1
|
Chang SA, Balouch A, Nahyoon WA, Raja AA. Detection of Trace Elements Se, Cu, and Zn Levels in Biological Samples of Drug Abusers: Halani Sindh, Pakistan. Biol Trace Elem Res 2024; 202:4869-4876. [PMID: 38196054 DOI: 10.1007/s12011-023-04056-6] [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: 10/12/2023] [Accepted: 12/30/2023] [Indexed: 01/11/2024]
Abstract
Illicit drug use has become a global issue, posing significant health, socioeconomic, and cultural risks. The study examined essential trace metals: selenium, zinc, and copper in blood concentrations, as well as in serum and scalp hair samples, from 240 male drug-abuse subjects/patients aged 18-45, categorized into three age groups. The study compared 45 healthy subjects of the same age group using an acid digestion method supported by a microwave oven during sample preparation. The technique of atomic absorption spectrometry was employed to identify essential and toxic elements, utilizing certified reference materials for accuracy. According to a recent study, plasma zinc and selenium concentrations in drug abusers are lower than those in referent subjects, potentially increasing vulnerability to infection due to poor nutritional status or other contaminants.
Collapse
Affiliation(s)
- Sajjad Ali Chang
- National Centre of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan.
- Institute of Forensic Sciences Forensic Medicine and Toxicology, Liaquat University of Medical and Health Sciences, Jamshoro, 76090, Pakistan.
| | - Aamna Balouch
- National Centre of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Waheed Ali Nahyoon
- Institute of Forensic Sciences Forensic Medicine and Toxicology, Liaquat University of Medical and Health Sciences, Jamshoro, 76090, Pakistan
| | - Aamir Ali Raja
- Jeejal Mau Institute of Physiotherapy and Rehabilitation Sciences, Liaquat University of Medical and Health Sciences, Jamshoro, 76090, Pakistan
| |
Collapse
|
2
|
Mori H, Goji A, Hara M. Upregulation of Intracellular Zinc Ion Level after Differentiation of the Neural Stem/Progenitor Cells In Vitro with the Changes in Gene Expression of Zinc Transporters. Biol Trace Elem Res 2024; 202:4699-4714. [PMID: 38180597 DOI: 10.1007/s12011-023-04033-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024]
Abstract
We measured the intracellular zinc ion concentration of murine fetal neural stem/progenitor cells (NSPCs) and that in the differentiated cells. The NSPCs cultured with 1.5 μM Zn2+ proliferated slightly faster than that in the zinc-deficient medium and the intracellular zinc concentration of the NSPCs and that of their differentiated cells (DCs) cultured with 1.5 μM Zn2+ was 1.34-fold and 2.00-fold higher than those in the zinc-deficient medium, respectively. The zinc transporter genes upregulated over the 3.5-fold change were Zip1, Zip4, Zip12, Zip13, ZnT1, ZnT8, and ZnT10 whereas the only downregulated one was Zip8 during the differentiation of NSPCs to DCs. The cell morphologies of both NSPCs and DCs in the low oxygen culture condition consisting of 2%O2 and 5%CO2, the high carbon dioxide condition consisting of 21%O2 and 10%CO2, and the normal condition consisting of 21%O2 and 5%CO2 were essentially the same each other. The expression of Zip4, Zip8, Zip12, and Zip14 was not drastically changed depending on the O2 and CO2 concentrations.
Collapse
Affiliation(s)
- Hideki Mori
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1 Gakuencho, Nakaku, Sakai, Osaka, 599-8531, Japan
| | - Akari Goji
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1 Gakuencho, Nakaku, Sakai, Osaka, 599-8531, Japan
| | - Masayuki Hara
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1 Gakuencho, Nakaku, Sakai, Osaka, 599-8531, Japan.
| |
Collapse
|
3
|
Zhao S, Wang L, Liang J, Jin F, Wang F. Preparation, characterization and microencapsulation of walnut (Juglans regia L.) peptides-zinc chelate. J Food Sci 2024; 89:5618-5632. [PMID: 39126687 DOI: 10.1111/1750-3841.17160] [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: 01/12/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 08/12/2024]
Abstract
In this research, a novel kind of walnut (Juglans regia L.) peptides-zinc (Zn-WPs) chelate was obtained using the mass ratio of the walnut peptides (WPs) to ZnSO4.7H2O of 3.5:1 at pH 8.5 and 50°C for 84 min, with the chelation rate of 84.5%. In comparison to walnut peptides (WPs), the contents of aspartic acid and glutamic acid in Zn-WPs chelate are approximately 27%, indicating that hydrophilic amino acids predominantly bind with walnut peptides. Following chelation with zinc ions, the ultraviolet-visible (UV) characteristic absorption peak shifted from 213 nm to 210 nm, while the average particle size of the chelate increased to 8.0 ± 0.14 µm, presenting a loose spherical structure under scanning electron microscopy. These findings suggest the formation of new substances. Fourier-transform infrared spectroscopy (FTIR) revealed carboxyl, amino, and peptide bonds as the chelation sites of WPs and zinc. The IC50 of walnut peptides-zinc (Zn-WPs) chelate is 2.91 mg/mL, indicative of a favorable DPPH radical scavenging rate. Furthermore, Zn-WPs chelate microcapsules were produced via the spray drying method, achieving an encapsulation rate of 75.67 ± 0.83% under optimal conditions. These microcapsules demonstrate robust stability across diverse environmental conditions. This study underscores the potential of Zn-WPs and its chelate microcapsules to enhance stability and bioactivity under varying circumstances. PRACTICAL APPLICATION: In this study, a new walnut peptide-zinc (Zn-WPs) chelate was prepared. The presence of zinc ions changes the structure and properties of walnut peptides and improves its stability. The production of Zn-WPs chelate microcapsules enables Zn-WPs to have strong in vitro stability under different pH and simulated gastrointestinal digestion conditions. These results provide novel insights for developing the walnut peptides as bioactive ingredients in functional foods.
Collapse
Affiliation(s)
- Sibao Zhao
- National Key Laboratory for Efficient Production of Forest Resources, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Lei Wang
- National Key Laboratory for Efficient Production of Forest Resources, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Jingyi Liang
- National Key Laboratory for Efficient Production of Forest Resources, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Feng Jin
- National Key Laboratory for Efficient Production of Forest Resources, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Fengjun Wang
- National Key Laboratory for Efficient Production of Forest Resources, Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| |
Collapse
|
4
|
Dean B, Hopper S, Scarr E. Changes in levels of the zinc transporter SLC39A12 in Brodmann's area 44: Effects of sex, suicide, CNS pH and schizophrenia. J Psychiatr Res 2024; 177:177-184. [PMID: 39024742 DOI: 10.1016/j.jpsychires.2024.07.017] [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: 04/29/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Disturbed CNS zinc homeostasis is suggested as part of the pathophysiology of schizophrenia. Our data, from multiple studies, suggests levels of cortical RNA for the solute carrier family 39 member 12 (SLC39A12), a putative zinc transporter, is higher in people with schizophrenia and is more perturbed in a sub-group of people with the disorder that can be separated because they have very low levels of muscarinic M1 receptors (MRDS). In this study qPCR was used to measure levels of two RNA splice variants of SLC39A12 (a and b) in Brodmann's area (BA) 44 from new cohorts of controls and people with schizophrenia. For the first time, in our study cohort as a whole, we report levels of both splice variants of SLC39A12 are lower in females compared to males and there are correlations between levels of SLC39A12 a and b and CNS pH. Levels of both splice variants were also lower in people with schizophrenia who were suicide completers compared to those who were not. Accounting for these factors, we showed levels of SLC39A12 a and b were higher in BA 44 in schizophrenia compared to controls. In further analyses, with and without our previous data on SLC39A12 a and b, we confirmed changes in levels of SLC39A12 RNAs were more profound in MRDS. In conclusion, our study argues there are higher levels of SLC39A12 a and b in BA 44 in schizophrenia which could be contributing to the breakdown in CNS zinc homeostasis suggested as part of the pathophysiology of schizophrenia, particularly in those with MRDS.
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| | - Shaun Hopper
- The Molecular Psychiatry Laboratory, The Florey, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
5
|
Chen H, Zheng Z, Cai X, Gao F. Causal links between serum micronutrients and epilepsy: a Mendelian randomization analysis. Front Neurol 2024; 15:1419289. [PMID: 39076846 PMCID: PMC11284170 DOI: 10.3389/fneur.2024.1419289] [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: 04/18/2024] [Accepted: 06/26/2024] [Indexed: 07/31/2024] Open
Abstract
Background Micronutrient levels play a critical role in epilepsy. This study investigates the impact of micronutrient levels on epilepsy via Mendelian randomization (MR). Methods A two-sample MR framework evaluated the genetic association between 15 serum micronutrients and epilepsy phenotypes. The analysis included calcium, iron, zinc, selenium, copper, magnesium, potassium, folate, vitamins B6, B12, C, D, E, retinol, and carotene against all epilepsy, generalized epilepsy, childhood absence epilepsy (CAE), juvenile absence epilepsy (JAE), juvenile myoclonic epilepsy (JME), generalized tonic-clonic seizures alone and with spike-wave electroencephalography (GTCS), and various focal epilepsy phenotypes [with hippocampal sclerosis (HS), lesions other than HS, lesion-negative]. The random-effects inverse-variance weighted (IVW) model was the primary method used, supported by heterogeneity and pleiotropy assessments. Multivariable Mendelian randomization analyses (MVMR) were used to identify micronutrients that are significantly causally associated with different epilepsy subtypes and to confirm the most potential causal risk factors for these subtypes. Results Zinc conferred an increased risk of focal epilepsy with HS (OR = 1.01; p = 0.045). Carotene was similarly linked to higher risks of lesion-negative cases (OR = 1.129; p = 0.037). Conversely, vitamin B6 was associated with reduced risks of focal epilepsy with HS (OR = 0.949; p = 0.020), and vitamin D was linked to decreased risks of both CAE (OR = 0.976, 95% CI: 0.959-0.993, p = 0.006) and JAE (OR = 0.986, 95% CI: 0.973-0.999, p = 0.032). These associations were robust, showing minimal heterogeneity and no evidence of pleiotropy across various sensitivity analyses. After adjustment using MVMR, significant causal relationships between vitamin D and both CAE and JAE remained. Furthermore, the causal relationship between zinc and vitamin B6 on focal epilepsy with HS became non-significant, while carotene shifted from a risk factor to a protective factor for focal epilepsy lesion-negative after adjusting for vitamin D. Conclusion MR estimates provide robust evidence for the causal effects of vitamin D on reducing the risk of CAE, and JAE, which might provide alternative treatment strategies.
