1
|
Yang J, Ma G, Du X, Xie J, Wang M, Wang W, Guo B, Wu S. Deciphering the Role of Shank3 in Dendritic Morphology and Synaptic Function Across Postnatal Developmental Stages in the Shank3B KO Mouse. Neurosci Bull 2025; 41:583-599. [PMID: 39693031 PMCID: PMC11978597 DOI: 10.1007/s12264-024-01330-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/14/2024] [Indexed: 12/19/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is marked by early-onset neurodevelopmental anomalies, yet the temporal dynamics of genetic contributions to these processes remain insufficiently understood. This study aimed to elucidate the role of the Shank3 gene, known to be associated with monogenic causes of autism, in early developmental processes to inform the timing and mechanisms for potential interventions for ASD. Utilizing the Shank3B knockout (KO) mouse model, we examined Shank3 expression and its impact on neuronal maturation through Golgi staining for dendritic morphology and electrophysiological recordings to measure synaptic function in the anterior cingulate cortex (ACC) across different postnatal stages. Our longitudinal analysis revealed that, while Shank3B KO mice displayed normal neuronal morphology at one week postnatal, significant impairments in dendritic growth and synaptic activity emerged by two to three weeks. These findings highlight the critical developmental window during which Shank3 is essential for neuronal and synaptic maturation in the ACC.
Collapse
Affiliation(s)
- Jing Yang
- Department of Basic Medicine, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Guaiguai Ma
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaohui Du
- Department of Basic Medicine, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Jinyi Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Mengmeng Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Baolin Guo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
- Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Gao W, Cai Q, Ying X, Zhao B. Establishing a Mouse Model of NL3R617W-Associated Autism Spectrum Disorder for a Functional Study. ACTAS ESPANOLAS DE PSIQUIATRIA 2025; 53:253-266. [PMID: 40071366 PMCID: PMC11898267 DOI: 10.62641/aep.v53i2.1780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and limited behavior. Despite the association of numerous synaptic gene mutations with ASD, the presence of behavioral abnormalities in mice expressing autism-associated R617W mutation in synaptic adhesion protein neuroligin-3 (NL3) has not been established. This work focuses on establishing a mouse model of ASD caused by NL3 R617W missense mutation (NL3R617W) and characterizing and profiling the molecular as well as behavioral features of the animal model. METHODS The expression and distribution of NL3R617W mutant protein in the 293T cell membrane and intracellular NL3 was detected by using immunofluorescence approach. Meanwhile, synaptic markers (Synapsin I, vesicular glutamate transporter (VGluT) I and vesicular γ-aminobutyric acid transporter (VGAT)) and synapse number were detected with a confocal fluorescence microscope. Thereafter, the effect on NL3R617W was verified. The expression of synaptic proteins, postsynaptic density protein-95 (PSD95) and Src homology domain and multiple ankyrin repeat domains protein 3 (SHANK3), was verified by Western blot. The interaction between NL3 and neurexin 1 (NRXN1) was studied by means of co-immunoprecipitation. The behavior of autistic mice induced by NL3R617W mutation was examined using the Morris water maze and the Y maze. NL3R617W mutant mice were assessed in the open field, and three-chamber test was conducted to assess and observe the presence of hyperactivity, repetitive behavior, friendliness, and social novelty. RESULTS The results indicated that the NL3 mutation could influence the interaction between NL3 and NRXN1, and inhibit the expression of VGluT I. Nevertheless, NL3 mutation would not influence the expression of NL3 on cell membrane, the intracellular distribution of NL3, or the endoplasmic reticulum retention. The outcomes of animal studies demonstrated that the ASD mice with NL3R617W exhibited a significant decrease in the capacity for spatial memory and exploration, as well as the expression levels of the postsynaptic scaffolding proteins, PSD95 and SHANK3 (p < 0.05). The number of excitatory synapses in hippocampal cornu ammonis (CA)1 and CA3 and the sensory cortex was also significantly reduced (p < 0.01). Compared to the control mice, the NL3R617W mutant mice were less active in the open field (p < 0.001), a finding consistent with the three-chamber test result showing reduced degree of activity. Furthermore, compared to the control mice, the NL3R617W mutant animals spent less time with stranger mice (p < 0.05). CONCLUSIONS NL3R617W mutation may inhibit the expression of postsynaptic scaffolding proteins by influencing the interaction with NRXN1, thus inhibiting synapse formation and reducing the number of excitatory synapses.
