Protein That Contributes to Synaptic Zinc Levels in the Mouse Auditory Cortex. J Neurosci 2025; 45:e2067242025. [PMID: 39809542 PMCID: PMC11949477 DOI: 10.1523/jneurosci.2067-24.2025] [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: 10/30/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Synaptically released zinc is a neuronal signaling system that arises from the actions of the presynaptic vesicular zinc transporter protein zinc transporter 3 (ZnT3). Mechanisms that regulate the actions of zinc at synapses are of great importance for many aspects of synaptic signaling in the brain. Here, we identify the astrocytic zinc transporter protein ZIP12 as a candidate mechanism that contributes to zinc clearance at cortical synapses. We identify small-molecule compounds that antagonize the function of ZIP12 in heterologous expression systems, and we use one of these compounds, ZIP12 modulator 8, to increase the concentration of ZnT3-dependent zinc at synapses in the brain of male and female mice to inhibit the activity of neuronal AMPA and NMDA glutamate receptors. These results identify a cellular mechanism and provide a pharmacological toolbox to target the molecular machinery that supports the actions of synaptic zinc in the brain.
Collapse
Affiliation(s)
- Abbey Manning
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Benjamin Z Mendelson
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Philip T R Bender
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Kaitlin Bainer
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Rayli Ruby
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Victoria R Shifflett
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Donald F Dariano
- Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Bradley A Webb
- Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| | - Werner J Geldenhuys
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, West Virginia 26506
| | - Charles T Anderson
- Departments of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506
| |
Collapse
|
6
|
Wang J, Wang W, Zheng G, Shi F, Wu S, Zhang Y. Associations of mixed metals exposure with cognitive impairment risk: a cross-sectional study in Chinese adults. Postgrad Med J 2025; 101:321-329. [PMID: 39484828 DOI: 10.1093/postmj/qgae154] [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: 04/27/2024] [Revised: 09/01/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Associations between exposure to single metals and cognitive impairment or related outcomes have been reported in many previous studies. However, co-exposure to more than one metal is common situation. In recent years, studies on the effects of exposure to multiple metals on cognitive impairment or related outcomes have increased, but remain very limited, with a focus on populations with occupational exposure to metals, children, and adolescents. The potential relationships between exposure to metal mixtures and risk of cognitive impairment in adults remain to be clarified. OBJECTIVE To determine the associations between blood metal mixtures and cognitive impairment risk. METHODS Inductively coupled plasma mass spectrometry (ICP-MS) was utilized to detect the blood levels of lead (Pb), iron (Fe), copper (Cu), calcium (Ca), magnesium (Mg), and zinc (Zn). Multivariable logistic regression and Bayesian kernel machine regression (BKMR) models were employed to assess the relationships of exposure to these blood metal mixtures with the risk of cognitive impairment. RESULTS It was found that four metals (Pb, Fe, Cu, and Mg) were positively correlated with cognitive impairment in each single metal model. The association of Pb and Cu remained significant after adjusting for these six metals, with the odds ratios (95% confidence intervals) in the highest quartiles of 9.51 (4.41-20.54, p-trend <0.01) and 4.87 (2.17-10.95, p-trend <0.01), respectively. The BKMR models indicated that co-exposure levels of Ca, Cu, Fe, Mg, Pb, and Zn were related to increased risk of cognitive impairment at ≥25th percentile compared with median, and Pb and Cu mainly contributed to the joint effect. In addition, the interaction effects of Mg and Pb/Pb and Cu on the risk of cognitive impairment were observed. SIGNIFICANCE Co-exposure of six metals (Pb, Fe, Cu, Ca, Mg, and Zn) increased the risk of cognitive impairment in Chinese adults, with Pb and Cu likely to have greater impact. Potential interaction effects of Mg and Pb, Pb and Cu on the risk of cognitive impairment may exist.
Collapse
Affiliation(s)
- Jierui Wang
- School of Public Health, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
- Department of Rheumatic Disease, Kailuan General Hospital, 57 Xinhua East Road, Tangshan, Hebei 063000, China
| | - Weixuan Wang
- The Laboratory Animal Center, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Gang Zheng
- Department of Occupational and Environmental Health, School of Military Preventive Medicine, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Fan Shi
- School of Public Health, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, 57 Xinhua East Road, Tangshan, Hebei 063000, China
| | - Yanshu Zhang
- The Laboratory Animal Center, North China University of Science and Technology, 21 Bohai Road, Cao Fei Dian, Tangshan, Hebei 063210, China
| |
Collapse
|
7
|
Krinochkin A, Valieva M, Starnovskaya E, Slovesnova N, Minin A, Belousova A, Sadieva L, Taniya O, Khasanov A, Novikov A, Bruskov V, Vatolina S, Kopchuk D, Slepukhin P, Sharutin V, Zyryanov G. New Fluorescent Dye for the Detection of Zn 2+ in Living Cells and Fixed Sections of the Rat Pancreas. J Fluoresc 2025; 35:1423-1439. [PMID: 38349481 DOI: 10.1007/s10895-024-03603-1] [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: 11/20/2023] [Accepted: 01/23/2024] [Indexed: 04/04/2025]
Abstract
We report the synthesis and characterization of a new 4-methoxyphenyl-2,2'-bipyridine-based ligand, such as 12, bearing dipicolylaminomethyl core as a receptor unit, as a probe for the fluorescence "turn-on" detection of Zn2+. Thus, in the presence of Zn2+ the probe 12 exhibited a fluorescence enhancement with a Stokes shift of ~ 180 nm and photoluminescence quantum yields value of ~ 1.0. In addition, 12 exhibited higher binding constant for Zn2+ (~ 2 × 105 M-1) with the LOD reaching the nanomolar level (~ 0.1 × 10-9 M) compare to the previously reported probe 1. The stoichiometry and structure of the [Zn(12)]2+ and [Zn(1)]2+ complexes were supported by XRD analysis, DFT calculations and 1H NMR experiments. It was postulated that, as a result of binding of Zn2+, the sample exhibited a bright "on" state via the PET-ICT processes. Molecular docking studies and confocal fluorescence microscopy experiments demonstrated that the probe 12 could be used for the fluorescence detection of Zn2+ not only in artificially enriched with zinc salts live cells, but also in fixed tissues with cations are in a bound state. The high binding constant of compound 12 to Zn2+ cation allows it to be used for the accurate localization of pancreatic beta cells (islets of Langerhans).
Collapse
Affiliation(s)
- Alexey Krinochkin
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Branch of the RAS, 22 S. Kovalevskoy Street, 620219, Yekaterinburg, Russia
| | - Maria Valieva
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Branch of the RAS, 22 S. Kovalevskoy Street, 620219, Yekaterinburg, Russia
| | | | - Nataliya Slovesnova
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
- Urals State Medical University, 3 Repina Street, 620028, Yekaterinburg, Russia
| | - Artem Minin
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
- M.N. Miheev Institute of Metal Physics, Ural Branch of the RAS, 18 S. Kovalevskoy Street, 620108, Yekaterinburg, Russia
| | - Anna Belousova
- Institute of Immunology and Physiology, Ural Branch of the RAS, 106 Pervomaiskaya Street, 620049, Yekaterinburg, Russia
| | - Leila Sadieva
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia.
| | - Olga Taniya
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
| | - Albert Khasanov
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
| | - Alexander Novikov
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., 199034, Saint Petersburg, Russia
| | - Vitaly Bruskov
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
| | - Svetlana Vatolina
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
| | - Dmitry Kopchuk
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Branch of the RAS, 22 S. Kovalevskoy Street, 620219, Yekaterinburg, Russia
| | - Pavel Slepukhin
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Branch of the RAS, 22 S. Kovalevskoy Street, 620219, Yekaterinburg, Russia
| | - Vladimir Sharutin
- Department of Chemistry, Institute of Natural Sciences, South Ural State University (National Research University), Lenin Avenue 76, 454080, Chelyabinsk, Russia
| | - Grigory Zyryanov
- Ural Federal University, 19 Mira Street, 620002, Yekaterinburg, Russia
- I. Ya. Postovskiy Institute of Organic Synthesis, Ural Branch of the RAS, 22 S. Kovalevskoy Street, 620219, Yekaterinburg, Russia
| |
Collapse
|
8
|
Maret W. The Arcana of Zinc. J Nutr 2025; 155:669-675. [PMID: 39788322 PMCID: PMC11934285 DOI: 10.1016/j.tjnut.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
This perspective discusses the essential micronutrient zinc, which functions in >3000 human proteins (the zinc proteome), and the implications of three aspects to ascertain an adequate zinc status for human health. First, the advent of highly sensitive fluorescent (bio)chemicals revealed cellular pools of zinc ions involved in signaling and secretion from cells for paracrine, autocrine, and possibly endocrine functions. Zinc signaling adds a yet unaccounted number of targeted proteins to the already impressive number of zinc proteins. Second, cellular zinc concentrations are remarkably high in the order of the concentrations of major metabolites and, therefore, at the cellular level zinc is not a trace element. Zinc is also not an antioxidant because zinc ions are redox-inactive in biology. However, zinc can express indirect pro-oxidant or proantioxidant effects depending on how cellular zinc is buffered. Zinc sites in proteins and other biomolecules can become redox-active when zinc is bound to the redox-active sulfur donor atom of cysteine. This interaction links zinc and redox metabolism, confers mobility on tightly bound zinc, and has implications for treating zinc deficiency. Third, the concept of zinc deficiency in blood as the only measure of an inadequate zinc status needs to be extended to zinc dyshomeostasis in cells because overwhelming the mechanisms controlling cellular zinc homeostasis can result in either not enough or too much available zinc. We need additional biomarkers of zinc status that determine cell-specific changes and perturbations of the system regulating cellular zinc, including functional deficits, and address the multiple genetic and environmental factors that can cause a conditioned zinc deficiency or overload. Considering the wider context of altered zinc availability in different organs, cells, and organelles impinges on whether zinc supplementation will be efficacious and adds another dimension to the already high health burden of zinc deficiency and its sequelae worldwide.