Collapse
Affiliation(s)
- Haohao Chen
- Department of Pharmacy, Shantou University Medical College, Shantou, Guangdong Province, China
- Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Zequn Zheng
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Xiaorui Cai
- Department of Pharmacy, The Affiliated Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Fenfei Gao
- Department of Pharmacy, Shantou University Medical College, Shantou, Guangdong Province, China
| |
Collapse
|
6
|
Hettiarachchi P, Niyangoda S, Shigemoto A, Solowiej IJ, Burdette SC, Johnson MA. Caged Zn 2+ Photolysis in Zebrafish Whole Brains Reveals Subsecond Modulation of Dopamine Uptake. ACS Chem Neurosci 2024; 15:772-782. [PMID: 38301116 PMCID: PMC11036533 DOI: 10.1021/acschemneuro.3c00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
Free, ionic zinc (Zn2+) modulates neurotransmitter dynamics in the brain. However, the sub-s effects of transient concentration changes of Zn2+ on neurotransmitter release and uptake are not well understood. To address this lack of knowledge, we have combined the photolysis of the novel caged Zn2+ compound [Zn(DPAdeCageOMe)]+ with fast scan cyclic voltammetry (FSCV) at carbon fiber microelectrodes in live, whole brain preparations from zebrafish (Danio rerio). After treating the brain with [Zn(DPAdeCageOMe)]+, Zn2+ was released by application of light that was gated through a computer-controlled shutter synchronized with the FSCV measurements and delivered through a 1 mm fiber optic cable. We systematically optimized the photocage concentration and light application parameters, including the total duration and light-to-electrical stimulation delay time. While sub-s Zn2+ application with this method inhibited DA reuptake, assessed by the first-order rate constant (k) and half-life (t1/2), it had no effect on the electrically stimulated DA overflow ([DA]STIM). Increasing the photocage concentration and light duration progressively inhibited uptake, with maximal effects occurring at 100 μM and 800 ms, respectively. Furthermore, uptake was inhibited 200 ms after Zn2+ photorelease, but no measurable effect occurred after 800 ms. We expect that application of this method to the zebrafish whole brain and other preparations will help expand the current knowledge of how Zn2+ affects neurotransmitter release/uptake in select neurological disease states.
Collapse
Affiliation(s)
- Piyanka Hettiarachchi
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Sayuri Niyangoda
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Austin Shigemoto
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA 01609
| | - Isabel J. Solowiej
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Shawn C. Burdette
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA 01609
| | - Michael A. Johnson
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
7
|
Bai HH, Wang KL, Zeng XR, Li J, Li Y, Xu JY, Zhang Y, Jiang HF, Yang X, Suo ZW, Hu XD. GPR39 regulated spinal glycinergic inhibition and mechanical inflammatory pain. SCIENCE ADVANCES 2024; 10:eadj3808. [PMID: 38306424 PMCID: PMC10836721 DOI: 10.1126/sciadv.adj3808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
G protein-coupled receptor 39 (GPR39) senses the change of extracellular divalent zinc ion and signals through multiple G proteins to a broad spectrum of downstream effectors. Here, we found that GPR39 was prevalent at inhibitory synapses of spinal cord somatostatin-positive (SOM+) interneurons, a mechanosensitive subpopulation that is critical for the conveyance of mechanical pain. GPR39 complexed specifically with inhibitory glycine receptors (GlyRs) and helped maintain glycinergic transmission in a manner independent of G protein signalings. Targeted knockdown of GPR39 in SOM+ interneurons reduced the glycinergic inhibition and facilitated the excitatory output from SOM+ interneurons to spinoparabrachial neurons that engaged superspinal neural circuits encoding both the sensory discriminative and affective motivational domains of pain experience. Our data showed that pharmacological activation of GPR39 or augmenting GPR39 interaction with GlyRs at the spinal level effectively alleviated the sensory and affective pain induced by complete Freund's adjuvant and implicated GPR39 as a promising therapeutic target for the treatment of inflammatory mechanical pain.
Collapse
Affiliation(s)
- Hu-Hu Bai
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- School of Life Science, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Kang-Li Wang
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiang-Ru Zeng
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jing Li
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yuan Li
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jia-Yu Xu
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yue Zhang
- School of Public Health, Gansu University of Chinese medicine, Lanzhou, Gansu 730000, P.R. China
| | - Hai-Feng Jiang
- School of Public Health, Gansu University of Chinese medicine, Lanzhou, Gansu 730000, P.R. China
| | - Xian Yang
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhan-Wei Suo
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiao-Dong Hu
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
8
|
Yu J, Ding Y, Wu D, Liu P. Rutin, Puerarin and Silymarin Regulated Aluminum-Induced Imbalance of Neurotransmitters and Metal Elements in Brain of Rats. Biol Trace Elem Res 2024; 202:548-557. [PMID: 37289414 DOI: 10.1007/s12011-023-03682-4] [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: 02/19/2023] [Accepted: 04/22/2023] [Indexed: 06/09/2023]
Abstract
Non-specifically binding of aluminum to various substances in the organism can result in toxicity. The accumulation of large amounts of aluminum can cause an imbalance in metal homeostasis and interfere with the synthesis and release of neurotransmitters. Flavonoids have strong metal chelating activity, which can reduce damage to the central nervous system. The purpose of this study was to investigate the protective effect of three representative flavonoids, rutin, puerarin and silymarin, on the brain toxicity induced by long-term exposure to aluminum trichloride (AlCl3). Sixty-four Wistar rats were randomly divided into eight groups (n = 8). The rats in six intervention groups were given 100 or 200 mg/kg BW/day of three different flavonoids for four weeks after a 4-week exposure to 281.40 mg/kg BW/day AlCl3·6H2O, while the rats in the AlCl3-toxicity and control groups were given the vehicle after the period of AlCl3 exposure. The results showed that rutin, puerarin, and silymarin could increase the concentrations of magnesium, iron, and zinc in the brains of the rats. Moreover, the intake of these three flavonoids regulated the homeostasis of amino acid neurotransmitters and adjusted the concentrations of monoamine neurotransmitters to normal levels. Taken together, our data suggest that rutin, puerarin, and silymarin could ameliorate AlCl3-induced brain toxicity in the rats by regulating imbalance of metal elements and neurotransmitters in the brains of rats.
Collapse
Affiliation(s)
- Jiasi Yu
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yun Ding
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dan Wu
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Ping Liu
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
9
|
Caraba IV, Caraba MN, Hutanu D, Sinitean A, Dumitrescu G, Popescu R. Trace Metal Accumulation in Rats Exposed to Mine Waters: A Case Study, Bor Area (Serbia). TOXICS 2023; 11:960. [PMID: 38133361 PMCID: PMC10748338 DOI: 10.3390/toxics11120960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023]
Abstract
Zinc (Zn), copper (Cu), iron (Fe), manganese (Mn), cadmium (Cd), and lead (Pb) levels were measured in the Bor City water supply system (control) and two watercourses exposed to mining wastewaters, i.e., the Lutarica River (one site) and the Kriveljska River (two sites). The same parameters were determined in the brain, heart, lungs, stomach, liver, spleen, kidneys, and testes of male Wistar rats given water from these sources for 2 months. Water Cu, Fe, Cd, and Pb were outside the safe range, excepting the reference site. Significant impacts on intra-organ metal homeostasis were detected, especially in the brain, stomach, kidneys, and testes. The dynamics and magnitude of these changes (versus controls) depended on the target organ, analyzed metal, and water origin. The greatest number of significant intra-organ associations between essential and non-essential metals were found for Cd-Zn, Cd-Cu, and Cd-Mn. A regression analysis suggested the kidneys as the most relevant organ for monitoring water manganese, and the stomach and brain for lead. These results highlight the environmental risks associated with mining wastewaters from the Bor area and could help scientists in mapping the spatial distribution and severity of trace metal contamination in water sources.
Collapse
Affiliation(s)
- Ion Valeriu Caraba
- Faculty of Bioengineering of Animal Resources, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului, 119, 300645 Timisoara, Romania; (I.V.C.); (G.D.)
| | - Marioara Nicoleta Caraba
- Department Biology-Chemistry, Faculty of Chemistry, Biology, Geography, West University of Timisoara, Pestalozzi 16, 300315 Timisoara, Romania; (D.H.); (A.S.)
| | - Delia Hutanu
- Department Biology-Chemistry, Faculty of Chemistry, Biology, Geography, West University of Timisoara, Pestalozzi 16, 300315 Timisoara, Romania; (D.H.); (A.S.)
| | - Adrian Sinitean
- Department Biology-Chemistry, Faculty of Chemistry, Biology, Geography, West University of Timisoara, Pestalozzi 16, 300315 Timisoara, Romania; (D.H.); (A.S.)
| | - Gabi Dumitrescu
- Faculty of Bioengineering of Animal Resources, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului, 119, 300645 Timisoara, Romania; (I.V.C.); (G.D.)
| | - Roxana Popescu
- ANAPATMOL Research Center, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| |
Collapse
|
10
|
Benarroch E. What Are the Functions of Zinc in the Nervous System? Neurology 2023; 101:714-720. [PMID: 37845046 PMCID: PMC10585682 DOI: 10.1212/wnl.0000000000207912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 10/18/2023] Open
|
11
|
Wang B, Fang T, Chen H. Zinc and Central Nervous System Disorders. Nutrients 2023; 15:2140. [PMID: 37432243 DOI: 10.3390/nu15092140] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 07/12/2023] Open
Abstract
Zinc (Zn2+) is the second most abundant necessary trace element in the human body, exerting a critical role in many physiological processes such as cellular proliferation, transcription, apoptosis, growth, immunity, and wound healing. It is an essential catalyst ion for many enzymes and transcription factors. The maintenance of Zn2+ homeostasis is essential for the central nervous system, in which Zn2+ is abundantly distributed and accumulates in presynaptic vesicles. Synaptic Zn2+ is necessary for neural transmission, playing a pivotal role in neurogenesis, cognition, memory, and learning. Emerging data suggest that disruption of Zn2+ homeostasis is associated with several central nervous system disorders including Alzheimer's disease, depression, Parkinson's disease, multiple sclerosis, schizophrenia, epilepsy, and traumatic brain injury. Here, we reviewed the correlation between Zn2+ and these central nervous system disorders. The potential mechanisms were also included. We hope that this review can provide new clues for the prevention and treatment of nervous system disorders.
Collapse
Affiliation(s)
- Bangqi Wang
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China
| | - Tianshu Fang
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China
| | - Hongping Chen
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
12
|
Ortho-Vanillin based multifunctional scaffold for selective detection of Al3+ and Zn2+ employing molecular logic with DFT study and cell imaging with live Grass pea. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2023.114663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
13
|
Abstract
Anorexia nervosa is a disorder associated with serious adverse health outcomes, for which there is currently considerable treatment ineffectiveness. Characterised by restrictive eating behaviours, distorted body image perceptions and excessive physical activity, there is growing recognition anorexia nervosa is associated with underlying dysfunction in excitatory and inhibitory neurometabolite metabolism and signalling. This narrative review critically explores the role of N-methyl-D-aspartate receptor-mediated excitatory and inhibitory neurometabolite dysfunction in anorexia nervosa and its associated biomarkers. The existing magnetic resonance spectroscopy literature in anorexia nervosa is reviewed and we outline the brain region-specific neurometabolite changes that have been reported and their connection to anorexia nervosa psychopathology. Considering the proposed role of dysfunctional neurotransmission in anorexia nervosa, the potential utility of zinc supplementation and sub-anaesthetic doses of ketamine in normalising this is discussed with reference to previous research in anorexia nervosa and other neuropsychiatric conditions. The rationale for future research to investigate the combined use of low-dose ketamine and zinc supplementation to potentially extend the therapeutic benefits in anorexia nervosa is subsequently explored and promising biological markers for assessing and potentially predicting treatment response are outlined.