Collapse
Affiliation(s)
- Wei Gao
- Children’s Health Department, Hongkou District Maternal and Child Health Institution, 200000 Shanghai, China
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| | - Qiao Cai
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| | - Xiaoming Ying
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| | - Bei Zhao
- Department of Pediatric, The First People’s Hospital of Taizhou, 318020 Taizhou, Zhejiang, China
| |
Collapse
|
3
|
Liang J, Zhao J, Yang L, Wang Q, Liao J, Li J, Zhuang W, Li F, He J, Tang Y, Chen H, Huang C. MSC-exosomes pretreated by Danshensu extracts pretreating to target the hsa-miR-27a-5p and STAT3-SHANK2 to enhanced antifibrotic therapy. Stem Cell Res Ther 2025; 16:40. [PMID: 39901236 PMCID: PMC11792327 DOI: 10.1186/s13287-025-04181-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Peritoneal fibrosis (PF) is a serious complication commonly associated with prolonged peritoneal dialysis. Mesenchymal stem cells (MSCs) and their exosomes (Exo) have shown significant therapeutic promise in treating fibrotic conditions. Danshensu (DSS), a bioactive compound from the traditional Chinese herb Danshen reverses fibrosis. This study aims to investigate a novel strategy to enhance the therapeutic efficacy against PF by DSS preconditioning MSCs-derived exosomes (DSS-Exo). METHODS The in vitro studies included the effects of DSS duration on MSCs, and the characterization of DSS-Exo and Exo, followed by the assessment of RNA and protein expression levels of peritoneal fibrosis markers and inflammatory cytokines levels after treating human peritoneal mesothelial (HMrSV5) cells. In vivo experiments were conducted on a PF mouse model to observe cell morphology, collagen deposition, fibrosis localization, and to evaluate peritoneal functions such as filtration rate, urea nitrogen clearance, peritoneal thickness, and protein leakage. Mechanistic insights were gained through the analysis of the STAT3/HIF-1α/VEGF signaling pathway, tissue dual-fluorescence localization,chromatin immunoprecipitation sequencing (ChIP-seq), and dual-luciferase reporter (DLR) assays. Additionally, the differential expression of miRNAs between DSS-Exo and Exo was explored and validation of key miRNA. RESULTS DSS-Exo significantly upregulated E-cadherin, downregulated VEGFA, α-SMA, CTGF and Fibronectin expression in HMrSV5 cells compared to untreated Exo. In vivo studies revealed that DSS-Exo enhanced the ability of Exo to improve peritoneal function,such as the peritoneal filtration rate and urea nitrogen, glucose clearance, while reducing peritoneal thickness and protein leakage, and cell morphology, reduce collagen deposition, and decrease the degree of fibrosis. Mechanistically, these exosomes inhibited the STAT3/HIF-1α/VEGF signaling pathway within peritoneal mesothelial tissues. Furthermore, ChIP-seq and DLR demonstrated that DSS-Exo affected STAT3 directly binds to SHANK2 promoter regions, forming hydrogen bonds between 5 key amino acids such as GLN-344, HIS-332 and 6 key bases such as DG-258, DG-261. miRNA profiling identified DSS-Exo increased hsa-miR-27a-5p_R-1 to regulated STAT3-SHANK2 and modulating the EMT. CONCLUSION This study highlighted the innovative use of Danshensu in enhancing MSC-derived exosome therapy for PF. The identification of the hsa-miR-27a-5p_R-1-STAT3-SHANK2 axis may reveal new molecular mechanisms underlying fibrosis, further research is needed to fully elucidate its impact on PF. The integration of Danshensu from traditional Chinese medicine into modern MSC exosome therapy represents a promising frontier in the development of novel treatments for fibrotic diseases.
Collapse
Affiliation(s)
- Jiabin Liang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingxiu Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Laboratory Science, ShunDe Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Yang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Wang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Liao
- Panyu Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Jianhao Li
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weizhao Zhuang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fanghong Li
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinxian He
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yukuan Tang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanwei Chen
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China.
- Panyu Health Management Center, Guangzhou, 511400, China.
| | - Chen Huang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China.
- Medical Imaging Institute of Panyu, Guangzhou, 511400, China.
| |
Collapse
|
4
|
Barón-Mendoza I, Márquez LA, Arenas AG, Guzmán-Condado J, Martínez-Rojas VA, Anguiano-Buenfil J, Mejía-Hernández M, Almazán JL, Pérez-Martínez L, Pedraza-Alva G, Galván EJ, Zepeda A. Single-nucleotide polymorphism analysis accurately predicts multiple impairments in hippocampal activity and memory performance in a murine model of idiopathic autism. Sci Rep 2025; 15:749. [PMID: 39755808 PMCID: PMC11700144 DOI: 10.1038/s41598-024-84521-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
Autism spectrum disorder (ASD) comprises alterations in brain anatomy and physiology that ultimately affect information processing and behavior. In most cases, autism is considered idiopathic, involving alterations in numerous genes whose functions are not extensively documented. We evaluated the C58/J mouse strain as an idiopathic model of ASD, emphasizing synaptic transmission as the basis of information processing. Through in silico analysis, we found that the C58/J strain carries single nucleotide polymorphisms (SNPs) compared to the C57BL/6J control strain related to synaptic structure and LTP induction. These SNPs have human orthologs previously associated with ASD. We then assessed chemical potentiation (cLTP) in synaptosomes, the electrophysiological properties of hippocampal CA3 cells, and the induction of LTP in ex-vivo slices. An increased proportion of synaptosomes expressing the GluA1 subunit of AMPA receptor and Nrx1β in the membrane was found in the C57BL/6J control strain, but not in C58/J mice, after cLTP induction. Additionally, several electrophysiological properties of CA3 pyramidal cells and hippocampal communication were altered. Behaviorally, C58/J mice exhibited hyperactivity and subtle memory changes. Our results demonstrate that an idiopathic model of ASD exhibits alterations in hippocampal physiology from the cellular to the circuitry and behavioral levels.
Collapse
Affiliation(s)
- Isabel Barón-Mendoza
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luis A Márquez
- Departamento de Farmacobiología, CINVESTAV Unidad Sur CdMx, Mexico City, Mexico
| | - Aliesha González Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Jessica Guzmán-Condado
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | | | | | - Montserrat Mejía-Hernández
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Jorge Luis Almazán
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología UNAM, Cuernavaca, Morelos, 62210, Mexico City, Mexico
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología UNAM, Cuernavaca, Morelos, 62210, Mexico City, Mexico
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología UNAM, Cuernavaca, Morelos, 62210, Mexico City, Mexico
| | - Emilio J Galván
- Departamento de Farmacobiología, CINVESTAV Unidad Sur CdMx, Mexico City, Mexico.