Collapse
Affiliation(s)
- Wolfgang Maret
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.
| |
Collapse
|
9
|
Kumar MS, Pakrashy S, Manna S, Choudhury SM, Das B, Ghosh A, Seikh AH, Dolai M, Das AK. Fluorogenic selective detection of Zn 2+ using a pyrazole- ortho-vanillin conjugate: insights from DFT, molecular docking, bioimaging and anticancer applications. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:2125-2133. [PMID: 39950208 DOI: 10.1039/d4ay02218a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
A fluorescent sensor, (E)-N'-(2-hydroxy-3-methoxybenzylidene)-3,5-dimethyl-1H-pyrazole-1-carbohydrazide (HMPC), was designed and synthesized for the selective fluorescence recognition of Zn2+ in semi-aqueous media. Notably, HMPC exhibited a red-shifted, two-fold fluorescence "turn-on" enhancement in response to Zn2+ at 490 nm, with a detection limit of 1.68 μM, which is significantly lower than the WHO guideline (76.0 μM). The binding constant of HMPC with Zn2+ was calculated to be 5 × 104 M-1. The fluorescence enhancement of HMPC in the presence of Zn2+ is attributed to the suppression of the PET process and the enhancement of ICT, leading to fluorescence via the CHEF mechanism. The sensing mechanism was demonstrated through UV-vis, fluorescence spectroscopy, Job plots, ESI-MS, and DFT calculations. For biological applications, cytotoxicity and cell imaging studies were performed using MCF-7 cells. Molecular docking studies revealed a high binding energy of HMPC (ΔG = -7.1 kcal mol-1) with the 4,5-diaryl isoxazole HSP90 chaperone protein, suggesting its potential as an anticancer agent. Additionally, its binding energy of -6.5 kcal mol-1 with the HDAC8 protein indicates greater efficacy than suberoylanilide hydroxamic acid (SAHA) in inhibiting HDAC, as it binds more strongly to the HDAC8 protein than SAHA (-7.4 kcal mol-1). Furthermore, due to its favorable ADME profile, HMPC may be suitable for oral administration, enhancing its potential as an anticancer drug.
Collapse
Affiliation(s)
- Malavika S Kumar
- Department of Chemistry, Christ University, Hosur Road, Bangalore, Karnataka, 560029, India.
| | - Sourav Pakrashy
- Department of Chemistry, Prabhat Kumar College, Vidyasagar University, Purba Medinipur, W. B., 721404, India.
| | - Sounik Manna
- Biochemistry, Molecular Endocrinology, and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore 721102, W. B., India
| | - Sujata Maiti Choudhury
- Biochemistry, Molecular Endocrinology, and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore 721102, W. B., India
| | - Bhriguram Das
- Department of Chemistry, Vidyasagar University, Paschim Medinipur, W. B., 721102, India
| | - Abhishek Ghosh
- Department of Applied Science, University of Quebec at Chicoutimi, Saguenay, QC, G7H 2B1, Canada
| | - Asiful H Seikh
- Department of Mechanical Engineering, College of Engineering, King Saud University, Riyadh 11421, Saudi Arabia
| | - Malay Dolai
- Department of Chemistry, Prabhat Kumar College, Vidyasagar University, Purba Medinipur, W. B., 721404, India.
| | - Avijit Kumar Das
- Department of Chemistry, Christ University, Hosur Road, Bangalore, Karnataka, 560029, India.
| |
Collapse
|
10
|
Tran NB, Lee SJ. Metallothionein-3-mediated intracellular zinc mediates antioxidant and anti-inflammatory responses in the complete Freund's adjuvant-induced inflammatory pain mouse model. Cell Death Discov 2025; 11:45. [PMID: 39904985 PMCID: PMC11794434 DOI: 10.1038/s41420-025-02322-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/03/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
Chronic inflammatory pain is often caused by peripheral tissue damage and persistent inflammation. This disease substantially affects patients' physical and social well-being. We investigated the role of metallothionein-3 (MT3) in modulating complete Freund's adjuvant (CFA)-induced intracellular Zn2+ activity in an MT3 knockout mouse model of inflammatory pain in the hind paw. The results demonstrated that increasing intracellular Zn2+ levels ameliorate deficits in motor behavior, as well as inflammation in the paw, spleen, and thymus. Furthermore, intracellular Zn2+ was crucial in regulating oxidative stress markers (glutathione, superoxide dismutase, catalase, and malondialdehyde) and inflammatory cytokines, such as tumor necrosis factor-α and interleukin-6, in MT3 knockout mice induced with CFA. This study highlights the critical role of MT3 in coordinating the intracellular interaction with Zn2+, which is vital for the immune systems's protective functions. These interactions are fundamental for maintaining metal ion homeostasis and regulating the synthesis of various biomolecules in the body.
Collapse
Affiliation(s)
- Ngoc Buu Tran
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeonbuk-do, 54896, Republic of Korea
| | - Sook-Jeong Lee
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeonbuk-do, 54896, Republic of Korea.
| |
Collapse
|
11
|
Duță C, Muscurel C, Dogaru CB, Stoian I. Selenoproteins: Zoom-In to Their Metal-Binding Properties in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:1305. [PMID: 39941073 PMCID: PMC11818150 DOI: 10.3390/ijms26031305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025] Open
Abstract
Selenoproteins contain selenium (Se), which is included in the 21st proteinogenic amino acid selenocysteine (Sec). Selenium (Se) is an essential trace element that exerts its biological actions mainly through selenoproteins. Selenoproteins have crucial roles in maintaining healthy brain activity. At the same time, brain-function-associated selenoproteins may also be involved in neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). The selenoproteins GPx4 (glutathione peroxidase 4), GPx1 (glutathione peroxidase 1), SELENOP (selenoprotein P), SELENOK (selenoprotein K), SELENOS (selenoprotein S), SELENOW (selenoprotein W), and SELENOT (selenoprotein T) are highly expressed, specifically in AD-related brain regions being closely correlated to brain function. Only a few selenoproteins, mentioned above (especially SELENOP), can bind transition and heavy metals. Metal ion homeostasis accomplishes the vital physiological function of the brain. Dyshomeostasis of these metals induces and entertains neurodegenerative diseases. In this review, we described some of the proposed and established mechanisms underlying the actions and properties of the above-mentioned selenoproteins having the characteristic feature of binding transition or heavy metals.
Collapse
Affiliation(s)
| | | | - Carmen Beatrice Dogaru
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.D.); (C.M.); (I.S.)
| | | |
Collapse
|
12
|
Mojarad-Jabali S, Roh KH. Peptide-based inhibitors and nanoparticles: Emerging therapeutics for Alzheimer's disease. Int J Pharm 2025; 669:125055. [PMID: 39653296 DOI: 10.1016/j.ijpharm.2024.125055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD) is an age-related progressive neurodegenerative disorder characterized by memory loss, cognitive decline, and behavioral changes, impacting millions of individuals worldwide. Despite significant research into its cellular and molecular mechanisms, no cure has been found to treat AD to date. For over two decades, research aimed at treating AD has focused on targeting amyloid-β (Aβ); however, these strategies have not demonstrated substantial effectiveness. Consequently, research is now expanding towards targeting other hallmarks of the disease, such as tau protein and brain metal ions. Among potential therapeutics against these pathophysiological targets, peptide-based inhibitors are notable for their high selectivity and low toxicity. Despite these advantages, they face obstacles such as a short half-life in vivo and low efficiencies in crossing the blood-brain barrier (BBB). The use of nanoparticles (NPs) to deliver peptide-based inhibitors to the brain offers unique advantages, such as enhanced stability against degradation, improvement in targeted delivery, and reduced potential for immunogenic responses. This review aims to provide a comprehensive overview of emerging peptides tested as treatments for AD against Aβ, tau protein, and brain metal ions and to evaluate NPs as a means to overcome the limitations. These peptide-based inhibitors are promising, as they not only alleviate symptoms but also aim to prevent progressive neuronal loss, and NPs can be highly effective in delivering these inhibitors.
Collapse
Affiliation(s)
- Solmaz Mojarad-Jabali
- Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, University of Alabama in Huntsville, Huntsville, AL 35899, United States; Biotechnology Science and Engineering Program, University of Alabama in Huntsville, Huntsville, AL 35899, United States.
| |
Collapse
|
13
|
Yi D, Li L, Li M. Subcellular Compartment-Specific Amplified Imaging of Metal Ions via Ribosomal RNA-Regulated DNAzyme Sensors. Angew Chem Int Ed Engl 2025; 64:e202412387. [PMID: 39480115 DOI: 10.1002/anie.202412387] [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: 07/02/2024] [Indexed: 11/02/2024]
Abstract
Although DNAzyme sensors have been widely developed for imaging metal ions, their application in specific subcellular compartments remains challenging due to low spatial controllability. Here we present a locally activatable, DNAzyme-based sensing technology that enables subcellular compartment-specific imaging of metal ions through ribosomal RNA (rRNA) regulated signal amplification. The system leverages a subcellularly encoded rRNA to locally activate DNAzyme-based sensors, and further drives signal amplification via multiple turnover cleavage of molecular beacons, to significantly enhance sensitivity and spatial precision for metal-ion imaging in specific organelles (e.g. mitochondria) or membraneless compartments (e.g. cytosol). Furthermore, we demonstrate that the system allows in situ monitoring of subcellular dynamics of mitochondrial Zn2+ during ischemia and the drug intervention. This study expands the DNAzyme toolbox for investigating the role of subcellular metal-ion dynamics in disease processes.
Collapse
Affiliation(s)
- Deyu Yi
- School of Chemistry and Biological Engineering Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, 30 XueYuan Road, Beijing, 100083, China
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, 11 ZhongGuanCun BeiYiTiao, Beijing, 100190, China
| | - Mengyuan Li
- School of Chemistry and Biological Engineering Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, 30 XueYuan Road, Beijing, 100083, China
| |
Collapse
|
14
|
Berger PK, Bansal R, Sawardekar S, Monk C, Peterson BS. Associations of Maternal Prenatal Zinc Consumption with Infant Brain Tissue Organization and Neurodevelopmental Outcomes. Nutrients 2025; 17:303. [PMID: 39861433 PMCID: PMC11767866 DOI: 10.3390/nu17020303] [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: 12/02/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES While studies in rat pups suggest that early zinc exposure is critical for optimal brain structure and function, associations of prenatal zinc intake with measures of brain development in infants are unknown. This study aimed to assess the associations of maternal zinc intake during pregnancy with MRI measures of brain tissue microstructure and neurodevelopmental outcomes, as well as to determine whether MRI measures of the brain mediated the relationship between maternal zinc intake and neurodevelopmental indices. METHODS Forty-one adolescent mothers were recruited for a longitudinal study during pregnancy. Maternal zinc intake was assessed during the third trimester of pregnancy using a 24 h dietary recall. Infant MRI scans were acquired at 3 weeks postpartum using a 3.0 Tesla scanner to measure fractional anisotropy (FA) and mean diffusivity (MD). Cognitive, language, and motor skills were assessed at 4, 14, and 24 months postpartum using the Bayley Scales of Infant Development. RESULTS Greater prenatal zinc intake was associated with reduced FA in cortical gray matter, particularly in the frontal lobe [medial superior frontal gyrus; β (95% CI) = -1.0 (-1.5, -0.5)], in developing white matter, and in subcortical gray matter nuclei. Greater prenatal zinc intake was associated with reduced MD in cortical gray matter and developing white matter [superior longitudinal fasciculus; -4.4 (-7.1, -1.7)]. Greater maternal zinc intake also was associated with higher cognitive development scores at 14 [0.1 (0.0, 0.1)] and 24 [0.1 (0.0, 0.2)] months of age; MRI indices of FA and MD did not mediate this relationship. CONCLUSIONS Maternal prenatal zinc intake was associated with more favorable measures of brain tissue microstructural maturation and cognitive development during infancy.