Collapse
|
14
|
Li Z, Liu Y, Wei R, Yong VW, Xue M. The Important Role of Zinc in Neurological Diseases. Biomolecules 2022; 13:28. [PMID: 36671413 PMCID: PMC9855948 DOI: 10.3390/biom13010028] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Zinc is one of the most abundant metal ions in the central nervous system (CNS), where it plays a crucial role in both physiological and pathological brain functions. Zinc promotes antioxidant effects, neurogenesis, and immune system responses. From neonatal brain development to the preservation and control of adult brain function, zinc is a vital homeostatic component of the CNS. Molecularly, zinc regulates gene expression with transcription factors and activates dozens of enzymes involved in neuronal metabolism. During development and in adulthood, zinc acts as a regulator of synaptic activity and neuronal plasticity at the cellular level. There are several neurological diseases that may be affected by changes in zinc status, and these include stroke, neurodegenerative diseases, traumatic brain injuries, and depression. Accordingly, zinc deficiency may result in declines in cognition and learning and an increase in oxidative stress, while zinc accumulation may lead to neurotoxicity and neuronal cell death. In this review, we explore the mechanisms of brain zinc balance, the role of zinc in neurological diseases, and strategies affecting zinc for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| | - Ruixue Wei
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| | - V. Wee Yong
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| |
Collapse
|
15
|
Abstract
Growing evidence indicates that a suboptimal intrauterine environment confers risk for schizophrenia. The developmental model of schizophrenia posits that aberrant brain growth during early brain development and adolescence may interact to contribute to this psychiatric disease in adulthood. Although a variety of factors may perturb the environment of the developing fetus and predispose for schizophrenia later, a common mechanism has yet to be elucidated. Micronutrient deficiencies during the perinatal period are known to induce potent effects on brain development by altering neurodevelopmental processes. Iron is an important candidate nutrient to consider because of its role in energy metabolism, monoamine synthesis, synaptogenesis, myelination, and the high prevalence of iron deficiency (ID) in the mother-infant dyad. Understanding the current state of science regarding perinatal ID as an early risk factor for schizophrenia is imperative to inform empirical work investigating the etiology of schizophrenia and develop prevention and intervention programs. In this narrative review, we focus on perinatal ID as a common mechanism underlying the fetal programming of schizophrenia. First, we review the neural aberrations associated with perinatal ID that indicate risk for schizophrenia in adulthood, including disruptions in dopaminergic neurotransmission, hippocampal-dependent learning and memory, and sensorimotor gating. Second, we review the pathophysiology of perinatal ID as a function of maternal ID during pregnancy and use epidemiological and cohort studies to link perinatal ID with risk of schizophrenia. Finally, we review potential confounding phenotypes, including nonanemic causes of perinatal brain ID and future risk of schizophrenia.
Collapse
Affiliation(s)
- Andrea M. Maxwell
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN 55455 (USA)
| | - Raghavendra B. Rao
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55455 (USA)
- Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN 55455 (USA)
| |
Collapse
|
16
|
Sun J, Xu S, Du Y, Yu K, Jiang Y, Weng H, Yuan W. Accumulation and Enrichment of Trace Elements by Yeast Cells and Their Applications: A Critical Review. Microorganisms 2022; 10:1746. [PMID: 36144348 PMCID: PMC9504137 DOI: 10.3390/microorganisms10091746] [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: 07/21/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/24/2022] Open
Abstract
Maintaining the homeostasis balance of trace elements is crucial for the health of organisms. Human health is threatened by diseases caused by a lack of trace elements. Saccharomyces cerevisiae has a wide and close relationship with human daily life and industrial applications. It can not only be used as fermentation products and single-cell proteins, but also as a trace elements supplement that is widely used in food, feed, and medicine. Trace-element-enriched yeast, viz., chromium-, iron-, zinc-, and selenium-enriched yeast, as an impactful microelements supplement, is more efficient, more environmentally friendly, and safer than its inorganic and organic counterparts. Over the last few decades, genetic engineering has been developing large-scaled genetic re-design and reconstruction in yeast. It is hoped that engineered yeast will include a higher concentration of trace elements. In this review, we compare the common supplement forms of several key trace elements. The mechanisms of detoxification and transport of trace elements in yeast are also reviewed thoroughly. Moreover, genes involved in the transport and detoxification of trace elements are summarized. A feasible way of metabolic engineering transformation of S. cerevisiae to produce trace-element-enriched yeast is examined. In addition, the economy, safety, and environmental protection of the engineered yeast are explored, and the future research direction of yeast enriched in trace elements is discussed.
Collapse
Affiliation(s)
- Jie Sun
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shiyi Xu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yongbao Du
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Kechen Yu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yi Jiang
- Hangzhou Se-easy Biotechnology Co., Ltd., Hangzhou 311100, China
| | - Hao Weng
- Hangzhou Se-easy Biotechnology Co., Ltd., Hangzhou 311100, China
| | - Wei Yuan
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
17
|
Teil M, Doudnikoff E, Thiolat ML, Bohic S, Bezard E, Dehay B. The Zinc Ionophore Clioquinol Reduces Parkinson's Disease Patient-Derived Brain Extracts-Induced Neurodegeneration. Mol Neurobiol 2022; 59:6245-6259. [PMID: 35915387 DOI: 10.1007/s12035-022-02974-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) is pathologically characterized by intracellular α-synuclein-rich protein aggregates, named Lewy bodies (LB), and by the progressive loss of dopaminergic neurons in the substantia nigra. Several heavy metals, including zinc (Zn), have been suggested to play a role in PD progression, although the exact role of Zn in neurodegeneration remains to be fully elucidated. To address this gap, we investigated the effects of Zn modulation on the progression of degeneration in mice injected with PD patient-derived LB-extracts carrying toxic α-synuclein aggregates. Zn modulation was achieved using either a clioquinol-enriched diet, a Zn ionophore that redistributes cellular Zn, or a Zn-enriched diet that increases Zn levels. Clioquinol treatment significantly prevented dopaminergic neurodegeneration and reduced α-synuclein-associated pathology in LB-injected mice, while no differences were observed with Zn supplementation. Biochemical analyses further demonstrate that the expression levels of vesicle-specific Zn transporter ZnT3 in the striatum of LB-injected mice treated with clioquinol were decreased, suggesting an intracellular redistribution of Zn. Additionally, we found that clioquinol modulates the autophagy-lysosomal pathway by enhancing lysosomal redistribution within the neuronal compartments. Collectively, we found that in vivo pharmacological chelation of Zn, by dampening Zn-mediated cytotoxicity, can result in an overall attenuation of PD-linked lysosomal alterations and dopaminergic neurodegeneration. The results support zinc chelation as a disease-modifying strategy for treating PD.
Collapse
Affiliation(s)
- Margaux Teil
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
| | | | | | - Sylvain Bohic
- Synchrotron Radiation for Biomedicine (STROBE), Univ. Grenoble Alpes, Inserm, UA7, 38000, Grenoble, France
| | - Erwan Bezard
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
| | - Benjamin Dehay
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France.
- Institut des Maladies Neurodégénératives, Université de Bordeaux, CNRS UMR 5293, Centre Broca Nouvelle-Aquitaine, 146 rue Léo Saignat, 33076, Bordeaux cedex, France.
| |
Collapse
|
18
|
Sun Y, Zhao J, Song X, Sun Z, Zhang R, Zhong J, Huang X, Dong Y, Yu Q, Dong F, Li Z, Fan L, Wang M, Peng C, Wang F. Effects of marginal zinc deficiency on learning and memory ability after birth. Food Funct 2022; 13:7204-7214. [PMID: 35713090 DOI: 10.1039/d2fo01074g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Zinc deficiency during pregnancy and severe zinc deficiency after birth both impaired learning and memory ability, but the effects of marginal zinc deficiency (MZD) after birth on learning and memory are unclear. In the first experiment, 4-week-old male rats were randomly divided into three groups: the marginal zinc-deficient group (MZG, 10 mg kg-1, 1/3 RNI), normal zinc group (NZG, 30 mg kg-1, RNI), and paired zinc group (PZG, 30 mg kg-1). After a 4-week feeding period, the brain weight, brain coefficient, and serum zinc concentration were measured, and hippocampal proteomics analysis was performed. In the second experiment, 4-week-old male rats were fed the same diet for 8 weeks. In addition to the previously mentioned indicators, the Morris water maze results, brain pathology, post-translational modifications (PTMs) of hippocampal proteins, and the concentrations of indicators known to be related to learning and memory were analyzed. In both experiments, compared with those of the NZG, the food intake, body weight and serum zinc of the MZG were significantly decreased, and the brain weight was unchanged, but the brain coefficient was increased. Two hippocampal proteomics analyses and PTM screening showed that MZD did not change the expression and PTM of proteins. The brain pathology, learning, memory and the concentrations of indicators known to be related to learning and memory were not changed by MZD. Our study confirmed that marginal zinc deficiency (10 mg kg-1, 1/3 RNI) had no effect on the learning and memory abilities of rats after birth.
Collapse
Affiliation(s)
- Yongzhi Sun
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Jiali Zhao
- Yiwu Maternity and Children Hospital, Yiwu, Zhejiang, China
| | - Xinyu Song
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Zhaohui Sun
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Rui Zhang
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Jiayi Zhong
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Xiaocai Huang
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Yingran Dong
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Qingli Yu
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Feng Dong
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Zixiang Li
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Lina Fan
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Maoqing Wang
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin 150081, China.
| | - Chenghai Peng
- Department of Cardiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150081, China.
| | - Fan Wang
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, China.
| |
Collapse
|
19
|
Andreassen SN, Toft-Bertelsen TL, Wardman JH, Villadsen R, MacAulay N. Transcriptional profiling of transport mechanisms and regulatory pathways in rat choroid plexus. Fluids Barriers CNS 2022; 19:44. [PMID: 35659263 PMCID: PMC9166438 DOI: 10.1186/s12987-022-00335-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dysregulation of brain fluid homeostasis associates with brain pathologies in which fluid accumulation leads to elevated intracranial pressure. Surgical intervention remains standard care, since specific and efficient pharmacological treatment options are limited for pathologies with disturbed brain fluid homeostasis. Such lack of therapeutic targets originates, in part, from the incomplete map of the molecular mechanisms underlying cerebrospinal fluid (CSF) secretion by the choroid plexus. METHODS The transcriptomic profile of rat choroid plexus was generated by RNA Sequencing (RNAseq) of whole tissue and epithelial cells captured by fluorescence-activated cell sorting (FACS), and compared to proximal tubules. The bioinformatic analysis comprised mapping to reference genome followed by filtering for type, location, and association with alias and protein function. The transporters and associated regulatory modules were arranged in discovery tables according to their transcriptional abundance and tied together in association network analysis. RESULTS The transcriptomic profile of choroid plexus displays high similarity between sex and species (human, rat, and mouse) and lesser similarity to another high-capacity fluid-transporting epithelium, the proximal tubules. The discovery tables provide lists of transport mechanisms that could participate in CSF secretion and suggest regulatory candidates. CONCLUSIONS With quantification of the transport protein transcript abundance in choroid plexus and their potentially linked regulatory modules, we envision a molecular tool to devise rational hypotheses regarding future delineation of choroidal transport proteins involved in CSF secretion and their regulation. Our vision is to obtain future pharmaceutical targets towards modulation of CSF production in pathologies involving disturbed brain water dynamics.