- Centro de Investigaciones Sobre El Envejecimiento, CIE-Cinvestav, México City, Mexico.
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
5
|
Xu QW, Larosa A, Wong TP. Roles of AMPA receptors in social behaviors. Front Synaptic Neurosci 2024; 16:1405510. [PMID: 39056071 PMCID: PMC11269240 DOI: 10.3389/fnsyn.2024.1405510] [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: 03/23/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
As a crucial player in excitatory synaptic transmission, AMPA receptors (AMPARs) contribute to the formation, regulation, and expression of social behaviors. AMPAR modifications have been associated with naturalistic social behaviors, such as aggression, sociability, and social memory, but are also noted in brain diseases featuring impaired social behavior. Understanding the role of AMPARs in social behaviors is timely to reveal therapeutic targets for treating social impairment in disorders, such as autism spectrum disorder and schizophrenia. In this review, we will discuss the contribution of the molecular composition, function, and plasticity of AMPARs to social behaviors. The impact of targeting AMPARs in treating brain disorders will also be discussed.
Collapse
Affiliation(s)
- Qi Wei Xu
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Amanda Larosa
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Tak Pan Wong
- Douglas Hospital Research Centre, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Manning A, Bender PTR, Boyd-Pratt H, Mendelson BZ, Hruska M, Anderson CT. Trans-synaptic Association of Vesicular Zinc Transporter 3 and Shank3 Supports Synapse-Specific Dendritic Spine Structure and Function in the Mouse Auditory Cortex. J Neurosci 2024; 44:e0619242024. [PMID: 38830758 PMCID: PMC11236586 DOI: 10.1523/jneurosci.0619-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Shank3 is a synaptic scaffolding protein that assists in tethering and organizing structural proteins and glutamatergic receptors in the postsynaptic density of excitatory synapses. The localization of Shank3 at excitatory synapses and the formation of stable Shank3 complexes is regulated by the binding of zinc to the C-terminal sterile-alpha-motif (SAM) domain of Shank3. Mutations in the SAM domain of Shank3 result in altered synaptic function and morphology, and disruption of zinc in synapses that express Shank3 leads to a reduction of postsynaptic proteins important for synaptic structure and function. This suggests that zinc supports the localization of postsynaptic proteins via Shank3. Many regions of the brain are highly enriched with free zinc inside glutamatergic vesicles at presynaptic terminals. At these synapses, zinc transporter 3 (ZnT3) moves zinc into vesicles where it is co-released with glutamate. Alterations in ZnT3 are implicated in multiple neurodevelopmental disorders, and ZnT3 knock-out (KO) mice-which lack synaptic zinc-show behavioral deficits associated with autism spectrum disorder and schizophrenia. Here we show that male and female ZnT3 KO mice have smaller dendritic spines and miniature excitatory postsynaptic current amplitudes than wildtype (WT) mice in the auditory cortex. Additionally, spine size deficits in ZnT3 KO mice are restricted to synapses that express Shank3. In WT mice, synapses that express both Shank3 and ZnT3 have larger spines compared to synapses that express Shank3 but not ZnT3. Together these findings suggest a mechanism whereby presynaptic ZnT3-dependent zinc supports postsynaptic structure and function via Shank3 in a synapse-specific manner.
Collapse
Affiliation(s)
- Abbey Manning
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Philip T R Bender
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Helen Boyd-Pratt
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
- Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Benjamin Z Mendelson
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Martin Hruska
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Charles T Anderson
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| |
Collapse
|
7
|
Tian Y, Yu F, Yun E, Lin JW, Man HY. mRNA nuclear retention reduces AMPAR expression and promotes autistic behavior in UBE3A-overexpressing mice. EMBO Rep 2024; 25:1282-1309. [PMID: 38316900 PMCID: PMC10933332 DOI: 10.1038/s44319-024-00073-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/07/2024] [Accepted: 01/16/2024] [Indexed: 02/07/2024] Open
Abstract
UBE3A is a common genetic factor in ASD etiology, and transgenic mice overexpressing UBE3A exhibit typical autistic-like behaviors. Because AMPA receptors (AMPARs) mediate most of the excitatory synaptic transmission in the brain, and synaptic dysregulation is considered one of the primary cellular mechanisms in ASD pathology, we investigate here the involvement of AMPARs in UBE3A-dependent ASD. We show that expression of the AMPAR GluA1 subunit is decreased in UBE3A-overexpressing mice, and that AMPAR-mediated neuronal activity is reduced. GluA1 mRNA is trapped in the nucleus of UBE3A-overexpressing neurons, suppressing GluA1 protein synthesis. Also, SARNP, an mRNA nuclear export protein, is downregulated in UBE3A-overexpressing neurons, causing GluA1 mRNA nuclear retention. Restoring SARNP levels not only rescues GluA1 mRNA localization and protein expression, but also normalizes neuronal activity and autistic behaviors in mice overexpressing UBE3A. These findings indicate that SARNP plays a crucial role in the cellular and behavioral phenotypes of UBE3A-induced ASD by regulating nuclear mRNA trafficking and protein translation of a key AMPAR subunit.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - Feiyuan Yu
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - Eunice Yun
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - Jen-Wei Lin
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA.