Collapse
Affiliation(s)
- Paige K. Berger
- Department of Pediatrics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ravi Bansal
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Child & Adolescent Psychiatry, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - Siddhant Sawardekar
- Division of Child & Adolescent Psychiatry, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - Catherine Monk
- Departments of Obstetrics and Gynecology and Psychiatry, Columbia University Medical Center, New York, NY 10032, USA;
| | - Bradley S. Peterson
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Child & Adolescent Psychiatry, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| |
Collapse
|
15
|
Woyciehowsky M, Larson P, Stephan AR, Dandridge SL, Idonije D, Berg KA, Lanthier A, Acuna SA, Stites SW, Gebhardt WJ, Holtzen SE, Rakshit A, Palmer AE. Systematic characterization of zinc in a series of breast cancer cell lines reveals significant changes in zinc homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632547. [PMID: 39868107 PMCID: PMC11761790 DOI: 10.1101/2025.01.11.632547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
An optimal amount of labile zinc (Zn 2+ ) is essential for proliferation of human cells, where Zn 2+ levels that are too high or too low cause cell cycle exit. Tumors of the breast have been characterized by high levels of total Zn 2+ . Given the role of Zn 2+ in proliferation of human cells and elevation of zinc in breast cancer tumors, we examined the concentration of total and labile Zn 2+ across a panel of 5 breast cancer cell lines, compared to the normal MCF10A cell line. We found that three cell lines (MDA-MB-231, MDA-MB-157, and SK-Br-3) showed elevated labile Zn 2+ in the cytosol, while T-47D showed significantly lower Zn 2+ , and MCF7 showed no change compared to MCF10A cells. There was no change in total Zn 2+ across the cell lines, as measured by ICP-MS, but we did observe a difference in the cells ability to accumulate Zn 2+ when Zn 2+ in the media was elevated. Therefore, we examined how proliferation of each cell line was affected by increases and decreases in the media. We found striking differences, where three cancer cell lines (MDA-MB-231, MDA-MB-157, and MCF7) showed robust proliferation in high Zn 2+ at concentrations that killed MCF10A, T-47D, and SK-Br-3 cells. We also discovered that 4 of the 5 cancer cell lines demonstrate compromised proliferation and increased cell death in low Zn 2+ , suggesting these cells may be addicted to Zn 2+ . Overall, our study suggests significant differences in Zn 2+ homeostasis and regulation in different types of breast cancer cells, with consequences for both proliferation and cell viability.
Collapse
|
16
|
Kokhabi P, Mollazadeh R, Hejazi SF, Nezhad AH, Pazoki-Toroudi H. Importance of Zinc Homeostasis for Normal Cardiac Rhythm. Curr Cardiol Rev 2025; 21:1-18. [PMID: 39301907 DOI: 10.2174/011573403x299868240904120621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/07/2024] [Accepted: 07/23/2024] [Indexed: 09/22/2024] Open
Abstract
Current arrhythmia therapies such as ion channel blockers, catheter ablation, or implantable cardioverter defibrillators have limitations and side effects, and given the proarrhythmic risk associated with conventional, ion channel-targeted anti-arrhythmic drug therapies, a new approach to arrhythmias may be warranted. Measuring and adjusting the level of specific ions that impact heart rhythm can be a simple and low-complication strategy for preventing or treating specific arrhythmias. In addition, new medicines targeting these ions may effectively treat arrhythmias. Numerous studies have shown that intracellular and extracellular zinc concentrations impact the heart's electrical activity. Zinc has been observed to affect cardiac rhythm through a range of mechanisms. These mechanisms encompass the modulation of sodium, calcium, and potassium ion channels, as well as the influence on beta-adrenergic receptors and the enzyme adenylate cyclase. Moreover, zinc can either counteract or induce oxidative stress, hinder calmodulin or the enzyme Ca (2+)/calmodulin-dependent protein kinase II (CaMKII), regulate cellular ATP levels, affect the processes of aging and autophagy, influence calcium ryanodine receptors, and control cellular inflammation. Additionally, zinc has been implicated in the modulation of circadian rhythm. In all the aforementioned cases, the effect of zinc on heart rhythm is largely influenced by its intracellular and extracellular concentrations. Optimal zinc levels are essential for maintaining a normal heart rhythm, while imbalances-whether deficiencies or excesses-can disrupt electrical activity and contribute to arrhythmias.
Collapse
Affiliation(s)
- Pejman Kokhabi
- School of Advanced Medical Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Reza Mollazadeh
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Fatemeh Hejazi
- School of Advanced Medical Sciences, Tonekabon Medical Branch, Islamic Azad University, Tonekabon, Iran
| | - Aida Hossein Nezhad
- School of Advanced Medical Sciences, Tonekabon Medical Branch, Islamic Azad University, Tonekabon, Iran
| | - Hamidreza Pazoki-Toroudi
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Franco CE, Rients EL, Diaz FE, Hansen SL, McGill JL. Dietary Zinc Supplementation in Steers Modulates Labile Zinc Concentration and Zinc Transporter Gene Expression in Circulating Immune Cells. Biol Trace Elem Res 2024; 202:5489-5501. [PMID: 38438601 PMCID: PMC11502596 DOI: 10.1007/s12011-024-04123-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/22/2024] [Indexed: 03/06/2024]
Abstract
Zinc (Zn) is critical for immune function, and marginal Zn deficiency in calves can lead to suboptimal growth and increased disease susceptibility. However, in contrast to other trace minerals such as copper, tissue concentrations of Zn do not change readily in conditions of supplementation or marginal deficiency. Therefore, the evaluation of Zn status remains challenging. Zinc transporters are essential for maintaining intracellular Zn homeostasis, and their expression may indicate changes in Zn status in the animal. Here, we investigated the effects of dietary Zn supplementation on labile Zn concentration and Zn transporter gene expression in circulating immune cells isolated from feedlot steers. Eighteen Angus crossbred steers (261 ± 14 kg) were blocked by body weight and randomly assigned to two dietary treatments: a control diet (58 mg Zn/kg DM, no supplemental Zn) or control plus 150 mg Zn/kg DM (HiZn; 207 mg Zn/kg DM total). After 33 days, Zn supplementation increased labile Zn concentrations (as FluoZin-3 fluorescence) in monocytes, granulocytes, and CD4 T cells (P < 0.05) but had the opposite effect on CD8 and γδ T cells (P < 0.05). Zn transporter gene expression was analyzed on purified immune cell populations collected on days 27 or 28. ZIP11 and ZnT1 gene expression was lower (P < 0.05) in CD4 T cells from HiZn compared to controls. Expression of ZIP6 in CD8 T cells (P = 0.02) and ZnT7 in B cells (P = 0.01) was upregulated in HiZn, while ZnT9 tended (P = 0.06) to increase in B cells from HiZn. These results suggest dietary Zn concentration affects both circulating immune cell Zn concentrations and Zn transporter gene expression in healthy steers.
Collapse
Affiliation(s)
- Carlos E Franco
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, 1907 ISU C-Drive, Ames, IA, USA
| | - Emma L Rients
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Fabian E Diaz
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, 1907 ISU C-Drive, Ames, IA, USA
| | | | - Jodi L McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, 1907 ISU C-Drive, Ames, IA, USA.
| |
Collapse
|
18
|
Dieu Nguyen HQ, Nam MH, Vigh J, Brzezinski J, Duncan L, Park D. Co-delivery of neurotrophic factors and a zinc chelator substantially promotes axon regeneration in the optic nerve crush model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624564. [PMID: 39605527 PMCID: PMC11601601 DOI: 10.1101/2024.11.20.624564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Traumatic optic neuropathies cause the death of retinal ganglion cells (RGCs) and axon degeneration. This is a result of the blockage of neurotrophic factor (NTF) supply from the brain and a vicious cycle of neurotoxicity, possibly mediated by increased levels of retinal Zn 2+ . Ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) are two NTFs that are known to support RGC survival and promote axon regeneration. Dipicolylamine (DPA) has a strong affinity to Zn 2+ and can selectively chelate this ion. To continuously supply NTFs and reduce elevated retinal Zn 2+ , we developed poly(serinol hexamethylene urea)-based sulfonated nanoparticles (S-PSHU NPs), that co-delivers CNTF, BDNF, and DPA. An in vitro release study was performed using the NTF-DPA-loaded S-PSHU NPs, demonstrating a sustained release of CNTF and BDNF for up to 8 weeks, while DPA was released for 4 weeks. In a rat optic nerve crush (ONC) model, DPA-loaded S-PSHU NPs exhibited dose-dependent elimination of retinal Zn 2+ . Similarly, in vitro primary RGC culture demonstrated that the activity of RGCs and axon growth were dependent on the dosage of CNTF and BDNF. In addition, the NTF-DPA-loaded S-PSHU NPs significantly improved RGC survival and axon regeneration following ONC in rats, with the regenerated axons extending to the distal segment of the brain, including the suprachiasmatic nucleus, lateral geniculate nucleus, and superior colliculus.
Collapse
|
19
|
Gao L, Meng Y, Luo X, Chen J, Wang X. ZnO Nanoparticles-Induced MRI Alterations to the Rat Olfactory Epithelium and Olfactory Bulb after Intranasal Instillation. TOXICS 2024; 12:724. [PMID: 39453144 PMCID: PMC11511357 DOI: 10.3390/toxics12100724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/28/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024]
Abstract
Since zinc oxide (ZnO) nanoparticles (NPs) have been widely applied, the nano community and the general public have paid great attention to the toxicity of ZnO NPs. We detected 20-nm ZnO NPs biotoxicity following nasal exposure utilizing the non-invasive and real-time magnetic resonance imaging (MRI) technique. MR images were scanned in the rat olfactory epithelium (OE) and olfactory bulb (OB) on a 4.7 T scanner following the treatment (as early as 1 day and up to 21 days after), and the histological changes were evaluated. The influence of the size of the ZnO NPs and chemical components was also investigated. Our study revealed that 20-nm ZnO NPs induced obvious structural disruption and inflammation in the OE and OB at the acute stage. The results suggest that the real-time and non-invasive advantages of MRI allow it to observe and assess, directly and dynamically, the potential toxicity of long-term exposure to ZnO NPs in the olfactory system. These findings indicate the size-dependent toxicity of ZnO NPs with respect to the olfactory bulb. Further study is needed to reveal the mechanism behind ZnO NPs' toxicity.