Collapse
Affiliation(s)
- Søren N Andreassen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Jonathan H Wardman
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
20
|
Monchanin C, Gabriela de Brito Sanchez M, Lecouvreur L, Boidard O, Méry G, Silvestre J, Le Roux G, Baqué D, Elger A, Barron AB, Lihoreau M, Devaud JM. Honey bees cannot sense harmful concentrations of metal pollutants in food. CHEMOSPHERE 2022; 297:134089. [PMID: 35240159 DOI: 10.1016/j.chemosphere.2022.134089] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/02/2022] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
Whether animals can actively avoid food contaminated with harmful compounds through taste is key to assess their ecotoxicological risks. Here, we investigated the ability of honey bees to perceive and avoid food resources contaminated with common metal pollutants known to impair behaviour at low concentrations. In laboratory assays, bees did not discriminate food contaminated with arsenic, lead or zinc and ingested it readily, up to estimated doses of 929.1 μg g-1 As, 6.45 mg g-1 Pb and 72.46 mg g-1 Zn. A decrease of intake and appetitive responses indicating metal detection was only observed at the highest concentrations of lead (3.6 mM) and zinc (122.3 mM) through contact with the antennae and the proboscis. Electrophysiological analyses confirmed that only high concentrations of the three metals in a sucrose solution induced a consistently reduced neural response to sucrose in antennal taste receptors (As: >0.1 μM, Pb: >1 mM; Zn: >100 mM). Overall, cellular and behavioural responses did not provide evidence for specific mechanisms that would support selective detection of toxic metals (arsenic, lead), as compared to zinc, which has important biological functions. Our results thus show that honey bees can avoid metal pollutants in their food only at high concentrations unlikely to be encountered in the environment. By contrast, they appear to be unable to detect low, yet harmful, concentrations found in flowers. Metal pollution at trace levels is therefore a major threat for pollinators.
Collapse
Affiliation(s)
- Coline Monchanin
- Centre de Recherches sur La Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, France; Department of Biological Sciences, Macquarie University, NSW, Australia
| | - Maria Gabriela de Brito Sanchez
- Centre de Recherches sur La Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, France
| | - Loreleï Lecouvreur
- Centre de Recherches sur La Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, France
| | - Océane Boidard
- Centre de Recherches sur La Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, France
| | - Grégoire Méry
- Centre de Recherches sur La Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, France
| | - Jérôme Silvestre
- Laboratoire Ecologie Fonctionnelle et Environnement, Université de Toulouse, CNRS, Toulouse, France
| | - Gaël Le Roux
- Laboratoire Ecologie Fonctionnelle et Environnement, Université de Toulouse, CNRS, Toulouse, France
| | - David Baqué
- Laboratoire Ecologie Fonctionnelle et Environnement, Université de Toulouse, CNRS, Toulouse, France
| | - Arnaud Elger
- Laboratoire Ecologie Fonctionnelle et Environnement, Université de Toulouse, CNRS, Toulouse, France
| | - Andrew B Barron
- Department of Biological Sciences, Macquarie University, NSW, Australia
| | - Mathieu Lihoreau
- Centre de Recherches sur La Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, France
| | - Jean-Marc Devaud
- Centre de Recherches sur La Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, France.
| |
Collapse
|
21
|
|
22
|
S100B dysregulation during brain development affects synaptic SHANK protein networks via alteration of zinc homeostasis. Transl Psychiatry 2021; 11:562. [PMID: 34741005 PMCID: PMC8571423 DOI: 10.1038/s41398-021-01694-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 11/08/2022] Open
Abstract
Autism Spectrum Disorders (ASD) are caused by a combination of genetic predisposition and nongenetic factors. Among the nongenetic factors, maternal immune system activation and zinc deficiency have been proposed. Intriguingly, as a genetic factor, copy-number variations in S100B, a pro-inflammatory damage-associated molecular pattern (DAMP), have been associated with ASD, and increased serum S100B has been found in ASD. Interestingly, it has been shown that increased S100B levels affect zinc homeostasis in vitro. Thus, here, we investigated the influence of increased S100B levels in vitro and in vivo during pregnancy in mice regarding zinc availability, the zinc-sensitive SHANK protein networks associated with ASD, and behavioral outcomes. We observed that S100B affects the synaptic SHANK2 and SHANK3 levels in a zinc-dependent manner, especially early in neuronal development. Animals exposed to high S100B levels in utero similarly show reduced levels of free zinc and SHANK2 in the brain. On the behavioral level, these mice display hyperactivity, increased stereotypic and abnormal social behaviors, and cognitive impairment. Pro-inflammatory factors and zinc-signaling alterations converge on the synaptic level revealing a common pathomechanism that may mechanistically explain a large share of ASD cases.
Collapse
|
23
|
Hansen KB, Wollmuth LP, Bowie D, Furukawa H, Menniti FS, Sobolevsky AI, Swanson GT, Swanger SA, Greger IH, Nakagawa T, McBain CJ, Jayaraman V, Low CM, Dell'Acqua ML, Diamond JS, Camp CR, Perszyk RE, Yuan H, Traynelis SF. Structure, Function, and Pharmacology of Glutamate Receptor Ion Channels. Pharmacol Rev 2021; 73:298-487. [PMID: 34753794 PMCID: PMC8626789 DOI: 10.1124/pharmrev.120.000131] [Citation(s) in RCA: 267] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Many physiologic effects of l-glutamate, the major excitatory neurotransmitter in the mammalian central nervous system, are mediated via signaling by ionotropic glutamate receptors (iGluRs). These ligand-gated ion channels are critical to brain function and are centrally implicated in numerous psychiatric and neurologic disorders. There are different classes of iGluRs with a variety of receptor subtypes in each class that play distinct roles in neuronal functions. The diversity in iGluR subtypes, with their unique functional properties and physiologic roles, has motivated a large number of studies. Our understanding of receptor subtypes has advanced considerably since the first iGluR subunit gene was cloned in 1989, and the research focus has expanded to encompass facets of biology that have been recently discovered and to exploit experimental paradigms made possible by technological advances. Here, we review insights from more than 3 decades of iGluR studies with an emphasis on the progress that has occurred in the past decade. We cover structure, function, pharmacology, roles in neurophysiology, and therapeutic implications for all classes of receptors assembled from the subunits encoded by the 18 ionotropic glutamate receptor genes. SIGNIFICANCE STATEMENT: Glutamate receptors play important roles in virtually all aspects of brain function and are either involved in mediating some clinical features of neurological disease or represent a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of this class of receptors will advance our understanding of many aspects of brain function at molecular, cellular, and system levels and provide new opportunities to treat patients.
Collapse
Affiliation(s)
- Kasper B Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Lonnie P Wollmuth
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Derek Bowie
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hiro Furukawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Frank S Menniti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Alexander I Sobolevsky
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Geoffrey T Swanson
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Sharon A Swanger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Ingo H Greger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Terunaga Nakagawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chris J McBain
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Vasanthi Jayaraman
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chian-Ming Low
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Mark L Dell'Acqua
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Jeffrey S Diamond
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chad R Camp
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Riley E Perszyk
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hongjie Yuan
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Stephen F Traynelis
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| |
Collapse
|
24
|
Kumar M, Swarnim S, Khanam S. Zinc Supplementation for Prevention of Febrile Seizures Recurrences in Children: A Systematic Review and Meta-Analysis. Indian Pediatr 2021. [DOI: 10.1007/s13312-021-2309-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
25
|
Peralta FA, Huidobro-Toro JP, Mera-Adasme R. Hybrid QM/MM Simulations Confirm Zn(II) Coordination Sphere That Includes Four Cysteines from the P2 × 4R Head Domain. Int J Mol Sci 2021; 22:ijms22147288. [PMID: 34298909 PMCID: PMC8303255 DOI: 10.3390/ijms22147288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/21/2022] Open
Abstract
To ascertain the role of Zn(II) as an allosteric modulator on P2X4R, QM/MM molecular dynamic simulations were performed on the WT and two P2X4R mutants suggested by previous electrophysiological data to affect Zn(II) binding. The Gibbs free energy for the reduction of the putative P2X4R Zn(II) binding site by glutathione was estimated at −22 kcal/mol. Simulations of the WT P2X4R head domain revealed a flexible coordination sphere dominated by an octahedral geometry encompassing C126, N127, C132, C149, C159 and a water molecule. The C132A mutation disrupted the metal binding site, leading to a coordination sphere with a majority of water ligands, and a displacement of the metal ion towards the solvent. The C132A/C159A mutant exhibited a tendency towards WT-like stability by incorporating the R148 backbone to the coordination sphere. Thus, the computational findings agree with previous experimental data showing Zn(II) modulation for the WT and C132A/C159A variants, but not for the C132A mutant. The results provide molecular insights into the nature of the Zn(II) modulation in P2X4R, and the effect of the C132A and C132A/C159A mutations, accounting for an elusive modulation mechanism possibly occurring in other extracellular or membrane protein.
Collapse
Affiliation(s)
| | - J. Pablo Huidobro-Toro
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile (USACH), Santiago 9170124, Chile
- Centro Para el Desarrollo de Nanociencia y Nanotecnología, (CEDENNA), Universidad de Santiago de Chile (USACH), Santiago 9170124, Chile
- Correspondence: (J.P.H.-T.); (R.M.-A.)
| | - Raúl Mera-Adasme
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile (USACH), Santiago 9170124, Chile
- Correspondence: (J.P.H.-T.); (R.M.-A.)
| |
Collapse
|
26
|
Jafari F, Mohammadi H, Amani R. The effect of zinc supplementation on brain derived neurotrophic factor: A meta-analysis. J Trace Elem Med Biol 2021; 66:126753. [PMID: 33831797 DOI: 10.1016/j.jtemb.2021.126753] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/31/2021] [Accepted: 03/29/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Zinc in one of the most abundant trace minerals in human body which is involved in numerous biological pathways and has variety of roles in the nervous system. It has been assumed that zinc exerts its role in nervous system through increasing brain derived neurotrophic factor (BDNF) concentrations. OBJECTIVES Present meta-analysis was aimed to review the effect of zinc supplementation on serum concentrations of BDNF. METHODS AND MATERIALS Four electronic databases (Pubmed, Scopus, Web of Science, Embase) were searched for identifying studies that examined BDNF levels prior and after zinc supplementation up to May 2020. According to the Cochrane guideline, a meta-analysis was performed to pool the effect size estimate (Hedges' test) of serum BDNF across studies. Risk of publication bias was assessed using a funnel plot and Egger's test. RESULTS Five studies were eligible and 238 participants were included. These studies enrolled subjects with premenstrual syndrome, diabetic retinopathy, major depression disorder, overweight/obese and obese with mild to moderate depressive disorders. Zinc supplementation failed to increase blood BDNF concentrations with effect size of 0.30 (95 % CI: -0.08, 0.67, P = 0.119). Funnel plot did not suggest publication bias. CONCLUSION Zinc supplementation may not significantly increase BDNF levels. However, the small number of included articles and significant heterogeneity between them can increase the risk of a false negative result; therefore, the results should be interpreted with caution.