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
8
|
Ross MM, Hernandez-Espinosa DR, Aizenman E. Neurodevelopmental Consequences of Dietary Zinc Deficiency: A Status Report. Biol Trace Elem Res 2023; 201:5616-5639. [PMID: 36964812 DOI: 10.1007/s12011-023-03630-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/11/2023] [Indexed: 03/26/2023]
Abstract
Zinc is a tightly regulated trace mineral element playing critical roles in growth, immunity, neurodevelopment, and synaptic and hormonal signaling. Although severe dietary zinc deficiency is relatively uncommon in the United States, dietary zinc deficiency is a substantial public health concern in low- and middle-income countries. Zinc status may be a key determinant of neurodevelopmental processes. Indeed, limited cohort studies have shown that serum zinc is lower in people diagnosed with autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and depression. These observations have sparked multiple studies investigating the mechanisms underlying zinc status and neurodevelopmental outcomes. Animal models of perinatal and adult dietary zinc restriction yield distinct behavioral phenotypes reminiscent of features of ASD, ADHD, and depression, including increased anxiety and immobility, repetitive behaviors, and altered social behaviors. At the cellular and molecular level, zinc has demonstrated roles in neurogenesis, regulation of cellular redox status, transcription factor trafficking, synaptogenesis, and the regulation of synaptic architecture via the Shank family of scaffolding proteins. Although mechanistic questions remain, the current evidence suggests that zinc status is important for adequate neuronal development and may be a yet overlooked factor in the pathogenesis of several psychiatric conditions. This review aims to summarize current knowledge of the role of zinc in the neurophysiology of the perinatal period, the many cellular targets of zinc in the developing brain, and the potential consequences of alterations in zinc homeostasis in early life.
Collapse
Affiliation(s)
- Madeline M Ross
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Diego R Hernandez-Espinosa
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
9
|
Ross MM, Aizenman E. GluA1-Shank3 interaction decreases in response to chronic neuronal depolarization. Neurosci Lett 2023; 809:137305. [PMID: 37210067 PMCID: PMC10330850 DOI: 10.1016/j.neulet.2023.137305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Interactions between AMPA receptors and synaptic scaffolding proteins are key regulators of synaptic receptor density and, thereby, synapse strength. Shank3 is one such scaffolding protein with high clinical relevance, as genetic variants and deletions of this protein have been linked to autism spectrum disorder. Shank3 acts as a master regulator of the postsynaptic density of glutamatergic synapses, interacting with ionotropic and metabotropic glutamate receptors and cytoskeletal elements to modulate synaptic structure. Notably, Shank3 has been shown to interact directly with the AMPAR subunit GluA1, and Shank3 knockout animals show deficits in AMPAR-mediated synaptic transmission. In this study, we sought to characterize the stability of GluA1-Shank3 interaction in response to chronic stimuli using a highly sensitive and specific proximity ligation assay. We found that GluA1-Shank3 interactions decrease in response to prolonged neuronal depolarization induced by elevated extracellular potassium, and that this reduced interaction is blocked by NMDA receptor antagonism. These results firmly establish the close interaction of GluA1 and Shank3 in cortical neurons in vitro, and that this select interaction is subject to modulation by depolarization.
Collapse
Affiliation(s)
- Madeline M Ross
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
10
|
Anchesi I, Schepici G, Chiricosta L, Gugliandolo A, Salamone S, Caprioglio D, Pollastro F, Mazzon E. Δ 8-THC Induces Up-Regulation of Glutamatergic Pathway Genes in Differentiated SH-SY5Y: A Transcriptomic Study. Int J Mol Sci 2023; 24:ijms24119486. [PMID: 37298437 DOI: 10.3390/ijms24119486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Cannabinoids, natural or synthetic, have antidepressant, anxiolytic, anticonvulsant, and anti-psychotic properties. Cannabidiol (CBD) and delta-9-tetrahydrocannabinol (Δ9-THC) are the most studied cannabinoids, but recently, attention has turned towards minor cannabinoids. Delta-8-tetrahydrocannabinol (Δ8-THC), an isomer of Δ9-THC, is a compound for which, to date, there is no evidence of its role in the modulation of synaptic pathways. The aim of our work was to evaluate the effects of Δ8-THC on differentiated SH-SY5Y human neuroblastoma cells. Using next generation sequencing (NGS), we investigated whether Δ8-THC could modify the transcriptomic profile of genes involved in synapse functions. Our results showed that Δ8-THC upregulates the expression of genes involved in the glutamatergic pathway and inhibits gene expression at cholinergic synapses. Conversely, Δ8-THC did not modify the transcriptomic profile of genes involved in the GABAergic and dopaminergic pathways.