Collapse
Affiliation(s)
- Lifeng Gao
- Department of Medical Imaging, School of Medicine, Jianghan University, Wuhan 430056, China; (L.G.); (X.L.); (J.C.)
| | - Yuguang Meng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China;
| | - Xiaowen Luo
- Department of Medical Imaging, School of Medicine, Jianghan University, Wuhan 430056, China; (L.G.); (X.L.); (J.C.)
| | - Jiangyuan Chen
- Department of Medical Imaging, School of Medicine, Jianghan University, Wuhan 430056, China; (L.G.); (X.L.); (J.C.)
| | - Xuxia Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China;
| |
Collapse
|
20
|
Nazemof N, Breznan D, Dirieh Y, Blais E, Johnston LJ, Tayabali AF, Gomes J, Kumarathasan P. Cytotoxic Potencies of Zinc Oxide Nanoforms in A549 and J774 Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1601. [PMID: 39404328 PMCID: PMC11482475 DOI: 10.3390/nano14191601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Zinc oxide nanoparticles (NPs) are used in a wide range of consumer products and in biomedical applications, resulting in an increased production of these materials with potential for exposure, thus causing human health concerns. Although there are many reports on the size-related toxicity of ZnO NPs, the toxicity of different nanoforms of this chemical, toxicity mechanisms, and potency determinants need clarification to support health risk characterization. A set of well-characterized ZnO nanoforms (e.g., uncoated ca. 30, 45, and 53 nm; coated with silicon oil, stearic acid, and (3-aminopropyl) triethoxysilane) were screened for in vitro cytotoxicity in two cell types, human lung epithelial cells (A549), and mouse monocyte/macrophage (J774) cells. ZnO (bulk) and ZnCl2 served as reference particles. Cytotoxicity was examined 24 h post-exposure by measuring CTB (viability), ATP (energy metabolism), and %LDH released (membrane integrity). Cellular oxidative stress (GSH-GSSG) and secreted proteins (targeted multiplex assay) were analyzed. Zinc oxide nanoform type-, dose-, and cell type-specific cytotoxic responses were seen, along with cellular oxidative stress. Cell-secreted protein profiles suggested ZnO NP exposure-related perturbations in signaling pathways relevant to inflammation/cell injury and corresponding biological processes, namely reactive oxygen species generation and apoptosis/necrosis, for some nanoforms, consistent with cellular oxidative stress and ATP status. The size, surface area, agglomeration state and metal contents of these ZnO nanoforms appeared to be physicochemical determinants of particle potencies. These findings warrant further research on high-content "OMICs" to validate and resolve toxicity pathways related to exposure to nanoforms to advance health risk-assessment efforts and to inform on safer materials.
Collapse
Affiliation(s)
- Nazila Nazemof
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada; (N.N.); (J.G.)
| | - Dalibor Breznan
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - Yasmine Dirieh
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - Erica Blais
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - Linda J. Johnston
- Metrology Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada;
| | - Azam F. Tayabali
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| | - James Gomes
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada; (N.N.); (J.G.)
| | - Premkumari Kumarathasan
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 7K4, Canada; (N.N.); (J.G.)
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada; (D.B.); (Y.D.); (E.B.); (A.F.T.)
| |
Collapse
|
21
|
Bizup B, Tzounopoulos T. On the genesis and unique functions of zinc neuromodulation. J Neurophysiol 2024; 132:1241-1254. [PMID: 39196675 PMCID: PMC11495185 DOI: 10.1152/jn.00285.2024] [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: 07/05/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/30/2024] Open
Abstract
In addition to the essential structural and catalytic functions of zinc, evolution has adopted synaptic zinc as a neuromodulator. In the brain, synaptic zinc is released primarily from glutamatergic neurons, notably in the neocortex, hippocampus, amygdala, and auditory brainstem. In these brain areas, synaptic zinc is essential for neuronal and sensory processing fine-tuning. But what niche does zinc fill in neural signaling that other neuromodulators do not? Here, we discuss the evolutionary history of zinc as a signaling agent and its eventual adoption as an essential neuromodulator in the mammalian brain. We then attempt to describe the unique roles that zinc has carved out of the vast and diverse landscape of neuromodulators.
Collapse
Affiliation(s)
- Brandon Bizup
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
22
|
Takeda A, Tamano H. Insight into brain metallothioneins from bidirectional Zn2+ signaling in synaptic dynamics. Metallomics 2024; 16:mfae039. [PMID: 39223100 DOI: 10.1093/mtomcs/mfae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/01/2024] [Indexed: 09/04/2024]
Abstract
The basal levels as the labile Zn2+ pools in the extracellular and intracellular compartments are in the range of ∼10 nM and ∼100 pM, respectively. The influx of extracellular Zn2+ is used for memory via cognitive activity and is regulated for synaptic plasticity, a cellular mechanism of memory. When Zn2+ influx into neurons excessively occurs, however, it becomes a critical trigger for cognitive decline and neurodegeneration, resulting in acute and chronic pathogenesis. Aging, a biological process, generally accelerates vulnerability to neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). The basal level of extracellular Zn2+ is age relatedly increased in the rat hippocampus, and the influx of extracellular Zn2+ contributes to accelerating vulnerability to the AD and PD pathogenesis in experimental animals with aging. Metallothioneins (MTs) are Zn2+-binding proteins for cellular Zn2+ homeostasis and involved in not only supplying functional Zn2+ required for cognitive activity, but also capturing excess (toxic) Zn2+ involved in cognitive decline and neurodegeneration. Therefore, it is estimated that regulation of MT synthesis is involved in both neuronal activity and neuroprotection. The present report provides recent knowledge regarding the protective/preventive potential of MT synthesis against not only normal aging but also the AD and PD pathogenesis in experimental animals, focused on MT function in bidirectional Zn2+ signaling in synaptic dynamics.
Collapse
Affiliation(s)
- Atsushi Takeda
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Haruna Tamano
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
- Shizuoka Tohto Medical College, 1949 Minamiema, Izunokuni, Shizuoka 410-2221, Japan
| |
Collapse
|
23
|
Yu F, Li X, Sheng C, Li L. DNA Nanotechnology Targeting Mitochondria: From Subcellular Molecular Imaging to Tailor-Made Therapeutics. Angew Chem Int Ed Engl 2024; 63:e202409351. [PMID: 38872505 DOI: 10.1002/anie.202409351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/15/2024]
Abstract
Mitochondria, one of the most important organelles, represent a crucial subcellular target for fundamental research and biomedical applications. Despite significant advances in the design of DNA nanotechnologies for a variety of bio-applications, the dearth of strategies that enable mitochondria targeting for subcellular molecular imaging and therapy remains an outstanding challenge in this field. In this Minireview, we summarize the recent progresses on the emerging design and application of DNA nanotechnology for mitochondria-targeted molecular imaging and tumor treatment. We first highlight the engineering of mitochondria-localized DNA nanosensors for in situ detection and imaging of diverse key molecules that are essential to maintain mitochondrial functions, including mitochondrial DNA and microRNA, enzymes, small molecules, and metal ions. Then, we compile the developments of DNA nanotechnologies for mitochondria-targeted anti-tumor therapy, including modularly designed DNA nanodevices for subcellular delivery of therapeutic agents, and programmed DNA assembly for mitochondrial interference. We will place an emphasis on clarification of the chemical principles of how DNA nanobiotechnology can be designed to target mitochondria for various biomedical applications. Finally, the remaining challenges and future directions in this emerging field will be discussed, hoping to inspire further development of advanced DNA toolkits for both academic and clinical research regarding mitochondria.
Collapse
Affiliation(s)
- Fangzhi Yu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiangfei Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chuangui Sheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
24
|
Asthana S, Maddeshiya T, Tamrakar A, Kumar P, Garg N, Pandey MD. L-Tryptophan-based pyrene conjugate for intracellular zinc-guided excimer emission and controlled nano-assembly. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:5633-5641. [PMID: 39139130 DOI: 10.1039/d4ay00979g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
This article describes intracellular zinc-induced excimer emission and tuning of self-assembly from L-tryptophan-pyrene conjugate (1). The zinc-guided excimer formation is due to the interaction of the pyrene moiety in an excited state. AFM studies show the structural modification in the supramolecular nano-assembly of 1 from dome-shaped to porous surface after complexation with zinc ions. Further, the interaction of 1 with Zn(II) ion is also studied using DFT, Job's plot, NMR titration and HRMS. The results of Zn(II) ion determination in natural water samples and RAW 264.7 cells demonstrate the practical utility of 1.
Collapse
Affiliation(s)
- Surabhi Asthana
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| | - Tarkeshwar Maddeshiya
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| | - Arpna Tamrakar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| | - Praveen Kumar
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Neha Garg
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Mrituanjay D Pandey
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| |
Collapse
|
25
|
Lee LCC, Lo KKW. Shining New Light on Biological Systems: Luminescent Transition Metal Complexes for Bioimaging and Biosensing Applications. Chem Rev 2024; 124:8825-9014. [PMID: 39052606 PMCID: PMC11328004 DOI: 10.1021/acs.chemrev.3c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Luminescence imaging is a powerful and versatile technique for investigating cell physiology and pathology in living systems, making significant contributions to life science research and clinical diagnosis. In recent years, luminescent transition metal complexes have gained significant attention for diagnostic and therapeutic applications due to their unique photophysical and photochemical properties. In this Review, we provide a comprehensive overview of the recent development of luminescent transition metal complexes for bioimaging and biosensing applications, with a focus on transition metal centers with a d6, d8, and d10 electronic configuration. We elucidate the structure-property relationships of luminescent transition metal complexes, exploring how their structural characteristics can be manipulated to control their biological behavior such as cellular uptake, localization, biocompatibility, pharmacokinetics, and biodistribution. Furthermore, we introduce the various design strategies that leverage the interesting photophysical properties of luminescent transition metal complexes for a wide variety of biological applications, including autofluorescence-free imaging, multimodal imaging, organelle imaging, biological sensing, microenvironment monitoring, bioorthogonal labeling, bacterial imaging, and cell viability assessment. Finally, we provide insights into the challenges and perspectives of luminescent transition metal complexes for bioimaging and biosensing applications, as well as their use in disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
26
|
Mo S, Kim MK, Jang JS, Lee SH, Hong SJ, Jung S, Kim HH. Unique expression and critical role of metallothionein 3 in the control of osteoclastogenesis and osteoporosis. Exp Mol Med 2024; 56:1791-1806. [PMID: 39085359 PMCID: PMC11372110 DOI: 10.1038/s12276-024-01290-3] [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: 11/17/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 08/02/2024] Open
Abstract
Bone homeostasis is maintained by an intricate balance between osteoclasts and osteoblasts, which becomes disturbed in osteoporosis. Metallothioneins (MTs) are major contributors in cellular zinc regulation. However, the role of MTs in bone cell regulation has remained unexplored. Single-cell RNA sequencing analysis discovered that, unlike the expression of other MT members, the expression of MT3 was unique to osteoclasts among various macrophage populations and was highly upregulated during osteoclast differentiation. This unique MT3 upregulation was validated experimentally and supported by ATAC sequencing data analyses. Downregulation of MT3 by gene knockdown or knockout resulted in excessive osteoclastogenesis and exacerbated bone loss in ovariectomy-induced osteoporosis. Transcriptome sequencing of MT3 knockdown osteoclasts and gene set enrichment analysis indicated that the oxidative stress and redox pathways were enriched, which was verified by MT3-dependent regulation of reactive oxygen species (ROS). In addition, MT3 deficiency increased the transcriptional activity of SP1 in a manner dependent on intracellular zinc levels. This MT3-zinc-SP1 axis was crucial for the control of osteoclasts, as zinc chelation and SP1 knockdown abrogated the promotion of SP1 activity and osteoclastogenesis by MT3 deletion. Moreover, SP1 bound to the NFATc1 promoter, and overexpression of an inactive SP1 mutant negated the effects of MT3 deletion on NFATc1 and osteoclastogenesis. In conclusion, MT3 plays a pivotal role in controlling osteoclastogenesis and bone metabolism via dual axes involving ROS and SP1. The present study demonstrated that MT3 elevation is a potential therapeutic strategy for osteolytic bone disorders, and it established for the first time that MT3 is a crucial bone mass regulator.