Collapse
Affiliation(s)
- Fatemeh Jafari
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Hamed Mohammadi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Reza Amani
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
27
|
Kuznetsov AV, Vainer VI, Volkova YM, Kartashov LE. Motility disorders and disintegration into separate cells of Trichoplax sp. H2 in the presence of Zn 2+ ions and L-cysteine molecules: A systems approach. Biosystems 2021; 206:104444. [PMID: 34023485 DOI: 10.1016/j.biosystems.2021.104444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/08/2021] [Accepted: 05/09/2021] [Indexed: 01/01/2023]
Abstract
Placozoa remain an ancient multicellular system with a dynamic body structure where calcium ions carry out a primary role in maintaining the integrity of the entire animal. Zinc ions can compete with calcium ions adsorption. We studied the effect of zinc ions and l-cysteine molecules on the interaction of Trichoplax sp. H2 cells. The regularity of formless motion was diminished in the presence of 20-25 μM of Zn2+ ions leading to the formation of branching animal forms. Locomotor ciliated cells moved chaotically and independently of each other leaving the Trichoplax body and opening a network of fiber cells. Application of 100 μM cysteine resulted in dissociation of the plate into separate cells. The combined chemical treatment shifted the effect in a random sample of animals toward disintegration, i.e. initially leading to disorder of collective cell movement and then to total body fragmentation. Two dissociation patterns of Trichoplax plate as "expanding ring" and "bicycle wheel" were revealed. Analysis of the interaction of Ca2+ and Zn2+ ions with cadherin showed that more than half (54%) of the amino acid residues with which Ca2+ and Zn2+ ions bind are common. The contact interaction of cells covered by the cadherin molecules is important for the coordinated movements of Trichoplax organism, while zinc ions are capable to break junctions between the cells. The involvement of other players, for example, l-cysteine in the regulation of Ca2+-dependent adhesion may be critical leading to the typical dissociation of Trichoplax body like in a calcium-free environment. A hypothesis about the essential role of calcium ions in the emergence of Metazoa ancestor is proposed.
Collapse
Affiliation(s)
- A V Kuznetsov
- A.O. Kovalevsky Institute of Biology of the Southern Seas RAS, Leninsky Avenue 38, Moscow, 119991, Russia.
| | - V I Vainer
- A.O. Kovalevsky Institute of Biology of the Southern Seas RAS, Leninsky Avenue 38, Moscow, 119991, Russia
| | - Yu M Volkova
- A.O. Kovalevsky Institute of Biology of the Southern Seas RAS, Leninsky Avenue 38, Moscow, 119991, Russia
| | - L E Kartashov
- A.O. Kovalevsky Institute of Biology of the Southern Seas RAS, Leninsky Avenue 38, Moscow, 119991, Russia
| |
Collapse
|
28
|
Supasai S, Adamo AM, Mathieu P, Marino RC, Hellmers AC, Cremonini E, Oteiza PI. Gestational zinc deficiency impairs brain astrogliogenesis in rats through multistep alterations of the JAK/STAT3 signaling pathway. Redox Biol 2021; 44:102017. [PMID: 34049221 PMCID: PMC8167189 DOI: 10.1016/j.redox.2021.102017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 10/27/2022] Open
Abstract
We previously showed that zinc (Zn) deficiency affects the STAT3 signaling pathway in part through redox-regulated mechanisms. Given that STAT3 is central to the process of astrogliogenesis, this study investigated the consequences of maternal marginal Zn deficiency on the developmental timing and key mechanisms of STAT3 activation, and its consequences on astrogliogenesis in the offspring. This work characterized the temporal profile of cortical STAT3 activation from the mid embryonic stage up to young adulthood in the offspring from dams fed a marginal Zn deficient diet (MZD) throughout gestation and until postnatal day (P) 2. All rats were fed a Zn sufficient diet (control) from P2 until P56. Maternal zinc deficiency disrupted cortical STAT3 activation at E19 and P2. This was accompanied by altered activation of JAK2 kinase due to changes in PTP1B phosphatase activity. The underlying mechanisms mediating the adverse impact of a decreased Zn availability on STAT3 activation in the offspring brain include: (i) impaired PTP1B degradation via the ubiquitin/proteasome pathway; (ii) tubulin oxidation, associated decreased interactions with STAT3 and consequent impaired nuclear translocation; and (iii) decreased nuclear STAT3 acetylation. Zn deficiency-associated decreased STAT3 activation adversely impacted astrogliogenesis, leading to a lower astrocyte number in the early postnatal and adult brain cortex. Thus, a decreased availability of Zn during early development can have a major and irreversible adverse effect on astrogliogenesis, in part via multistep alterations in the STAT3 pathway.
Collapse
Affiliation(s)
- Suangsuda Supasai
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Ana M Adamo
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Patricia Mathieu
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Regina C Marino
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Adelaide C Hellmers
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Eleonora Cremonini
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Patricia I Oteiza
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA, 95616, USA; Department of Environmental Toxicology, University of California, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
29
|
Arvas B, Ucar B, Acar T, Arvas MB, Sahin Y, Aydogan F, Yolacan C. A new coumarin based Schiff base fluorescence probe for zinc ion. Tetrahedron 2021. [DOI: 10.1016/j.tet.2021.132127] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
30
|
Bou Khalil R, Yazbek JC. Potential importance of supplementation with zinc for autism spectrum disorder. Encephale 2021; 47:514-517. [PMID: 33863509 DOI: 10.1016/j.encep.2020.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 11/29/2022]
Abstract
Zinc is an essential micronutrient for cellular proliferation and subsequent body and brain development. Zinc deficiency is becoming a major public health issue equally in under-developed and developed countries. The lack of sufficient zinc, whether related to environmental or internal factors, is an important environmental stressor that is eligible to become elucidated as a contributing factor for the pathogenesis of autism spectrum disorder (ASD). The aim of this manuscript is to briefly overview available data regarding the relationship of zinc deficiency with the development of ASD and to relate these data with currently known pathogenetic mechanisms of this disorder namely brain growth disturbances and neuropeptides secretion. Zinc deficiency impacts brain connectivity and growth and alters adequate neurotransmission. In addition, zinc deficiency may indirectly act on the brain by disturbing the immune system and by altering the normal gut-brain connection. Zinc seems to be important for the social effect of neuropeptides. Zinc supplementation in pregnant women and newborn children with the aim of preventing ASD needs further consideration.
Collapse
Affiliation(s)
- R Bou Khalil
- Hôtel Dieu de France- Hôtel-Dieu de France, Saint Joseph University, A. Naccache boulevard, Achrafieh, 166830 Beirut, Lebanon.
| | - J-C Yazbek
- Hôtel Dieu de France- Hôtel-Dieu de France, Saint Joseph University, A. Naccache boulevard, Achrafieh, 166830 Beirut, Lebanon
| |
Collapse
|
31
|
Blampied M, Bell C, Gilbert C, Rucklidge JJ. Broad spectrum micronutrient formulas for the treatment of symptoms of depression, stress, and/or anxiety: a systematic review. Expert Rev Neurother 2021; 20:351-371. [PMID: 32178540 DOI: 10.1080/14737175.2020.1740595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Vitamin and mineral nutritional supplements are becoming increasingly popular as alternative treatments for anxiety and depression, as issues such as side effects from medication, failure to respond to psychotherapy and workforce limitations pose barriers for successful treatment.Areas covered: This review covered double-blind, randomized controlled trials (DBRCTs) testing formulas including at least four vitamins and/or minerals used for the treatment of symptoms of anxiety, stress, or depression in adults not currently taking medication for psychiatric difficulties.Expert opinion: The majority of the 23 trials reviewed were conducted on people without psychological difficulties, limiting the generalizability of the results in people with diagnosed mood and anxiety difficulties. Sixteen studies demonstrated positive effects for symptoms of anxiety, depression, or stress. Micronutrient supplementation in healthy nonclinical adults has limited benefits for mood and anxiety symptoms, although may convey some subtle general improvements. The evidence for adults with physical or mental ill health is more positive although limited by small samples and variability in nutrients studied. Broad-spectrum nutrient products may be more effective than a selected few. While an effect of micronutrients cannot be dismissed, the variability of the studies makes it extremely challenging to identify specific treatment benefits.
Collapse
Affiliation(s)
- Meredith Blampied
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - Caroline Bell
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Claire Gilbert
- Canterbury District Health Board, Christchurch, New Zealand
| | - Julia J Rucklidge
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| |
Collapse
|
32
|
Barber-Zucker S, Moran A, Zarivach R. Metal transport mechanism of the cation diffusion facilitator (CDF) protein family - a structural perspective on human CDF (ZnT)-related diseases. RSC Chem Biol 2021; 2:486-498. [PMID: 34458794 PMCID: PMC8341793 DOI: 10.1039/d0cb00181c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/26/2020] [Indexed: 11/21/2022] Open
Abstract
Divalent d-block metal cations (DDMCs) participate in many cellular functions; however, their accumulation in cells can be cytotoxic. The cation diffusion facilitator (CDF) family is a ubiquitous family of transmembrane DDMC exporters that ensures their homeostasis. Severe diseases, such as type II diabetes, Parkinson's and Alzheimer's disease, were linked to dysfunctional human CDF proteins, ZnT-1-10 (SLC30A1-10). Each member of the CDF family reduces the cytosolic concentration of a specific DDMC by transporting it from the cytoplasm to the extracellular environment or into intracellular compartments. This process is usually achieved by utilizing the proton motive force. In addition to their activity as DDMC transporters, CDFs also have other cellular functions such as the regulation of ion channels and enzymatic activity. The combination of structural and biophysical studies of different bacterial and eukaryotic CDF proteins led to significant progress in the understanding of the mutual interaction among CDFs and DDMCs, their involvement in ion binding and selectivity, conformational changes and the consequent transporting mechanisms. Here, we review these studies, provide our mechanistic interpretation of CDF proteins based on the current literature and relate the above to known human CDF-related diseases. Our analysis provides a common structure-function relationship to this important protein family and closes the gap between eukaryote and prokaryote CDFs.