Collapse
Affiliation(s)
- Ivan Anchesi
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Giovanni Schepici
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Diego Caprioglio
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
11
|
Costa RT, Santos MB, Alberto-Silva C, Carrettiero DC, Ribeiro CAJ. Methylmalonic Acid Impairs Cell Respiration and Glutamate Uptake in C6 Rat Glioma Cells: Implications for Methylmalonic Acidemia. Cell Mol Neurobiol 2023; 43:1163-1180. [PMID: 35674974 PMCID: PMC11414442 DOI: 10.1007/s10571-022-01236-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/20/2022] [Indexed: 11/28/2022]
Abstract
Methylmalonic acidemia is an organic acidemia caused by deficient activity of L-methylmalonyl-CoA mutase or its cofactor cyanocobalamin and it is biochemically characterized by an accumulation of methylmalonic acid (MMA) in tissue and body fluids of patients. The main clinical manifestations of this disease are neurological and observable symptoms during metabolic decompensation are encephalopathy, cerebral atrophy, coma, and seizures, which commonly appear in newborns. This study aimed to investigate the toxic effects of MMA in a glial cell line presenting astrocytic features. Astroglial C6 cells were exposed to MMA (0.1-10 mM) for 24 or 48 h and cell metabolic viability, glucose consumption, and oxygen consumption rate, as well as glutamate uptake and ATP content were analyzed. The possible preventive effects of bezafibrate were also evaluated. MMA significantly reduced cell metabolic viability after 48-h period and increased glucose consumption during the same period of incubation. Regarding the energy homeostasis, MMA significantly reduced respiratory parameters of cells after 48-h exposure, indicating that cell metabolism is compromised at resting and reserve capacity state, which might influence the cell capacity to meet energetic demands. Glutamate uptake and ATP content were also compromised after exposure to MMA, which can be influenced energy metabolism impairment, affecting the functionality of the astroglial cells. Our findings suggest that these effects could be involved in the pathophysiology of neurological dysfunction of this disease. Methylmalonic acid compromises mitochondrial functioning leading to reduced ATP production and reduces glutamate uptake by C6 astroglial cells.
Collapse
Affiliation(s)
- Renata T Costa
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - Marcella B Santos
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - Carlos Alberto-Silva
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - Daniel C Carrettiero
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil
| | - César A J Ribeiro
- Centro de Ciências Naturais E Humanas (CCNH), UFABC - Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo, SP, CEP 09606-045, Brazil.
| |
Collapse
|
12
|
Alshawaf AJ, Alnassar SA, Al-Mohanna FA. The interplay of intracellular calcium and zinc ions in response to electric field stimulation in primary rat cortical neurons in vitro. Front Cell Neurosci 2023; 17:1118335. [PMID: 37180947 PMCID: PMC10174245 DOI: 10.3389/fncel.2023.1118335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Recent pharmacological studies demonstrate a role for zinc (Zn2+) in shaping intracellular calcium (Ca2+) dynamics and vice versa in excitable cells including neurons and cardiomyocytes. Herein, we sought to examine the dynamic of intracellular release of Ca2+ and Zn2+ upon modifying excitability of primary rat cortical neurons using electric field stimulation (EFS) in vitro. We show that exposure to EFS with an intensity of 7.69 V/cm induces transient membrane hyperpolarization together with transient elevations in the cytosolic levels of Ca2+ and Zn2+ ions. The EFS-induced hyperpolarization was inhibited by prior treatment of cells with the K+ channel opener diazoxide. Chemical hyperpolarization had no apparent effect on either Ca2+ or Zn2+. The source of EFS-induced rise in Ca2+ and Zn2+ seemed to be intracellular, and that the dynamic inferred of an interplay between Ca2+ and Zn2+ ions, whereby the removal of extracellular Ca2+ augmented the release of intracellular Ca2+ and Zn2+ and caused a stronger and more sustained hyperpolarization. We demonstrate that Zn2+ is released from intracellular vesicles located in the soma, with major co-localizations in the lysosomes and endoplasmic reticulum. These studies further support the use of EFS as a tool to interrogate the kinetics of intracellular ions in response to changing membrane potential in vitro.
Collapse
Affiliation(s)
- Abdullah J. Alshawaf
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sarah A. Alnassar
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Futwan A. Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- *Correspondence: Futwan A. Al-Mohanna,
| |
Collapse
|
13
|
Lee K, Mills Z, Cheung P, Cheyne JE, Montgomery JM. The Role of Zinc and NMDA Receptors in Autism Spectrum Disorders. Pharmaceuticals (Basel) 2022; 16:ph16010001. [PMID: 36678498 PMCID: PMC9866730 DOI: 10.3390/ph16010001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
NMDA-type glutamate receptors are critical for synaptic plasticity in the central nervous system. Their unique properties and age-dependent arrangement of subunit types underpin their role as a coincidence detector of pre- and postsynaptic activity during brain development and maturation. NMDAR function is highly modulated by zinc, which is co-released with glutamate and concentrates in postsynaptic spines. Both NMDARs and zinc have been strongly linked to autism spectrum disorders (ASDs), suggesting that NMDARs are an important player in the beneficial effects observed with zinc in both animal models and children with ASDs. Significant evidence is emerging that these beneficial effects occur via zinc-dependent regulation of SHANK proteins, which form the backbone of the postsynaptic density. For example, dietary zinc supplementation enhances SHANK2 or SHANK3 synaptic recruitment and rescues NMDAR deficits and hypofunction in Shank3ex13-16-/- and Tbr1+/- ASD mice. Across multiple studies, synaptic changes occur in parallel with a reversal of ASD-associated behaviours, highlighting the zinc-dependent regulation of NMDARs and glutamatergic synapses as therapeutic targets for severe forms of ASDs, either pre- or postnatally. The data from rodent models set a strong foundation for future translational studies in human cells and people affected by ASDs.