Collapse
Affiliation(s)
- Shenzheng Mo
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Min Kyung Kim
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Bone Science R&D Center, Tissue Regeneration Institute, Osstem Implant, Seoul, 07789, Republic of Korea
| | - Ji Sun Jang
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seung Hye Lee
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seo Jin Hong
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Suhan Jung
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea.
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
27
|
González Díaz A, Cataldi R, Mannini B, Vendruscolo M. Preparation and Characterization of Zn(II)-Stabilized Aβ 42 Oligomers. ACS Chem Neurosci 2024; 15:2586-2599. [PMID: 38979921 PMCID: PMC11258685 DOI: 10.1021/acschemneuro.4c00084] [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: 02/06/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
Aβ oligomers are being investigated as cytotoxic agents in Alzheimer's disease (AD). Because of their transient nature and conformational heterogeneity, the relationship between the structure and activity of these oligomers is still poorly understood. Hence, methods for stabilizing Aβ oligomeric species relevant to AD are needed to uncover the structural determinants of their cytotoxicity. Here, we build on the observation that metal ions and metabolites have been shown to interact with Aβ, influencing its aggregation and stabilizing its oligomeric species. We thus developed a method that uses zinc ions, Zn(II), to stabilize oligomers produced by the 42-residue form of Aβ (Aβ42), which is dysregulated in AD. These Aβ42-Zn(II) oligomers are small in size, spanning the 10-30 nm range, stable at physiological temperature, and with a broad toxic profile in human neuroblastoma cells. These oligomers offer a tool to study the mechanisms of toxicity of Aβ oligomers in cellular and animal AD models.
Collapse
Affiliation(s)
- Alicia González Díaz
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Rodrigo Cataldi
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Benedetta Mannini
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
- Department
of Experimental and Clinical Biomedical Sciences Mario Serio, University
of Florence, 50134 Florence, Italy
| | - Michele Vendruscolo
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
28
|
Yu L, Sun F, Wang Y, Li W, Zheng Y, Shen G, Wang Y, Chen M. Effects of MgO nanoparticle addition on the mechanical properties, degradation properties, antibacterial properties and in vitro and in vivo biological properties of 3D-printed Zn scaffolds. Bioact Mater 2024; 37:72-85. [PMID: 38523703 PMCID: PMC10958222 DOI: 10.1016/j.bioactmat.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/29/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024] Open
Abstract
Bone tissue engineering is the main method for repairing large segment bone defects. In this study, a layer of bioactive MgO nanoparticles was wrapped on the surface of spherical Zn powders, which allowed the MgO nanoparticles to be incorporated into 3D-printed Zn matrix and improved the biodegradation and biocompatibility of the Zn matrix. The results showed that porous pure Zn scaffolds and Zn/MgO scaffolds with skeletal-gyroid (G) model structure were successfully prepared by selective laser melting (SLM). The average porosity of two porous scaffolds was 59.3 and 60.0%, respectively. The pores were uniformly distributed with an average pore size of 558.6-569.3 μm. MgO nanoparticles regulated the corrosion rate of scaffolds, resulting in a more uniform corrosion degradation behavior of the Zn/MgO scaffolds in simulated body fluid solution. The degradation ratio of Zn/MgO composite scaffolds in vivo was increased compared to pure Zn scaffolds, reaching 15.6% at 12 weeks. The yield strength (10.8 ± 2.4 MPa) of the Zn/MgO composite scaffold was comparable to that of cancellous bone, and the antimicrobial rate were higher than 99%. The Zn/MgO composite scaffolds could better guide bone tissue regeneration in rat cranial bone repair experiments (completely filling the scaffolds at 12 weeks). Therefore, porous Zn/MgO scaffolds with G-model structure prepared with SLM are a promising biodegradable bone tissue engineering scaffold.
Collapse
Affiliation(s)
- Leiting Yu
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Fengdong Sun
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Yuanyuan Wang
- School of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Wei Li
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Guangxin Shen
- Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Yao Wang
- School of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Minfang Chen
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin, 300384, China
- National Demonstration Center for Experimental Function Materials Education, Tianjin University of Technology, Tianjin, 300384, China
| |
Collapse
|
29
|
Schoofs H, Schmit J, Rink L. Zinc Toxicity: Understanding the Limits. Molecules 2024; 29:3130. [PMID: 38999082 PMCID: PMC11243279 DOI: 10.3390/molecules29133130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Zinc, a vital trace element, holds significant importance in numerous physiological processes within the body. It participates in over 300 enzymatic reactions, metabolic functions, regulation of gene expression, apoptosis and immune modulation, thereby demonstrating its essential role in maintaining overall health and well-being. While zinc deficiency is associated with significant health risks, an excess of this trace element can also lead to harmful effects. According to the World Health Organization (WHO), 6.7 to 15 mg per day are referred to be the dietary reference value. An excess of the recommended daily intake may result in symptoms such as anemia, neutropenia and zinc-induced copper deficiency. The European Food Safety Authority (EFSA) defines the tolerable upper intake level (UL) as 25 mg per day, whereas the Food and Drug Administration (FDA) allows 40 mg per day. This review will summarize the current knowledge regarding the calculation of UL and other health risks associated with zinc. For example, zinc intake is not limited to oral consumption; other routes, such as inhalation or topical application, may also pose risks of zinc intoxication.
Collapse
Affiliation(s)
- Hannah Schoofs
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52074 Aachen, Germany
| | - Joyce Schmit
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52074 Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
30
|
Csomos A, Madarász M, Turczel G, Cseri L, Bodor A, Matuscsák A, Katona G, Kovács E, Rózsa B, Mucsi Z. A GFP Inspired 8-Methoxyquinoline-Derived Fluorescent Molecular Sensor for the Detection of Zn 2+ by Two-Photon Microscopy. Chemistry 2024; 30:e202400009. [PMID: 38446718 DOI: 10.1002/chem.202400009] [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: 01/02/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/08/2024]
Abstract
An effective, GFP-inspired fluorescent Zn2+ sensor is developed for two-photon microscopy and related biological application that features an 8-methoxyquinoline moiety. Excellent photophysical characteristics including a 37-fold fluorescence enhancement with excitation and emission maxima at 440 nm and 505 nm, respectively, as well as a high two-photon cross-section of 73 GM at 880 nm are reported. Based on the experimental data, the relationship between the structure and properties was elucidated and explained backed up by DFT calculations, particularly the observed PeT phenomenon for the turn-on process. Biological validation and detailed experimental and theoretical characterization of the free and the zinc-bound compounds are presented.
Collapse
Affiliation(s)
- Attila Csomos
- Femtonics Ltd., Tűzoltó utca 59, H-1094, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
| | - Miklós Madarász
- BrainVisionCenter, Liliom utca 43-45, H-1094, Budapest, Hungary
| | - Gábor Turczel
- NMR Research Laboratory, HUN-REN Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117, Budapest, Hungary
| | - Levente Cseri
- BrainVisionCenter, Liliom utca 43-45, H-1094, Budapest, Hungary
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Műegyetem rakpart 3, H-1111, Budapest, Hungary
| | - Andrea Bodor
- Analytical and BioNMR Laboratory, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
| | - Anett Matuscsák
- Laboratory of 3D functional network and dendritic imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, H-1083, Budapest, Hungary
| | - Gergely Katona
- Two-Photon Measurement Technology Research Group, Pázmány Péter Catholic University Práter, utca 50/a, H-1083, Budapest, Hungary
| | - Ervin Kovács
- Two-Photon Measurement Technology Research Group, Pázmány Péter Catholic University Práter, utca 50/a, H-1083, Budapest, Hungary
- Polymer Chemistry and Physics Research Group, HUN-REN Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117, Budapest, Hungary
| | - Balázs Rózsa
- BrainVisionCenter, Liliom utca 43-45, H-1094, Budapest, Hungary
- Laboratory of 3D functional network and dendritic imaging, HUN-REN Institute of Experimental Medicine, Szigony utca 43, H-1083, Budapest, Hungary
- Two-Photon Measurement Technology Research Group, Pázmány Péter Catholic University Práter, utca 50/a, H-1083, Budapest, Hungary
| | - Zoltán Mucsi
- Femtonics Ltd., Tűzoltó utca 59, H-1094, Budapest, Hungary
- BrainVisionCenter, Liliom utca 43-45, H-1094, Budapest, Hungary
- Faculty of Materials and Chemical Sciences, University of Miskolc, H-3515, Miskolc, Hungary
| |
Collapse
|
31
|
Cheng J, Kolba N, Tako E. The effect of dietary zinc and zinc physiological status on the composition of the gut microbiome in vivo. Crit Rev Food Sci Nutr 2024; 64:6432-6451. [PMID: 36688291 DOI: 10.1080/10408398.2023.2169857] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Zinc serves critical catalytic, regulatory, and structural roles. Hosts and their resident gut microbiota both require zinc, leading to competition, where a balance must be maintained. This systematic review examined evidence on dietary zinc and physiological status (zinc deficiency or high zinc/zinc overload) effects on gut microbiota. This review was conducted according to PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analyses) guidelines and registered in PROSPERO (CRD42021250566). PubMed, Web of Science, and Scopus databases were searched for in vivo (animal) studies, resulting in eight selected studies. Study quality limitations were evaluated using the SYRCLE risk of bias tool and according to ARRIVE guidelines. The results demonstrated that zinc deficiency led to inconsistent changes in α-diversity and short-chain fatty acid production but led to alterations in bacterial taxa with functions in carbohydrate metabolism, glycan metabolism, and intestinal mucin degradation. High dietary zinc/zinc overload generally resulted in either unchanged or decreased α-diversity, decreased short-chain fatty acid production, and increased bacterial metal resistance and antibiotic resistance genes. Additional studies in human and animal models are needed to further understand zinc physiological status effects on the intestinal microbiome and clarify the applicability of utilizing the gut microbiome as a potential zinc status biomarker.