Collapse
Affiliation(s)
- Shiran Barber-Zucker
- Department of Life Sciences, the National Institute for Biotechnology in the Negev and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev P.O.B. 653 Beer Sheva 8410501 Israel +972-8-6472970 +972-8-6472970 +972-8-6428447 +972-8-6461999
| | - Arie Moran
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev P.O.B. 653 Beer Sheva 8410501 Israel
| | - Raz Zarivach
- Department of Life Sciences, the National Institute for Biotechnology in the Negev and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev P.O.B. 653 Beer Sheva 8410501 Israel +972-8-6472970 +972-8-6472970 +972-8-6428447 +972-8-6461999
| |
Collapse
|
33
|
Saha U, Mabhai S, Das B, Kumar GS, Brandão P, Dolai M. Combined theoretical and experimental investigation of a DNA interactive poly-hydroxyl enamine tautomer exhibiting “turn on” sensing for Zn 2+ in pseudo-aqueous medium. NEW J CHEM 2021. [DOI: 10.1039/d1nj03510j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Crystallographically established (solid state structure at 150 K temperature) synthesized enamine ligand (H4L) showed interconvertible equilibrium (ΔE = 7.37 kcal) of its tautomers, displayed zinc sensing and also found to exhibit DNA binding activity at the minor groove of double-stranded (ds) DNA.
Collapse
Affiliation(s)
- Urmila Saha
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, W.B., India
| | - Subhabrata Mabhai
- Department of Chemistry, Mahishadal Raj College, Purba Medinipur 721628, W.B., India
| | - Bhriguram Das
- Department of Chemistry, Vidyasagar University, Paschim Medinipur 721102, W. B., India
| | - Gopinatha Suresh Kumar
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, W.B., India
| | - Paula Brandão
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Malay Dolai
- Department of Chemistry, Prabhat Kumar College, Purba Medinipur 721404, W.B., India
| |
Collapse
|
34
|
Kumar N, Roopa, Bhalla V, Kumar M. Beyond zinc coordination: Bioimaging applications of Zn(II)-complexes. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213550] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
35
|
Jorratt P, Hoschl C, Ovsepian SV. Endogenous antagonists of N-methyl-d-aspartate receptor in schizophrenia. Alzheimers Dement 2020; 17:888-905. [PMID: 33336545 DOI: 10.1002/alz.12244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/24/2020] [Indexed: 12/28/2022]
Abstract
Schizophrenia is a chronic neuropsychiatric brain disorder that has devastating personal impact and rising healthcare costs. Dysregulation of glutamatergic neurotransmission has been implicated in the pathobiology of the disease, attributed largely to the hypofunction of the N-methyl-d-aspartate (NMDA) receptor. Currently, there is a major gap in mechanistic analysis as to how endogenous modulators of the NMDA receptors contribute to the onset and progression of the disease. We present a systematic review of the neurobiology and the role of endogenous NMDA receptor antagonists in animal models of schizophrenia, and in patients. We discuss their neurochemical origin, release from neurons and glia with action mechanisms, and functional effects, which might contribute toward the impairment of neuronal processes underlying this complex pathological state. We consider clinical evidence suggesting dysregulations of endogenous NMDA receptor in schizophrenia, and highlight the pressing need in future studies and emerging directions, to restore the NMDA receptor functions for therapeutic benefits.
Collapse
Affiliation(s)
- Pascal Jorratt
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague 10, Czech Republic
| | - Cyril Hoschl
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague 10, Czech Republic
| | - Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague 10, Czech Republic
| |
Collapse
|
36
|
Baj J, Forma A, Sitarz E, Karakuła K, Flieger W, Sitarz M, Grochowski C, Maciejewski R, Karakula-Juchnowicz H. Beyond the Mind-Serum Trace Element Levels in Schizophrenic Patients: A Systematic Review. Int J Mol Sci 2020; 21:ijms21249566. [PMID: 33334078 PMCID: PMC7765526 DOI: 10.3390/ijms21249566] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/06/2020] [Accepted: 12/12/2020] [Indexed: 12/19/2022] Open
Abstract
The alterations in serum trace element levels are common phenomena observed in patients with different psychiatric conditions such as schizophrenia, autism spectrum disorder, or major depressive disorder. The fluctuations in the trace element concentrations might act as potential diagnostic and prognostic biomarkers of many psychiatric and neurological disorders. This paper aimed to assess the alterations in serum trace element concentrations in patients with a diagnosed schizophrenia. The authors made a systematic review, extracting papers from the PubMed, Web of Science, and Scopus databases according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Among 5009 articles identified through database searching, 59 of them were assessed for eligibility. Ultimately, 33 articles were included in the qualitative synthesis. This review includes the analysis of serum levels of the following trace elements: iron, nickel, molybdenum, phosphorus, lead, chromium, antimony, uranium, magnesium, aluminum, zinc, copper, selenium, calcium, and manganese. Currently, there is no consistency regarding serum trace element levels in schizophrenic patients. Thus, it cannot be considered as a reliable prognostic or diagnostic marker of schizophrenia. However, it can be assumed that altered concentrations of those elements are crucial regarding the onset and exaggeration of either psychotic or negative symptoms or cognitive dysfunctions.
Collapse
Affiliation(s)
- Jacek Baj
- Department of Human Anatomy, Medical University of Lublin, 20-400 Lublin, Poland;
- Correspondence:
| | - Alicja Forma
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Elżbieta Sitarz
- Chair and 1st Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland; (E.S.); (K.K.); (H.K.-J.)
| | - Kaja Karakuła
- Chair and 1st Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland; (E.S.); (K.K.); (H.K.-J.)
| | - Wojciech Flieger
- Faculty of Medicine, Medical University of Lublin, Aleje Racławickie 1, 20-059 Lublin, Poland;
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Cezary Grochowski
- Laboratory of Virtual Man, Chair of Anatomy, Medical University of Lublin, 20-400 Lublin, Poland;
| | - Ryszard Maciejewski
- Department of Human Anatomy, Medical University of Lublin, 20-400 Lublin, Poland;
| | - Hanna Karakula-Juchnowicz
- Chair and 1st Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland; (E.S.); (K.K.); (H.K.-J.)
- Department of Clinical Neuropsychiatry, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland
| |
Collapse
|
37
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
38
|
Jia W, Song Y, Yang L, Kong J, Boczek T, He Z, Wang Y, Zhang X, Hu H, Shao D, Tang H, Xia L, Xu X, Guo F. The changes of serum zinc, copper, and selenium levels in epileptic patients: a systematic review and meta-analysis. Expert Rev Clin Pharmacol 2020; 13:1047-1058. [PMID: 32856976 DOI: 10.1080/17512433.2020.1816821] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION It is widely accepted that trace elements have been implicated in various metabolic processes. Valproic acid (VPA) is a remarkably safe and effective antiepileptic drug. There is no consensus option regarding the fluctuations in serum zinc (Zn), copper (Cu), and selenium (Se) in epileptic patients treated with VPA. We applied a meta-analysis to systematically assess the effects of VPA on serum ions in these patients. AREAS COVERED In this study, we performed a meta-analysis of the changes in serum Zn, Cu, and Se levels in human samples of healthy controls, epileptic patients, and patients treated with VPA. Twenty-two published analyzable studies were selected by searching the databases of PubMed, China National Knowledge Infrastructure (CNKI), Google Scholar, Web of Science, EMBASE, WAN FANG and Vip. EXPERT OPINION Serum Se levels in epileptic patients were decreased compared to healthy controls. Serum Zn levels in patients with VPA treatment were significantly lower than those in epileptic patients. The results of this meta-analysis are instructive for the intake of trace elements such as Zn, Cu, and Se in the diet balance of patients with epilepsy treated with VPA. Meanwhile, this study provides a theoretical basis for the combined use of other drugs that affect the intake and absorption of trace elements and VPA.
Collapse
Affiliation(s)
- Wanying Jia
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University , Shenyang, China
| | - Yang Song
- School of Humanities and Social Sciences, China Medical University , Shenyang, China
| | - Lei Yang
- Tianjin Customs, Technical Center for Safety of Industrial Products , Tianjin, China
| | - Jingjing Kong
- Department of Gerontology, The First Affiliated Hospital of Dalian Medical University , Dalian, China
| | - Tomasz Boczek
- Department of Ophthalmology, Stanford University School of Medicine , Palo Alto, CA, USA
| | - Zhenwei He
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University , Shenyang, China
| | - Yuting Wang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University , Shenyang, China
| | - Xiaohong Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University , Shenyang, China
| | - Huiyuan Hu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University , Shenyang, China
| | - Dongxue Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University , Shenyang, China
| | - Hong Tang
- School of Public Health, China Medical University , Shenyang, China
| | - Liguang Xia
- Department of Pediatric Surgery, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University , Wenzhou, China
| | - Xiaoxue Xu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University , Shenyang, China
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University , Shenyang, China
| |
Collapse
|
39
|
New Insights of the Zn(II)-Induced P2 × 4R Positive Allosteric Modulation: Role of Head Receptor Domain SS2/SS3, E160 and D170. Int J Mol Sci 2020; 21:ijms21186940. [PMID: 32971737 PMCID: PMC7555825 DOI: 10.3390/ijms21186940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 01/18/2023] Open
Abstract
P2 × 4R is allosterically modulated by Zn(II), and despite the efforts to understand the mechanism, there is not a consensus proposal; C132 is a critical amino acid for the Zn(II) modulation, and this residue is located in the receptor head domain, forming disulfide SS3. To ascertain the role of the SS2/SS3 microenvironment on the rP2 × 4R Zn(II)-induced allosteric modulation, we investigated the contribution of each individual SS2/SS3 cysteine plus carboxylic acid residues E118, E160, and D170, located in the immediate vicinity of the SS2/SS3 disulfide bonds. To this aim, we combined electrophysiological recordings with protein chemical alkylation using thiol reagents such as N-ethylmaleimide or iodoacetamide, and a mutation of key amino acid residues together with P2 × 4 receptor bioinformatics. P2 × 4R alkylation in the presence of the metal obliterated the allosteric modulation, a finding supported by the site-directed mutagenesis of C132 and C149 by a corresponding alanine. In addition, while E118Q was sensitive to Zn(II) modulation, the wild type receptor, mutants E160Q and D170N, were not, suggesting that these acid residues participate in the modulatory mechanism. Poisson–Boltzmann analysis indicated that the E160Q and D170N mutants showed a shift towards more positive electrostatic potential in the SS2/SS3 microenvironment. Present results highlight the role of C132 and C149 as putative Zn(II) ligands; in addition, we infer that acid residues E160 and D170 play a role attracting Zn(II) to the head receptor domain.