Collapse
|
14
|
Austin C, Curtin P, Arora M, Reichenberg A, Curtin A, Iwai-Shimada M, Wright RO, Wright RJ, Remnelius KL, Isaksson J, Bölte S, Nakayama SF. Elemental Dynamics in Hair Accurately Predict Future Autism Spectrum Disorder Diagnosis: An International Multi-Center Study. J Clin Med 2022; 11:jcm11237154. [PMID: 36498727 PMCID: PMC9740182 DOI: 10.3390/jcm11237154] [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: 10/24/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition diagnosed in approximately 2% of children. Reliance on the emergence of clinically observable behavioral patterns only delays the mean age of diagnosis to approximately 4 years. However, neural pathways critical to language and social functions develop during infancy, and current diagnostic protocols miss the age when therapy would be most effective. We developed non-invasive ASD biomarkers using mass spectrometry analyses of elemental metabolism in single hair strands, coupled with machine learning. We undertook a national prospective study in Japan, where hair samples were collected at 1 month and clinical diagnosis was undertaken at 4 years. Next, we analyzed a national sample of Swedish twins and, in our third study, participants from a specialist ASD center in the US. In a blinded analysis, a predictive algorithm detected ASD risk as early as 1 month with 96.4% sensitivity, 75.4% specificity, and 81.4% accuracy (n = 486; 175 cases). These findings emphasize that the dynamics in elemental metabolism are systemically dysregulated in autism, and these signatures can be detected and leveraged in hair samples to predict the emergence of ASD as early as 1 month of age.
Collapse
Affiliation(s)
- Christine Austin
- Linus Biotechnology Inc., New York, NY 10013, USA
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Paul Curtin
- Linus Biotechnology Inc., New York, NY 10013, USA
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, NY 10029, USA
- Correspondence: (P.C.); (M.A.)
| | - Manish Arora
- Linus Biotechnology Inc., New York, NY 10013, USA
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, NY 10029, USA
- Correspondence: (P.C.); (M.A.)
| | - Abraham Reichenberg
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, NY 10029, USA
- Seaver Autism Center, Department of Psychiatry, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Austen Curtin
- Linus Biotechnology Inc., New York, NY 10013, USA
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Miyuki Iwai-Shimada
- Exposure Dynamics Research Section, Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Japan
| | - Robert O. Wright
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Rosalind J. Wright
- Environmental Medicine and Public Health, Mount Sinai School of Medicine, New York, NY 10029, USA
- Department of Pediatrics, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Karl Lundin Remnelius
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women’s and Children’s Health, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, 11330 Stockholm, Sweden
| | - Johan Isaksson
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women’s and Children’s Health, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, 11330 Stockholm, Sweden
- Department of Medical Sciences, Child and Adolescent Psychiatry Unit, Uppsala University, 75185 Uppsala, Sweden
| | - Sven Bölte
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research, Department of Women’s and Children’s Health, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, 11330 Stockholm, Sweden
- Child and Adolescent Psychiatry, Stockholm Health Care Services, Region Stockholm, 11861 Stockholm, Sweden
- Curtin Autism Research Group, Curtin School of Allied Health, Curtin University, Perth, WA 6102, Australia
| | - Shoji F. Nakayama
- Exposure Dynamics Research Section, Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Japan
| |
Collapse
|
15
|
Krall R, Gale JR, Ross MM, Tzounopoulos T, Aizenman E. Intracellular zinc signaling influences NMDA receptor function by enhancing the interaction of ZnT1 with GluN2A. Neurosci Lett 2022; 790:136896. [PMID: 36202195 PMCID: PMC10153101 DOI: 10.1016/j.neulet.2022.136896] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022]
Abstract
Zinc, loaded into glutamate-containing presynaptic vesicles and released into the synapse in an activity-dependent manner, modulates neurotransmission through its actions on postsynaptic targets, prominently via high-affinity inhibition of GluN2A-containing NMDA receptors. Recently, we identified a postsynaptic transport mechanism that regulates endogenous zinc inhibition of NMDARs. In this new model of zinc regulation, the postsynaptic transporter ZnT1 mediates zinc inhibition of NMDARs by binding to GluN2A. Through this interaction, ZnT1, a transporter that moves zinc from the cytoplasm to the extracellular domain, generates a zinc microdomain that modulates NMDAR-mediated neurotransmission. As ZnT1 expression is transcriptionally driven by the metal-responsive transcription factor 1 (MTF-1), we found that intracellular zinc strongly drives MTF-1 in cortical neurons in vitro and increases the number of GluN2A-ZnT1 interactions, thereby enhancing tonic zinc inhibition of NMDAR-mediated currents. Importantly, this effect is absent when the interaction between GluN2A and ZnT1 is disrupted by a cell-permeable peptide. These results suggest that zinc-regulated gene expression can dynamically regulate NMDAR-mediated synaptic processes.
Collapse
Affiliation(s)
- Rebecca Krall
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Hearing Research Center, University of Pittsburgh, PA, USA
| | - Jenna R Gale
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline M Ross
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, University of Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Hearing Research Center, University of Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Yasuda H, Tsutsui T. Metallomics analysis for early assessment and individualized intervention of neurodevelopmental disorders. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6695310. [PMID: 36087072 DOI: 10.1093/mtomcs/mfac067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/28/2022] [Indexed: 11/12/2022]
Abstract
The children today are in the midst of the epidemic of neurodevelopmental disorders. In this metallomics study for the scalp hair samples of total 2550 children with autistic disorders (2108 males and 442 females aged 0-15 year), it was demonstrated that near one half of the infantile individuals aged 0-3 year are suffering from zinc deficiency and toxic metal burdens. Zinc level correlated closely to the index of zinc/iron ratio more than zinc/copper ratio. Furthermore, there were significant relationships between zinc deficiency and toxic metal burdens such as lead and aluminum which were inversely associated with not only zinc level but also zinc/iron ratio with higher regression coefficients of r = -0.486 and -0.551 (p < 0.00001), respectively. High-significant inverse association was detected between zinc and molybdenum concentration (r = -0.509) and also between zinc/iron ratio and molybdenum (r = -0.548). These findings suggest that infantile zinc deficiency relates to the high burdens of not only toxic but also some essential metals such as molybdenum, iron and manganese and that these various mineral imbalances play principal roles to the etiology of neurodevelopmental disorders. We expect that the early assessment and intervention of the mineral imbalances (or dis-homeostasis) in individual child open an avenue for evidence-based individualized treatment of neurodevelopmental disorders and also of the comorbid immune disorders, in near future.