Collapse
Affiliation(s)
- Jacquelyn Cheng
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Nikolai Kolba
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Elad Tako
- Department of Food Science, Cornell University, Ithaca, New York, USA
| |
Collapse
|
32
|
Bhole RP, Chikhale RV, Rathi KM. Current biomarkers and treatment strategies in Alzheimer disease: An overview and future perspectives. IBRO Neurosci Rep 2024; 16:8-42. [PMID: 38169888 PMCID: PMC10758887 DOI: 10.1016/j.ibneur.2023.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024] Open
Abstract
Alzheimer's disease (AD), a progressive degenerative disorder first identified by Alois Alzheimer in 1907, poses a significant public health challenge. Despite its prevalence and impact, there is currently no definitive ante mortem diagnosis for AD pathogenesis. By 2050, the United States may face a staggering 13.8 million AD patients. This review provides a concise summary of current AD biomarkers, available treatments, and potential future therapeutic approaches. The review begins by outlining existing drug targets and mechanisms in AD, along with a discussion of current treatment options. We explore various approaches targeting Amyloid β (Aβ), Tau Protein aggregation, Tau Kinases, Glycogen Synthase kinase-3β, CDK-5 inhibitors, Heat Shock Proteins (HSP), oxidative stress, inflammation, metals, Apolipoprotein E (ApoE) modulators, and Notch signaling. Additionally, we examine the historical use of Estradiol (E2) as an AD therapy, as well as the outcomes of Randomized Controlled Trials (RCTs) that evaluated antioxidants (e.g., vitamin E) and omega-3 polyunsaturated fatty acids as alternative treatment options. Notably, positive effects of docosahexaenoic acid nutriment in older adults with cognitive impairment or AD are highlighted. Furthermore, this review offers insights into ongoing clinical trials and potential therapies, shedding light on the dynamic research landscape in AD treatment.
Collapse
Affiliation(s)
- Ritesh P. Bhole
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
- Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | | | - Karishma M. Rathi
- Department of Pharmacy Practice, Dr. D. Y. Patil institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
| |
Collapse
|
33
|
Gómez-Castro CZ, Quintanar L, Vela A. An N-terminal acidic β-sheet domain is responsible for the metal-accumulation properties of amyloid-β protofibrils: a molecular dynamics study. J Biol Inorg Chem 2024; 29:407-425. [PMID: 38811408 PMCID: PMC11186886 DOI: 10.1007/s00775-024-02061-1] [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: 12/20/2023] [Accepted: 04/10/2024] [Indexed: 05/31/2024]
Abstract
The influence of metal ions on the structure of amyloid- β (Aβ) protofibril models was studied through molecular dynamics to explore the molecular mechanisms underlying metal-induced Aβ aggregation relevant in Alzheimer's disease (AD). The models included 36-, 48-, and 188-mers of the Aβ42 sequence and two disease-modifying variants. Primary structural effects were observed at the N-terminal domain, as it became susceptible to the presence of cations. Specially when β-sheets predominate, this motif orients N-terminal acidic residues toward one single face of the β-sheet, resulting in the formation of an acidic region that attracts cations from the media and promotes the folding of the N-terminal region, with implications in amyloid aggregation. The molecular phenotype of the protofibril models based on Aβ variants shows that the AD-causative D7N mutation promotes the formation of N-terminal β-sheets and accumulates more Zn2+, in contrast to the non-amyloidogenic rodent sequence that hinders the β-sheets and is more selective for Na+ over Zn2+ cations. It is proposed that forming an acidic β-sheet domain and accumulating cations is a plausible molecular mechanism connecting the elevated affinity and concentration of metals in Aβ fibrils to their high content of β-sheet structure at the N-terminal sequence.
Collapse
Affiliation(s)
- Carlos Z Gómez-Castro
- Conahcyt-Universidad Autónoma del Estado de Hidalgo, Km 4.5 Carr. Pachuca-Tulancingo, Mineral de La Reforma, 42184, Hidalgo, Mexico.
| | - Liliana Quintanar
- Department of Chemistry, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX, San Pedro Zacatenco, 07360, Gustavo A. Madero, Mexico.
| | - Alberto Vela
- Department of Chemistry, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX, San Pedro Zacatenco, 07360, Gustavo A. Madero, Mexico.
| |
Collapse
|
34
|
Chen Y. Advances in Organic Fluorescent Probes for Intracellular Zn 2+ Detection and Bioimaging. Molecules 2024; 29:2542. [PMID: 38893419 PMCID: PMC11173588 DOI: 10.3390/molecules29112542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/13/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Zinc ions (Zn2+) play a key role in maintaining and regulating protein structures and functions. To better understand the intracellular Zn2+ homeostasis and signaling role, various fluorescent sensors have been developed that allow the monitoring of Zn2+ concentrations and bioimaging in live cells in real time. This review highlights the recent development of organic fluorescent probes for the detection and imaging of intracellular Zn2+, including the design and construction of the probes, fluorescent response mechanisms, and their applications to intracellular Zn2+ detection and imaging on-site. Finally, the current challenges and prospects are discussed.
Collapse
Affiliation(s)
- Yi Chen
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
35
|
Elitt CM, Ross MM, Wang J, Fahrni CJ, Rosenberg PA. Developmental regulation of zinc homeostasis in differentiating oligodendrocytes. Neurosci Lett 2024; 831:137727. [PMID: 38467270 DOI: 10.1016/j.neulet.2024.137727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Oligodendrocytes develop through sequential stages and understanding pathways regulating their differentiation remains an important area of investigation. Zinc is required for the function of enzymes, proteins and transcription factors, including those important in myelination and mitosis. Our previous studies using the ratiometric zinc sensor chromis-1 demonstrated a reduction in intracellular free zinc concentrations in mature MBP+ oligodendrocytes compared with earlier stages (Bourassa et al., 2018). We performed a more detailed developmental study to better understand the temporal course of zinc homeostasis across the oligodendrocyte lineage. Using chromis-1, we found a transient increase in free zinc after O4+,O1- pre-oligodendrocytes were switched from proliferation medium into terminal differentiation medium. To gather other evidence for dynamic regulation of free zinc during oligodendrocyte development, qPCR was used to evaluate mRNA expression of major zinc storage proteins metallothioneins (MTs) and metal regulatory transcription factor 1 (MTF1), which controls expression of MTs. MT1, MT2 and MTF1 mRNAs were increased several fold in mature oligodendrocytes compared to oligodendrocytes in proliferation medium. To assess the depth of the zinc buffer, we assayed zinc release from intracellular stores using the oxidizing thiol reagent 2,2'-dithiodipyridine (DTDP). Exposure to DTDP resulted in ∼ 100% increase in free zinc in pre-oligodendrocytes but, paradoxically more modest ∼ 60% increase in mature oligodendrocytes despite increased expression of MTs. These results suggest that zinc homeostasis is regulated during oligodendrocyte development, that oligodendrocytes are a useful model for studying zinc homeostasis in the central nervous system, and that regulation of zinc homeostasis may be important in oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Christopher M Elitt
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States.
| | - Madeline M Ross
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| | - Jianlin Wang
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| | - Christoph J Fahrni
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, United States; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Paul A Rosenberg
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| |
Collapse
|
36
|
Chen M, Qin Y, Peng Y, Mai R, Teng H, Qi Z, Mo J. Advancing stroke therapy: the potential of MOF-based nanozymes in biomedical applications. Front Bioeng Biotechnol 2024; 12:1363227. [PMID: 38798955 PMCID: PMC11119330 DOI: 10.3389/fbioe.2024.1363227] [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: 12/30/2023] [Accepted: 04/12/2024] [Indexed: 05/29/2024] Open
Abstract
In this study, we explored the growing use of metal-organic framework (MOF)-based Nanozymes in biomedical research, with a specific emphasis on their applications in stroke therapy. We have discussed the complex nature of stroke pathophysiology, highlighting the crucial role of reactive oxygen species (ROS), and acknowledging the limitations of natural enzymes in addressing these challenges. We have also discussed the role of nanozymes, particularly those based on MOFs, their structural similarities to natural enzymes, and their potential to improve reactivity in various biomedical applications. The categorization of MOF nanozymes based on enzyme-mimicking activities is discussed, and their applications in stroke therapy are explored. We have reported the potential of MOF in treating stroke by regulating ROS levels, alleviation inflammation, and reducing neuron apoptosis. Additionally, we have addressed the challenges in developing efficient antioxidant nanozyme systems for stroke treatment. The review concludes with the promise of addressing these challenges and highlights the promising future of MOF nanozymes in diverse medical applications, particularly in the field of stroke treatment.
Collapse
Affiliation(s)
- Meirong Chen
- The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Medical College of Guangxi University, Nanning, China
| | - Yang Qin
- Department of Graduate and Postgraduate Education Management, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yongmei Peng
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Ruyu Mai
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Huanyao Teng
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Zhongquan Qi
- Medical College of Guangxi University, Nanning, China
| | - Jingxin Mo
- The Guangxi Clinical Research Center for Neurological Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Lab of Neurology, The Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
37
|
Reda AT, Park JY, Park YT. Zinc Oxide-Based Nanomaterials for Microbiostatic Activities: A Review. J Funct Biomater 2024; 15:103. [PMID: 38667560 PMCID: PMC11050959 DOI: 10.3390/jfb15040103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The world is fighting infectious diseases. Therefore, effective antimicrobials are required to prevent the spread of microbes and protect human health. Zinc oxide (ZnO) nano-materials are known for their antimicrobial activities. Because of their distinctive physical and chemical characteristics, they can be used in medical and environmental applications. ZnO-based composites are among the leading sources of antimicrobial research. They are effective at killing (microbicidal) and inhibiting the growth (microbiostatic) of numerous microorganisms, such as bacteria, viruses, and fungi. Although most studies have focused on the microbicidal features, there is a lack of reviews on their microbiostatic effects. This review provides a detailed overview of available reports on the microbiostatic activities of ZnO-based nano-materials against different microorganisms. Additionally, the factors that affect the efficacy of these materials, their time course, and a comparison of the available antimicrobials are highlighted in this review. The basic properties of ZnO, challenges of working with microorganisms, and working mechanisms of microbiostatic activities are also examined. This review underscores the importance of further research to better understand ZnO-based nano-materials for controlling microbial growth.