Collapse
|
40
|
Pan CY, Lin FY, Kao LS, Huang CC, Liu PS. Zinc oxide nanoparticles modulate the gene expression of ZnT1 and ZIP8 to manipulate zinc homeostasis and stress-induced cytotoxicity in human neuroblastoma SH-SY5Y cells. PLoS One 2020; 15:e0232729. [PMID: 32915786 PMCID: PMC7485861 DOI: 10.1371/journal.pone.0232729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/27/2020] [Indexed: 11/19/2022] Open
Abstract
Zinc ions (Zn2+) are important messenger molecules involved in various physiological functions. To maintain the homeostasis of cytosolic Zn2+ concentration ([Zn2+]c), Zrt/Irt-related proteins (ZIPs) and Zn2+ transporters (ZnTs) are the two families of proteins responsible for decreasing and increasing the [Zn2+]c, respectively, by fluxing Zn2+ across the membranes of the cell and intracellular compartments in opposite directions. Most studies focus on the cytotoxicity incurred by a high concentration of [Zn2+]c and less investigate the [Zn2+]c at physiological levels. Zinc oxide-nanoparticle (ZnO-NP) is blood brain barrier-permeable and elevates the [Zn2+]c to different levels according to the concentrations of ZnO-NP applied. In this study, we mildly elevated the [Zn2+]c by ZnO-NP at concentrations below 1 μg/ml, which had little cytotoxicity, in cultured human neuroblastoma SH-SY5Y cells and characterized the importance of Zn2+ transporters in 6-hydroxy dopamine (6-OHDA)-induced cell death. The results show that ZnO-NP at low concentrations elevated the [Zn2+]c transiently in 6 hr, then declined gradually to a basal level in 24 hr. Knocking down the expression levels of ZnT1 (located mostly at the plasma membrane) and ZIP8 (present in endosomes and lysosomes) increased and decreased the ZnO-NP-induced elevation of [Zn2+]c, respectively. ZnO-NP treatment reduced the basal levels of reactive oxygen species and Bax/Bcl-2 mRNA ratios; in addition, ZnO-NP decreased the 6-OHDA-induced ROS production, p53 expression, and cell death. These results show that ZnO-NP-induced mild elevation in [Zn2+]c activates beneficial effects in reducing the 6-OHDA-induced cytotoxic effects. Therefore, brain-delivery of ZnO-NP can be regarded as a potential therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Chien-Yuan Pan
- Department of Life Science and Institute of Zoology, National Taiwan University, Taipei, Taiwan
| | - Fang-Yu Lin
- Department of Microbiology, Soochow University, Taipei, Taiwan
| | - Lung-Sen Kao
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Chang Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Shan Liu
- Department of Microbiology, Soochow University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
41
|
Magnesium, Calcium, Potassium, Sodium, Phosphorus, Selenium, Zinc, and Chromium Levels in Alcohol Use Disorder: A Review. J Clin Med 2020; 9:jcm9061901. [PMID: 32570709 PMCID: PMC7357092 DOI: 10.3390/jcm9061901] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
Macronutrients and trace elements are important components of living tissues that have different metabolic properties and functions. Trace elements participate in the regulation of immunity through humoral and cellular mechanisms, nerve conduction, muscle spasms, membrane potential regulation as well as mitochondrial activity and enzymatic reactions. Excessive alcohol consumption disrupts the concentrations of crucial trace elements, also increasing the risk of enhanced oxidative stress and alcohol-related liver diseases. In this review, we present the status of selected macroelements and trace elements in the serum and plasma of people chronically consuming alcohol. Such knowledge helps to understand the mechanisms of chronic alcohol-use disorder and to progress and prevent withdrawal effects, also improving treatment strategies.
Collapse
|
42
|
Wiqas A, LeSauter J, Taub A, Austin RN, Silver R. Elevated zinc transporter ZnT3 in the dentate gyrus of mast cell-deficient mice. Eur J Neurosci 2019; 51:1504-1513. [PMID: 31502721 DOI: 10.1111/ejn.14575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/01/2019] [Accepted: 07/25/2019] [Indexed: 11/27/2022]
Abstract
Zinc is important in neurogenesis, but excessive levels can cause apoptosis and other pathologies leading to cognitive impairments. Mast cells are present in many brain regions including the hippocampus, an area rich in vesicular zinc. Mast cells contain zinc-rich granules and a well-developed mechanism for uptake of zinc ions; both features point to the potential for a role in zinc homeostasis. Prior work using the Timm stain supported this hypothesis, as increased labile zinc was detected in the hippocampus of mast cell-deficient mice compared to wild-type mice while no differences in total zinc were found between the two genotypes in the whole brain or other tissues. The current report further examines differences in zinc homeostasis between wild-type and mast cell-deficient mice by exploring the zinc transporter ZnT3, which transports labile zinc into synaptic vesicles. The first study used immunocytochemistry to localize ZnT3 within the mossy fibre layer of the hippocampus to determine whether there was differential expression of ZnT3 in wild-type versus mast cell-deficient mice. The second study used inductively coupled plasma mass spectrometry (ICP-MS) to determine total zinc content in the whole dentate gyrus of the two genotypes. The immunocytochemical results indicate that there are higher levels of ZnT3 localized to the mossy fibre layer of the dentate gyrus of mast cell-deficient mice than in wild-type mice. The ICP-MS data reveal no differences in total zinc in dentate gyrus as a whole. The results are consistent with the hypothesis that mast cells participate in zinc homeostasis at the level of synaptic vesicles.
Collapse
Affiliation(s)
- Amen Wiqas
- Department of Biology, Barnard College, Columbia University, New York, New York
| | - Joseph LeSauter
- Department of Neuroscience, Barnard College, Columbia University, New York, New York
| | - Alana Taub
- Department of Psychology, Columbia University, New York, New York
| | | | - Rae Silver
- Department of Neuroscience, Barnard College, Columbia University, New York, New York.,Department of Psychology, Columbia University, New York, New York
| |
Collapse
|
43
|
Othman H, Ammari M, Lassoued A, Sakly M, Abdelmelek H. Zinc improves clomipramine effects on depressive and locomotor behavior and reverses its oxidative stress in rats. Behav Brain Res 2019; 374:112122. [PMID: 31376442 DOI: 10.1016/j.bbr.2019.112122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 10/26/2022]
Abstract
Depression is a common mental disease affecting a lot of people of all ages around the world. Today, improving the therapeutic effects of currently used antidepressants such as clomipramine and, especially when they are administered at high doses is a topic of interest. The study aims to evaluate the eventual role of zinc (30 mg/Kg) in ameliorating clomipramine (75 mg/Kg) effects on behavior and oxidative stress equilibrium following a 6 day treatment in male Wistar rats. Our main findings showed that zinc improved clomipramine antidepressant and locomotor effects. Moreover, zinc reversed the oxidative stress induced by this drug in the liver. Thus, zinc at 30 mg/Kg may constitute an efficient adjuvant for clomipramine used at a high dose (75 mg/Kg) by boosting its efficacy on behavior and alleviating its negative effects on oxidative balance in liver.
Collapse
Affiliation(s)
- Haifa Othman
- University of Carthage, Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, Jarzouna, 7021, Tunisia.
| | - Mohamed Ammari
- University of Carthage, Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, Jarzouna, 7021, Tunisia; University of Tunis El Manar, Higher Institute of Applied Biological Sciences of Tunis, 9, Rue Zouhair Essafi, 1006, Tunis, Tunisia
| | - Amal Lassoued
- University of Carthage, Faculty of Sciences of Bizerte, Environment Biomonitoring Laboratory, Jarzouna, 7021, Tunisia
| | - Mohsen Sakly
- University of Carthage, Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, Jarzouna, 7021, Tunisia
| | - Hafedh Abdelmelek
- University of Carthage, Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, Jarzouna, 7021, Tunisia
| |
Collapse
|
44
|
Petrović S, Guzsvány V, Ranković N, Beljin J, Rončević S, Dalmacija B, Ashrafi AM, Kónya Z, Švancara I, Vytřas K. Trace level voltammetric determination of Zn(II) in selected nutrition related samples by bismuth-oxychloride-multiwalled carbon nanotube composite based electrode. Microchem J 2019. [DOI: 10.1016/j.microc.2018.12.053] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
45
|
Luo Q, Chen Q, Wang W, Desrivières S, Quinlan EB, Jia T, Macare C, Robert GH, Cui J, Guedj M, Palaniyappan L, Kherif F, Banaschewski T, Bokde ALW, Büchel C, Flor H, Frouin V, Garavan H, Gowland P, Heinz A, Ittermann B, Martinot JL, Artiges E, Paillère-Martinot ML, Nees F, Orfanos DP, Poustka L, Fröhner JH, Smolka MN, Walter H, Whelan R, Callicott JH, Mattay VS, Pausova Z, Dartigues JF, Tzourio C, Crivello F, Berman KF, Li F, Paus T, Weinberger DR, Murray RM, Schumann G, Feng J. Association of a Schizophrenia-Risk Nonsynonymous Variant With Putamen Volume in Adolescents: A Voxelwise and Genome-Wide Association Study. JAMA Psychiatry 2019; 76:435-445. [PMID: 30649180 PMCID: PMC6450291 DOI: 10.1001/jamapsychiatry.2018.4126] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 10/16/2018] [Indexed: 02/03/2023]
Abstract
Importance Deviation from normal adolescent brain development precedes manifestations of many major psychiatric symptoms. Such altered developmental trajectories in adolescents may be linked to genetic risk for psychopathology. Objective To identify genetic variants associated with adolescent brain structure and explore psychopathologic relevance of such associations. Design, Setting, and Participants Voxelwise genome-wide association study in a cohort of healthy adolescents aged 14 years and validation of the findings using 4 independent samples across the life span with allele-specific expression analysis of top hits. Group comparison of the identified gene-brain association among patients with schizophrenia, unaffected siblings, and healthy control individuals. This was a population-based, multicenter study combined with a clinical sample that included participants from the IMAGEN cohort, Saguenay Youth Study, Three-City Study, and Lieber Institute for Brain Development sample cohorts and UK biobank who were assessed for both brain imaging and genetic sequencing. Clinical samples included patients with schizophrenia and unaffected siblings of patients from the Lieber Institute for Brain Development study. Data were analyzed between October 2015 and April 2018. Main Outcomes and Measures Gray matter volume was assessed by neuroimaging and genetic variants were genotyped by Illumina BeadChip. Results The discovery sample included 1721 adolescents (873 girls [50.7%]), with a mean (SD) age of 14.44 (0.41) years. The replication samples consisted of 8690 healthy adults (4497 women [51.8%]) from 4 independent studies across the life span. A nonsynonymous genetic variant (minor T allele of rs13107325 in SLC39A8, a gene implicated in schizophrenia) was associated with greater gray matter volume of the putamen (variance explained of 4.21% in the left hemisphere; 8.66; 95% CI, 6.59-10.81; P = 5.35 × 10-18; and 4.44% in the right hemisphere; t = 8.90; 95% CI, 6.75-11.19; P = 6.80 × 10-19) and also with a lower gene expression of SLC39A8 specifically in the putamen (t127 = -3.87; P = 1.70 × 10-4). The identified association was validated in samples across the life span but was significantly weakened in both patients with schizophrenia (z = -3.05; P = .002; n = 157) and unaffected siblings (z = -2.08; P = .04; n = 149). Conclusions and Relevance Our results show that a missense mutation in gene SLC39A8 is associated with larger gray matter volume in the putamen and that this association is significantly weakened in schizophrenia. These results may suggest a role for aberrant ion transport in the etiology of psychosis and provide a target for preemptive developmental interventions aimed at restoring the functional effect of this mutation.