Collapse
Affiliation(s)
- Hiroshi Yasuda
- La Belle Vie Research Laboratory, Tokyo; Japan.,Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa, Japan
| | | |
Collapse
|
17
|
Zhang C, Dischler A, Glover K, Qin Y. Neuronal signalling of zinc: from detection and modulation to function. Open Biol 2022; 12:220188. [PMID: 36067793 PMCID: PMC9448499 DOI: 10.1098/rsob.220188] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Zinc is an essential trace element that stabilizes protein structures and allosterically modulates a plethora of enzymes, ion channels and neurotransmitter receptors. Labile zinc (Zn2+) acts as an intracellular and intercellular signalling molecule in response to various stimuli, which is especially important in the central nervous system. Zincergic neurons, characterized by Zn2+ deposits in synaptic vesicles and presynaptic Zn2+ release, are found in the cortex, hippocampus, amygdala, olfactory bulb and spinal cord. To provide an overview of synaptic Zn2+ and intracellular Zn2+ signalling in neurons, the present paper summarizes the fluorescent sensors used to detect Zn2+ signals, the cellular mechanisms regulating the generation and buffering of Zn2+ signals, as well as the current perspectives on their pleiotropic effects on phosphorylation signalling, synapse formation, synaptic plasticity, as well as sensory and cognitive function.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Anna Dischler
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Kaitlyn Glover
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Yan Qin
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| |
Collapse
|
18
|
Davies TC. The position of geochemical variables as causal co-factors of diseases of unknown aetiology. SN APPLIED SCIENCES 2022; 4:236. [PMID: 35909942 PMCID: PMC9326422 DOI: 10.1007/s42452-022-05113-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract The term diseases of unknown aetiology (DUA) or idiopathic diseases is used to describe diseases that are of uncertain or unknown cause or origin. Among plausible geoenvironmental co-factors in causation of DUA, this article focusses on the entry of trace elements, including metals and metalloids into humans, and their involvement in humoral and cellular immune responses, representing potentially toxic agents with implications as co-factors for certain DUA. Several trace elements/metals/metalloids (micronutrients) play vital roles as co-factors for essential enzymes and antioxidant molecules, thus, conferring protection against disease. However, inborn errors of trace element/metal/metalloid metabolisms can occur to produce toxicity, such as when there are basic defects in the element transport mechanism. Ultimately, it is the amount of trace element, metal or metalloid that is taken up, its mode of accumulation in human tissues, and related geomedical attributes such as the chemical form and bioavailability that decisively determine whether the exerted effects are toxic or beneficial. Several case descriptions of DUA that are common worldwide are given to illustrate our knowledge so far of how trace element/metal/metalloid interactions in the immune system may engender its dysregulation and be implicated as causal co-factors of DUA. Article highlights The importance of a proper understanding of geochemical perturbations in human metabolisms is emphasisedIt is proferred that such an understanding would aid greatly in the decipherment of diseases of unknown aetiology (DUA)The thesis presented may pave the way towards better diagnosis and therapy of DUA.
Collapse
Affiliation(s)
- Theophilus C. Davies
- Present Address: Faculty of Natural Sciences, Mangosuthu University of Technology, 511 Mangosuthu Highway, 4031, KwaZulu Natal, South Africa
| |
Collapse
|
19
|
Sauer AK, Hagmeyer S, Grabrucker AM. Prenatal Zinc Deficient Mice as a Model for Autism Spectrum Disorders. Int J Mol Sci 2022; 23:ijms23116082. [PMID: 35682762 PMCID: PMC9181257 DOI: 10.3390/ijms23116082] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies have shown a clear association between early life zinc deficiency and Autism Spectrum Disorders (ASD). In line with this, mouse models have revealed prenatal zinc deficiency as a profound risk factor for neurobiological and behavioral abnormalities in the offspring reminiscent of ASD behavior. From these studies, a complex pathology emerges, with alterations in the gastrointestinal and immune system and synaptic signaling in the brain, as a major consequence of prenatal zinc deficiency. The features represent a critical link in a causal chain that leads to various neuronal dysfunctions and behavioral phenotypes observed in prenatal zinc deficient (PZD) mice and probably other mouse models for ASD. Given that the complete phenotype of PZD mice may be key to understanding how non-genetic factors can modify the clinical features and severity of autistic patients and explain the observed heterogeneity, here, we summarize published data on PZD mice. We critically review the emerging evidence that prenatal zinc deficiency is at the core of several environmental risk factors associated with ASD, being mechanistically linked to ASD-associated genetic factors. In addition, we highlight future directions and outstanding questions, including potential symptomatic, disease-modifying, and preventive treatment strategies.