Collapse
Affiliation(s)
| | | | - Yong Tae Park
- Department of Mechanical Engineering, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin, Gyeonggi 17058, Republic of Korea; (A.T.R.)
| |
Collapse
|
38
|
Shenderovich IG. The Scope of the Applicability of Non-relativistic DFT Calculations of NMR Chemical Shifts in Pyridine-Metal Complexes for Applied Applications. Chemphyschem 2024; 25:e202300986. [PMID: 38259119 DOI: 10.1002/cphc.202300986] [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: 12/21/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 01/24/2024]
Abstract
Heavy metals are toxic, but it is impossible to stop using them. Considering the variety of molecular systems in which they can be present, the multicomponent nature and disorder of the structure of such systems, one of the most effective methods for studying them is NMR spectroscopy. This determines the need to calculate NMR chemical shifts for expected model systems. For elements beyond the third row of the periodic table, corrections for relativistic effects are necessary when calculating NMR parameters. Such corrections may be necessary even for light atoms due to the shielding effect of a neighboring heavy atom. This work examines the extent to which non-relativistic DFT calculations are able to reproduce experimental 15N and 113Cd NMR chemical shift tensors in pyridine-metal coordination complexes. It is shown that while for the calculation of 15N NMR chemical shift tensors there is no real need to consider relativistic corrections, for 113Cd, on the contrary, none of the tested calculation methods could reproduce the experimentally obtained tensor to any extent correctly.
Collapse
Affiliation(s)
- Ilya G Shenderovich
- NMR Department, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040, Regensburg, Germany
| |
Collapse
|
39
|
Zhang Q, Lou C, Li H, Li Y, Zhang H, Li Z, Qi G, Cai X, Luo Q, Fan L, Li X, Lao W, Zhu W, Li X. Zinc hybrid polyester barrier membrane accelerates guided tissue regeneration. J Control Release 2024; 368:676-690. [PMID: 38458572 DOI: 10.1016/j.jconrel.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Barrier membranes play a pivotal role in the success of guided periodontal tissue regeneration. The biodegradable barriers predominantly used in clinical practice often lack sufficient barrier strength, antibacterial properties, and bioactivity, frequently leading to suboptimal regeneration outcomes. Although with advantages in mechanical strength, biodegradability and plasticity, bioinert aliphatic polyesters as barrier materials are usually polymerized via toxic catalysts, hard to be functionalized and lack of antibacterial properties. To address these challenges, we propose a new concept that controlled release of bioactive substance on the whole degradation course can give a bioinert aliphatic polyester bioactivity. Thus, a Zn-based catalytic system for polycondensation of dicarboxylic acids and diols is created to prepare zinc covalent hybrid polyester (PBS/ZnO). The atomically-dispersed Zn2+ ions entering main chain of polyester molecules endow PBS/ZnO barrier with antibacterial properties, barrier strength, excellent biocompatibility and histocompatibility. Further studies reveal that relying on long-term controlled release of Zn2+ ions, the PBS/ZnO membrane greatly expedites osteogenetic effect in guided tissue regeneration (GTR) by enhancing the mitochondrial function of macrophages to induce M2 polarization. These findings show a novel preparation strategy of bioactive polyester biomaterials based on long term controlled release of bioactive substance that integrates catalysis, material structures and function customization.
Collapse
Affiliation(s)
- Qiao Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Chaoqian Lou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Hang Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Yanyan Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Hongjie Zhang
- College of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zimeng Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Ganggang Qi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Xia Cai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Qiaojie Luo
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Lijie Fan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Xiaojun Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China.
| | - Weiwei Lao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China.
| | - Weipu Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Adsorption and Separation Materials & Technologies of Zhejiang Province, Hangzhou 310027, China
| | - Xiaodong Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China.
| |
Collapse
|
40
|
Zhang C, Li Q, Xing J, Yang Y, Zhu M, Lin L, Yu Y, Cai X, Wang X. Tannic acid and zinc ion coordination of nanase for the treatment of inflammatory bowel disease by promoting mucosal repair and removing reactive oxygen and nitrogen species. Acta Biomater 2024; 177:347-360. [PMID: 38373525 DOI: 10.1016/j.actbio.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/18/2024] [Accepted: 02/11/2024] [Indexed: 02/21/2024]
Abstract
Colon mucosal overexpression of reactive oxygen and nitrogen species (RONS) accelerates the development of inflammatory bowel disease (IBD) and destroys the mucosa and its barrier. IBD can be alleviated by removing RONS from the inflamed colon. The preparation of strong and efficient nanoantioxidants remains a challenge despite the development of numerous nanoantioxidants. In this paper, Zn-TA nanoparticles with fine hollow microstructure (HZn-TA) were successfully prepared and could be effectively used to treat IBD. In the first step, ZIF-8 nanoparticles were synthesized by a one-pot method. On this basis, HZn-TA nanoparticles were etched by TA, and a multifunctional nanase was developed for the treatment of IBD. RONS, including reactive oxygen species (ROS) and nitric oxide (NO), can be eliminated to increase cell survival following Hydrogen peroxide (H2O2) stimulation, including reactive oxygen species (ROS) and nitric oxide (NO with hydrogen peroxide (H2O2). In a model for preventing and delaying acute colitis, clearance of RONS has been shown to reduce intestinal inflammation in mice by reducing colon damage, proinflammatory cytokine levels, the spleen index, and body weight. Intestinal mucosal healing can be promoted by HZn-TA nanoparticles, which can upregulate zonula occludens protein 1 (ZO-1) and claudin-1 expression. Based on the results of this study, HZn-TA nanoparticles were able to effectively treat IBD with minimal adverse effects by being biocompatible, multienzyme active, and capable of scavenging RONS. Therefore, we pioneered the application of HZn-TA nanoparticles for the treatment of IBD, which are capable of clearing RONS without significant adverse effects. STATEMENT OF SIGNIFICANCE: ➢ HZn-TA nanoparticles were successfully prepared and could be effectively used to treat IBD. ➢ Intestinal mucosal healing can be promoted by HZn-TA nanoparticles, which can upregulate ZO-1 and claudin-1 expression. ➢ HZn-TA nanoparticles were able to effectively treat IBD with minimal adverse effects by being biocompatible, multienzyme active, and capable of scavenging RONS.
Collapse
Affiliation(s)
- Cong Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, PR China; Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230026, PR China
| | - Qingrong Li
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, PR China
| | - Jianghao Xing
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, PR China
| | - Yan Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Mengmei Zhu
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, PR China
| | - Liting Lin
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, PR China
| | - Yue Yu
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230026, PR China.
| | - Xiaojun Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, PR China.
| | - Xianwen Wang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei 230032, PR China; School of Biomedical Engineering, Anhui Medical University, Hefei 230032, PR China.
| |
Collapse
|
41
|
Liu R, Jiang D, Yun Y, Feng Z, Zheng F, Xiang Y, Fan H, Zhang J. Photoactivatable Engineering of CRISPR/Cas9-Inducible DNAzyme Probe for In Situ Imaging of Nuclear Zinc Ions. Angew Chem Int Ed Engl 2024; 63:e202315536. [PMID: 38253802 DOI: 10.1002/anie.202315536] [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: 10/15/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 01/24/2024]
Abstract
DNAzyme-based fluorescent probes for imaging metal ions in living cells have received much attention recently. However, employing in situ metal ions imaging within subcellular organelles, such as nucleus, remains a significant challenge. We developed a three-stranded DNAzyme probe (TSDP) that contained a 20-base-pair (20-bp) recognition site of a CRISPR/Cas9, which blocks the DNAzyme activity. When Cas9, with its specialized nuclear localization function, forms an active complex with sgRNA within the cell nucleus, it cleaves the TSDP at the recognition site, resulting in the in situ formation of catalytic DNAzyme structure. With this design, the CRISPR/Cas9-inducible imaging of nuclear Zn2+ is demonstrated in living cells. Moreover, the superiority of CRISPR-DNAzyme for spatiotemporal control imaging was demonstrated by integrating it with photoactivation strategy and Boolean logic gate for dynamic monitoring nuclear Zn2+ in both HeLa cells and mice. Collectively, this conceptual design expands the DNAzyme toolbox for visualizing nuclear metal ions and thus provides new analytical methods for nuclear metal-associated biology.
Collapse
Affiliation(s)
- Ran Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Difei Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu, 212003, China
| | - Yangfang Yun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Zhe Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Fenfen Zheng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Changhui Rd. 666, Zhenjiang, Jiangsu, 212003, China
| | - Yu Xiang
- Department of Chemistry, Beijing Key Laboratory for Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | - Huanhuan Fan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Jingjing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| |
Collapse
|
42
|
Park JM, Park S, Seo YS, Kim JH, Lee MY. Cytosolic zinc mediates the cytotoxicity of thiol-reactive electrophiles in rat vascular smooth muscle cells. Food Chem Toxicol 2024; 185:114446. [PMID: 38244666 DOI: 10.1016/j.fct.2024.114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/07/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
The aberrant increase or dysregulation of cytosolic Zn2+ concentration ([Zn2+]cyt) has been associated with cellular dysfunction and cytotoxicity. In this study, we postulated that Zn2+ mediates the cytotoxicity of thiol-reactive electrophiles. This notion was grounded on earlier research, which revealed that thiol-reactive electrophiles may disrupt Zn2+-binding motifs, consequently causing Zn2+ to be released from Zn2+-binding proteins, and leading to a surge in [Zn2+]cyt. The thiol-reactive electrophiles N-ethylmaleimide (NEM) and diamide were observed to induce an increase in [Zn2+]cyt, possibly through the impairment of Zn2+-binding motifs, and subsequent stimulation of reactive oxygen species (ROS) formation, resulting in cytotoxicity in primary cultured rat vascular smooth muscle cells. These processes were negated by the thiol donor N-acetyl-L-cysteine and the Zn2+ chelator TPEN. Similar outcomes were detected with co-treatment involving Zn2+ and Zn2+ ionophores such as pyrithione or disulfiram. Moreover, TPEN was found to inhibit cytotoxicity triggered by short-term exposure to various thiol-reactive electrophiles including hydrogen peroxide, acrylamide, acrylonitrile, diethyl maleate, iodoacetic acid, and iodoacetamide. In conclusion, our findings suggest that cytosolic Zn2+ acts as a universal mediator in the cytotoxic effects produced by thiol-reactive electrophiles.