Collapse
Affiliation(s)
- Qiang Luo
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- School of Life Sciences and State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Social Genetic and Developmental Psychiatry Centre, London, England
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Wenjia Wang
- Pharnext, Issy-les-Moulineaux, Ile de France, France
- Institut National de la Santé et de la Recherche Médicale Unit 897, University of Bordeaux, Bordeaux, Aquitaine, France
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Social Genetic and Developmental Psychiatry Centre, London, England
| | - Erin Burke Quinlan
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Social Genetic and Developmental Psychiatry Centre, London, England
| | - Tianye Jia
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Social Genetic and Developmental Psychiatry Centre, London, England
| | - Christine Macare
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Social Genetic and Developmental Psychiatry Centre, London, England
| | - Gabriel H. Robert
- EA 4712 “Behavior and Basal Ganglia,” Rennes University 1, Rennes, France
| | - Jing Cui
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Mickaël Guedj
- Pharnext, Issy-les-Moulineaux, Ile de France, France
| | - Lena Palaniyappan
- Departments of Psychiatry and Medical Biophysics, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Ferath Kherif
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
| | - Arun L. W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Vincent Frouin
- NeuroSpin, Commissariat à L'énergie Atomique, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, England
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Germany
| | - Bernd Ittermann
- Physikalisch-Technische Bundesanstalt Braunschweig and Berlin, Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale Unit 1000, Neuroimaging and Psychiatry, University Paris Sud–Paris Saclay, University Paris Descartes, Paris, France
- Service Hospitalier Frédéric Joliot, Orsay, France
- Maison de Solenn, Paris, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale Unit 1000, Neuroimaging and Psychiatry, University Paris Sud–Paris Saclay, University Paris Descartes, Paris, France
- Service Hospitalier Frédéric Joliot, Orsay, France
- GH Nord Essonne Psychiatry Department, Orsay, France
| | - Marie-Laure Paillère-Martinot
- Institut National de la Santé et de la Recherche Médicale Unit 1000, Neuroimaging and Psychiatry, University Paris Sud–Paris Saclay, University Paris Descartes, Paris, France
- Assistance Publique–Hôpitaux de Paris, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
- Clinic for Child and Adolescent Psychiatry, Medical University of Vienna, Währinger Gürtel, Vienna, Austria
| | - Juliane H. Fröhner
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Michael N. Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité, Universitätsmedizin Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Joseph H. Callicott
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Venkata S. Mattay
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Departments of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jean-François Dartigues
- Institut National de la Santé et de la Recherche Médicale Unit 1219, Université de Bordeaux, Bordeaux, France
| | - Christophe Tzourio
- Institut National de la Santé et de la Recherche Médicale Unit 1219, Université de Bordeaux, Bordeaux, France
| | - Fabrice Crivello
- University de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France
- Centre National de la Recherche Scientifique, Institut des Maladies Neurodégénératives, Bordeaux, France
- Commissariat à L'énergie Atomiquecea, Institut des Maladies Neurodégénératives-Equipe 5, Bordeaux, France
| | - Karen F. Berman
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Fei Li
- Developmental and Behavioral Pediatric Department and Child Primary Care Department, MOE-Shanghai Key Lab for Children's Environmental Health, Xinhua Hospital Affiliated To Shang Jiaotong University School of Medicine, Shanghai, China
| | - Tomáš Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Daniel R. Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- McKusick Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Robin M. Murray
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Social Genetic and Developmental Psychiatry Centre, London, England
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Social Genetic and Developmental Psychiatry Centre, London, England
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- School of Life Sciences and State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, England
- Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
- Shanghai Center for Mathematical Sciences, Shanghai, China
| |
Collapse
|
46
|
Qi Z, Liu KJ. The interaction of zinc and the blood-brain barrier under physiological and ischemic conditions. Toxicol Appl Pharmacol 2019; 364:114-119. [PMID: 30594689 PMCID: PMC6331270 DOI: 10.1016/j.taap.2018.12.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/02/2018] [Accepted: 12/25/2018] [Indexed: 12/30/2022]
Abstract
Zinc is the second most abundant metal in human and serves as an essential trace element in the body. During the past decades, zinc has been found to play important roles in central nervous system, such as the development of neurons and synaptic activities. An imbalance of zinc is associated with brain diseases. The blood-brain barrier (BBB) maintains the homeostasis of the microenvironment, regulating the balance of zinc in the brain. A compromised BBB is the main cause of severe complications in cerebral ischemic patients, such as hemorrhage transformation, inflammation and edema. Recent studies reported that zinc in the brain may be a potential target for integrative protection against ischemic brain injury. Although zinc has long been regarded as important transmitters in central nervous system, the critical role of zinc dyshomeostasis in damage to the BBB has not been fully recognized. In this review, we summarize the role of the BBB in regulating homeostasis of zinc in physiological conditions and the effects of changes in zinc levels on the permeability of the BBB in cerebral ischemia. The integrity of BBB maintains the homeostasis of zinc in pathological conditions, while the balance of zinc in the brain and the circulation maintains the normal function of the BBB. Interrupting the zinc/BBB system will disturb the microenvironment in the brain, leading to pathological diseases. In stroke patients, zinc may serve as a potential target for protecting the BBB and reducing hemorrhage transformation, inflammation and edema.
Collapse
Affiliation(s)
- Zhifeng Qi
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ke Jian Liu
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
47
|
Filippini T, Cilloni S, Malavolti M, Violi F, Malagoli C, Tesauro M, Bottecchi I, Ferrari A, Vescovi L, Vinceti M. Dietary intake of cadmium, chromium, copper, manganese, selenium and zinc in a Northern Italy community. J Trace Elem Med Biol 2018; 50:508-517. [PMID: 29548610 DOI: 10.1016/j.jtemb.2018.03.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/26/2018] [Accepted: 03/02/2018] [Indexed: 02/06/2023]
Abstract
This study provides the dietary intakes of six trace elements (cadmium, chromium, copper, manganese, selenium and zinc), generally characterized by both nutritional and toxicological features depending on their exposure. Being diet the most relevant source of exposure to trace elements in non-professionally exposed subjects, we measured content of these trace elements in foods composing the typical Italian diet using inductively coupled plasma-mass spectrometry, and assessing dietary habits using a validated semi-quantitative food frequency questionnaire we eventually estimated dietary daily intake of trace elements in a Northern Italian community. In the 890 analyzed food samples, the main contributors to cadmium intake are cereals, vegetables and sweets, while cereals, beverages and vegetable are to primary source of manganese. The primary contributors for copper are cereals, fresh fruits and vegetables, while for chromium are beverages, cereals and meat. The main source of selenium intake are cereals and meat, followed by fish, seafood and milk and dairy products, while of zinc intake are meat, cereals, milk and dairy products. In our Italian population sample, the estimated median (interquartile range) dietary daily intakes are 5.00 (3.17-7.65), 56.70 (36.08-86.70) and 66.53 (40.04-101.32) μg/day for cadmium, chromium and selenium, and corresponding figures are 0.98 (0.61-1.49), 2.34 (1.46-3.52) and 8.50 (5.21-12.48) mg/day for copper, manganese and zinc. The estimated intakes are generally within the average intake reported in other European populations, and in such cases well above the daily dietary intakes recommended by national international agencies, avoiding the risk of excess or deficiency. The present estimated intake data can be used to examine a specific trace element of interest and would afford enhanced health protection from those trace elements characterized by both nutritional and toxicological effects.
Collapse
Affiliation(s)
- Tommaso Filippini
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy
| | - Silvia Cilloni
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy
| | - Marcella Malavolti
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy
| | - Federica Violi
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy
| | - Carlotta Malagoli
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy
| | - Marina Tesauro
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Ilaria Bottecchi
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy
| | - Angela Ferrari
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy
| | | | - Marco Vinceti
- CREAGEN, Environmental, Genetic and Nutritional Epidemiology Research Center, Section of Public Health - Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 287 Via Campi, 41125 Modena, Italy; Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
48
|
Wolf C, Weth A, Walcher S, Lax C, Baumgartner W. Modeling of Zinc Dynamics in the Synaptic Cleft: Implications for Cadherin Mediated Adhesion and Synaptic Plasticity. Front Mol Neurosci 2018; 11:306. [PMID: 30233309 PMCID: PMC6131644 DOI: 10.3389/fnmol.2018.00306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022] Open
Abstract
While the numerous influences of synaptically released zinc on synaptic efficiency during long-term potentiation have been discussed by many authors already, we focused on the possible effect of zinc on cadherins and therefore its contribution to morphological changes in the context of synaptic plasticity. The difficulty with gaining insights into the dynamics of zinc-cadherin interaction is the inability to directly observe it on a suitable timescale. Therefore our approach was to establish an analytical model of the zinc diffusion dynamics in the synaptic cleft and experimentally validate, if the theoretical concentrations at the periphery of the synaptic cleft are sufficient to significantly modulate cadherin-mediated adhesion. Our results emphasize, that synaptically released zinc might have a strong accelerating effect on the morphological changes involved in long-term synaptic plasticity. The approach presented here might also prove useful for investigations on other synaptically released trace metals.
Collapse
Affiliation(s)
- Christoph Wolf
- Institute of Medical Biomechatronics, Johannes Kepler University Linz, Linz, Austria
| | - Agnes Weth
- Institute of Medical Biomechatronics, Johannes Kepler University Linz, Linz, Austria
| | | | - Christian Lax
- Lehrstuhl A für Mathematik, RWTH-Aachen University, Aachen, Germany
| | - Werner Baumgartner
- Institute of Medical Biomechatronics, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
49
|
Abstract
Evidence from both preclinical and clinical studies suggest the importance of zinc homeostasis in seizures/epilepsy. Undoubtedly, zinc, via modulation of a variety of targets, is necessary for maintaining the balance between neuronal excitation and inhibition, while an imbalance between excitation and inhibition underlies seizures. However, the relationship between zinc signaling and seizures/epilepsy is complex as both extracellular and intracellular zinc may produce either protective or detrimental effects. This review provides an overview of preclinical/behavioral, functional and molecular studies, as well as clinical data on the involvement of zinc in the pathophysiology and treatment of seizures/epilepsy. Furthermore, the potential of targeting elements associated with zinc signaling or homeostasis and zinc levels as a therapeutic strategy for epilepsy is discussed.
Collapse
Affiliation(s)
- Urszula Doboszewska
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Katarzyna Młyniec
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland
| | - Aleksandra Wlaź
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Gabriel Nowak
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland; Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Piotr Wlaź
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
50
|
Abstract
Appraising success in meeting the world's nutritional needs has largely focused on infant mortality and anthropometric measurements with an emphasis on the first 1,000 days (conception to approximately age 2 years). This ignores the unique nutritional needs of the human brain. Although the intrauterine environment and the early postnatal years are important, equally critical periods follow during which the brain's intricate wiring is established for a lifetime of experience-driven remodeling. At the peak of this process during childhood, the human brain may account for 50% of the body's basal nutritional requirement. Thus, the consequences of proper nutritional management of the brain play out over a lifetime. Our motivation in preparing this review was to move the human brain into a more central position in the planning of nutritional programs. Here we review the macro- and micronutrient requirements of the human brain and how they are delivered, from conception to adulthood.
Collapse
Affiliation(s)
- Manu S. Goyal
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| | - Lora L. Iannotti
- Brown School, Institute for Public Health, Washington University, St. Louis, Missouri 63130, USA
| | - Marcus E. Raichle
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| |
Collapse
|