Collapse
Affiliation(s)
- Jung-Min Park
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Suin Park
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Yoon-Seok Seo
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Jae-Hyeong Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Moo-Yeol Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
43
|
Huang Y, Guo X, Lu S, Chen Q, Wang Z, Lai L, Liu Q, Zhu X, Luo L, Li J, Huang Y, Gao H, Zhang Z, Bu Q, Cen X. Long-term exposure to cadmium disrupts neurodevelopment in mature cerebral organoids. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168923. [PMID: 38065485 DOI: 10.1016/j.scitotenv.2023.168923] [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: 10/06/2023] [Revised: 11/25/2023] [Accepted: 11/25/2023] [Indexed: 01/18/2024]
Abstract
Cadmium (Cd) is a pervasive environmental pollutant. Increasing evidence suggests that Cd exposure during pregnancy can induce adverse neurodevelopmental outcomes. However, due to the limitations of neural cell and animal models, it is challenging to study the developmental neurotoxicity and underlying toxicity mechanism of long-term exposure to environmental pollutants during human brain development. In this study, chronic Cd exposure was performed in human mature cerebral organoids for 49 or 77 days. Our study found that prolonged exposure to Cd resulted in the inhibition of cerebral organoid growth and the disruption of neural differentiation and cortical layer organization. These potential consequences of chronic Cd exposure may include impaired GFAP expression, a reduction in SOX2+ neuronal progenitor cells, an increase in TUJ1+ immature neurons, as well as an initial increase and a subsequent decrease in both TBR2+ intermediate progenitors and CTIP2+ deep layer cortical neurons. Transcriptomic analyses revealed that long-term exposure to Cd disrupted zinc and copper ion homeostasis through excessive synthesis of metallothionein and disturbed synaptogenesis, as evidenced by inhibited postsynaptic protein. Our study employed mature cerebral organoids to evaluate the developmental neurotoxicity induced by long-term Cd exposure.
Collapse
Affiliation(s)
- Yan Huang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xinhua Guo
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Shiya Lu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qiqi Chen
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqiu Wang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Li Lai
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Qian Liu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Xizhi Zhu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Li Luo
- Department of Gynaecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yina Huang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Gao
- Department of Food Science and Technology, College of Biomass and Engineering, Sichuan University, Chengdu 610065, China
| | - Zunzhen Zhang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qian Bu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
44
|
Bizup B, Brutsaert S, Cunningham CL, Thathiah A, Tzounopoulos T. Cochlear zinc signaling dysregulation is associated with noise-induced hearing loss, and zinc chelation enhances cochlear recovery. Proc Natl Acad Sci U S A 2024; 121:e2310561121. [PMID: 38354264 PMCID: PMC10895357 DOI: 10.1073/pnas.2310561121] [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: 06/22/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Exposure to loud noise triggers sensory organ damage and degeneration that, in turn, leads to hearing loss. Despite the troublesome impact of noise-induced hearing loss (NIHL) in individuals and societies, treatment strategies that protect and restore hearing are few and insufficient. As such, identification and mechanistic understanding of the signaling pathways involved in NIHL are required. Biological zinc is mostly bound to proteins, where it plays major structural or catalytic roles; however, there is also a pool of unbound, mobile (labile) zinc. Labile zinc is mostly found in vesicles in secretory tissues, where it is released and plays a critical signaling role. In the brain, labile zinc fine-tunes neurotransmission and sensory processing. However, injury-induced dysregulation of labile zinc signaling contributes to neurodegeneration. Here, we tested whether zinc dysregulation occurs and contributes to NIHL in mice. We found that ZnT3, the vesicular zinc transporter responsible for loading zinc into vesicles, is expressed in cochlear hair cells and the spiral limbus, with labile zinc also present in the same areas. Soon after noise trauma, ZnT3 and zinc levels are significantly increased, and their subcellular localization is vastly altered. Disruption of zinc signaling, either via ZnT3 deletion or pharmacological zinc chelation, mitigated NIHL, as evidenced by enhanced auditory brainstem responses, distortion product otoacoustic emissions, and number of hair cell synapses. These data reveal that noise-induced zinc dysregulation is associated with cochlear dysfunction and recovery after NIHL, and point to zinc chelation as a potential treatment for mitigating NIHL.
Collapse
Affiliation(s)
- Brandon Bizup
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA15261
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA15261
| | - Sofie Brutsaert
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA15261
| | - Christopher L. Cunningham
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA15261
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA15261
| | - Amantha Thathiah
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA15261
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA15261
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA15261
| |
Collapse
|
45
|
Baek Y, Lee M. Solid-state NMR spectroscopic analysis for structure determination of a zinc-bound catalytic amyloid fibril. Methods Enzymol 2024; 697:435-471. [PMID: 38816132 DOI: 10.1016/bs.mie.2024.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Zinc ions are commonly involved in enzyme catalysis and protein structure stabilization, but their coordination geometry of zinc-protein complex is rarely determined. Here, in this chapter, we introduce a systematic solid-state NMR approach to determine the oligomeric assembly and Zn2+ coordination geometry of a de novo designed amyloid fibrils that catalyze zinc dependent ester hydrolysis. NMR chemical shifts and intermolecular contacts confirm that the peptide forms parallel-in-register β-sheets, with the two forms of Zn2+ bound histidines in each peptide. The amphiphilic parallel β-sheets assemble into stacked bilayers that are stabilized by hydrophobic side chains between β-sheets. The conformations of the histidine side chains, determined by 13C-15N distance measurements, reveal how histidines protrude from the β-sheet. 1H-15N correlation spectra show that the single-Zn2+ coordinated histidine associated with dynamic water. The resulting structure provides insight into how metal ions contribute to stabilizing the protein structure and driving its catalytic reactivity.
Collapse
Affiliation(s)
- Yoongyeong Baek
- Department of Chemistry, Drexel University, Philadelphia, PA, United States
| | - Myungwoon Lee
- Department of Chemistry, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
46
|
Bennett MC, Reinhart KM, Weisend JE, Morton RA, Carlson AP, Shuttleworth CW. Synaptic Zn 2+ contributes to deleterious consequences of spreading depolarizations. Neurobiol Dis 2024; 191:106407. [PMID: 38199272 PMCID: PMC10869643 DOI: 10.1016/j.nbd.2024.106407] [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: 10/26/2023] [Revised: 12/23/2023] [Accepted: 01/07/2024] [Indexed: 01/12/2024] Open
Abstract
Spreading depolarizations (SDs) are profound waves of neuroglial depolarization that can propagate repetitively through injured brain. Recent clinical work has established SD as an important contributor to expansion of acute brain injuries and have begun to extend SD studies into other neurological disorders. A critical challenge is to determine how to selectively prevent deleterious consequences of SD. In the present study, we determined whether a wave of profound Zn2+ release is a key contributor to deleterious consequences of SD, and whether this can be targeted pharmacologically. Focal KCl microinjection was used to initiate SD in the CA1 region of the hippocampus in murine brain slices. An extracellular Zn2+ chelator with rapid kinetics (ZX1) increased SD propagation rates and improved recovery of extracellular DC potential shifts. Under conditions of metabolic compromise, tissues showed sustained impairment of functional and structural recovery following a single SD. ZX1 effectively improved recovery of synaptic potentials and intrinsic optical signals in these vulnerable conditions. Fluorescence imaging and genetic deletion of a presynaptic Zn2+ transporter confirmed synaptic release as the primary contributor to extracellular accumulation and deleterious consequences of Zn2+ during SD. These results demonstrate a role for synaptic Zn2+ release in deleterious consequences of SD and show that targeted extracellular chelation could be useful for disorders where repetitive SD enlarges infarcts in injured tissues.
Collapse
Affiliation(s)
- Michael C Bennett
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Katelyn M Reinhart
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Jordan E Weisend
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Russell A Morton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Andrew P Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| |
Collapse
|
47
|
Li H, Hao J, Liu X. Research progress and perspective of metallic implant biomaterials for craniomaxillofacial surgeries. Biomater Sci 2024; 12:252-269. [PMID: 38170634 DOI: 10.1039/d2bm01414a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Craniomaxillofacial bone serves a variety of functions. However, the increasing number of cases of craniomaxillofacial bone injury and the use of selective rare implants make the treatment difficult, and the cure rate is low. If such a bone injury is not properly treated, it can lead to a slew of complications that can seriously disrupt a patient's daily life. For example, premature closure of cranial sutures or skull fractures can lead to increased intracranial pressure, which can lead to headaches, vomiting, and even brain hernia. At present, implant placement is one of the most common approaches to repair craniomaxillofacial bone injury or abnormal closure, especially with biomedical metallic implants. This review analyzes the research progress in the design and development of degradable and non-degradable metallic implants in craniomaxillofacial surgery. The mechanical properties, corrosion behaviours, as well as in vitro and in vivo performances of these materials are summarized. The challenges and future research directions of metallic biomaterials used in craniomaxillofacial surgery are also identified.
Collapse
Affiliation(s)
- Huafang Li
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Jiaqi Hao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Xiwei Liu
- Lepu Medical Technology Co., Ltd, Beijing 102200, China
| |
Collapse
|
48
|
Alshehri RF, El-Feky HH, Askar AM, Amin AS, Aish M. Utilization of a novel PVC- optical sensor for high sensitive and selective determination of zinc ion in real samples. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 305:123424. [PMID: 37806243 DOI: 10.1016/j.saa.2023.123424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/24/2023] [Accepted: 09/16/2023] [Indexed: 10/10/2023]
Abstract
A novel and highly specific bulk membrane optode was developed for the ultra-sensitive detection of zinc(II) in biological, pharmaceutical, and water samples. The polymer inclusion membrane (PIM) utilized in this study consists of 50% polyvinyl chloride (PVC) as a base polymer, 9.00% DOP (dioctylphthalate) as the plasticizer, and 40.0% D2EHPA (di(2-ethlyhexyl)phosphoric acid) as the carrier. To facilitate the spectrophotometric determination of zinc(II), a colorimetric reagent, namely 4-(2-arsonophenylazo) salicylic acid (APASA) {1.00%, m/v}, was employed. When Zn(II) was extracted into the PIM, it led to the creation of the zinc-D2EHPA complex. This complex then underwent a reaction with APASA, resulting in the formation of a red Zn - APASA complex with a maximum absorption wavelength (λmax) of 558 nm. To optimize the response of the optode, a central composite design was employed, considering variables such as the amount of additive and reagent, response time, and pH. When operated under the specific optimal conditions, the sensor demonstrated a limit of quantification (LOQ) of 0.74 ng/mL (equivalent to 1.17 × 10-8 M) and a limit of detection (LOD) of 0.22 ng/mL (equivalent to 3.44 × 10-9 M). The optode membrane demonstrated excellent reproducibility, stability, and a relatively long lifespan, making it suitable for precise and accurate monitoring of Zn(II) ion content. Regeneration of the optode was achieved effectively using 0.25 nitric acid solution, and its response exhibited reversibility and reproducibility, showed a relative standard deviation of less than 1.33%. Moreover, the PIM-APASA optode exhibited a high level of effectiveness in accurately determining the presence of Zn(II) ions in real environmental samples.
Collapse
Affiliation(s)
- Reem F Alshehri
- Chemistry Department, College of Science& Arts, Taibah University, Saudi Arabia
| | - Hesham H El-Feky
- Chemistry Department, Faculty of Science, Benha University, Benha, Egypt
| | - Abdelrazek M Askar
- Chemistry Department, Faculty of Science, Benha University, Benha, Egypt
| | - Alaa S Amin
- Chemistry Department, Faculty of Science, Benha University, Benha, Egypt.
| | - Mai Aish
- Chemistry Department, Faculty of Science, Port Said University, Port Said, Egypt
| |
Collapse
|
49
|
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
|
50
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|