Collapse
Affiliation(s)
- Ann Katrin Sauer
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
- Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
| | - Simone Hagmeyer
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
| | - Andreas M. Grabrucker
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
- Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
- Correspondence: ; Tel.: +353-61-237756
| |
Collapse
|
20
|
Garí M, Grzesiak M, Krekora M, Kaczmarek P, Jankowska A, Król A, Kaleta D, Jerzyńska J, Janasik B, Kuraś R, Tartaglione AM, Calamandrei G, Hanke W, Polańska K. Prenatal exposure to neurotoxic metals and micronutrients and neurodevelopmental outcomes in early school age children from Poland. ENVIRONMENTAL RESEARCH 2022; 204:112049. [PMID: 34520749 DOI: 10.1016/j.envres.2021.112049] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
Exposure to environmental factors, such as neurotoxic metals and micronutrients, during critical periods of development can contribute to long-term consequences in offspring's health, including neurodevelopmental outcomes. The aim of this study was to evaluate the association between simultaneous prenatal exposure to metals [lead (Pb), cadmium (Cd), mercury (Hg)] and micronutrients [selenium (Se), zinc (Zn), copper (Cu)] and neurodevelopmental outcomes in school-age children from the Polish Mother and Child Cohort (REPRO_PL). Metals and micronutrients concentrations were measured in cord blood (Pb, Cd, Se, Zn, Cu) and in maternal hair (Hg) collected during the 3rd trimester of pregnancy. Behavioral and emotional problems, as well as children's cognitive and psychomotor development, were assessed in 436 school-age children using the Strengths and Difficulties Questionnaire (SDQ, filled in by the mothers) and the Polish adaptation of the Intelligence and Development Scales (IDS, administered by trained psychologists). Multivariate regression models were applied after imputation of missing values, using two approaches: (i) a joint analysis taking into account all metals and micronutrients simultaneously, and (ii) an ExWAS study (single-exposure model). In the SDQ, Hyperactivity/Inattention problems and Total difficulties were associated with higher Hg concentrations in maternal hair (0.18, 95% CI: 0.05; 0.3; and 0.14, 95% CI: 0.01; 0.3, respectively), whereas Emotional symptoms were inversely associated with Se and Zn levels in cord blood (-0.13, 95% CI: -0.3; 0.004; and -0.10, 95% CI: -0.2; 0.02, respectively). In the IDS, cord blood Pb levels were found to be negatively associated with Fluid and Crystallized IQ (-0.12, 95% CI: -0.3; 0.02; and -0.14, 95% CI: -0.3; 0.007, respectively) as well as Mathematical skills (-0.15, 95% CI: -0.3; 0.01). The current research has been able to simultaneously assess the exposure to various interacting chemicals during the prenatal period. We demonstrate that prenatal co-exposures to Pb, Hg, Zn and Se have long-term influences on the neuropsychological outcome of school-age children.
Collapse
Affiliation(s)
- Mercè Garí
- Institute of Computational Biology, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Mariusz Grzesiak
- Department of Perinatology, Obstetrics and Gynecology, "Polish Mother's Memorial Hospital" Research Institute, Lodz, Poland; Department of Gynecology and Obstetrics, IInd Chair of Gynecology and Obstetrics, Medical University of Lodz, Poland.
| | - Michał Krekora
- Department of Gynecology and Obstetrics, IInd Chair of Gynecology and Obstetrics, Medical University of Lodz, Poland; Department of Obstetrics and Gynecology, "Polish Mother's Memorial Hospital" Research Institute, Lodz, Poland.
| | - Piotr Kaczmarek
- Department of Perinatology, Obstetrics and Gynecology, "Polish Mother's Memorial Hospital" Research Institute, Lodz, Poland.
| | - Agnieszka Jankowska
- Department of Environmental and Occupational Health Hazards, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | - Anna Król
- Department of Environmental and Occupational Health Hazards, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | - Dorota Kaleta
- Department of Hygiene and Epidemiology, Medical University of Lodz, Poland.
| | - Joanna Jerzyńska
- Department of Paediatrics and Allergy, Copernicus Memorial Hospital, Medical University of Lodz, Poland.
| | - Beata Janasik
- Department of Biological and Environmental Monitoring, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | - Renata Kuraś
- Department of Biological and Environmental Monitoring, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | - Anna Maria Tartaglione
- Centre for Behavioural Sciences and Mental Health, National Institute of Health, Rome, Italy.
| | - Gemma Calamandrei
- Centre for Behavioural Sciences and Mental Health, National Institute of Health, Rome, Italy.
| | - Wojciech Hanke
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | - Kinga Polańska
- Department of Environmental and Occupational Health Hazards, Nofer Institute of Occupational Medicine, Lodz, Poland; Department of Hygiene and Epidemiology, Medical University of Lodz, Poland.
| |
Collapse
|
21
|
Sandoval KC, Thackray SE, Wong A, Niewinski N, Chipak C, Rehal S, Dyck RH. Lack of Vesicular Zinc Does Not Affect the Behavioral Phenotype of Polyinosinic:Polycytidylic Acid-Induced Maternal Immune Activation Mice. Front Behav Neurosci 2022; 16:769322. [PMID: 35273483 PMCID: PMC8902171 DOI: 10.3389/fnbeh.2022.769322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Zinc is important in neural and synaptic development and neuronal transmission. Within the brain, zinc transporter 3 (ZnT3) is essential for zinc uptake into vesicles. Loss of vesicular zinc has been shown to produce neurodevelopmental disorder (NDD)-like behavior, such as decreased social interaction and increased anxiety- and repetitive-like behavior. Maternal immune activation (MIA) has been identified as an environmental factor for NDDs, such as autism spectrum disorders (ASDs) and schizophrenia (SZ), in offspring, which occurs during pregnancy when the mother’s immune system reacts to the exposure to viruses or infectious diseases. In this study, we investigated the interaction effect of a genetic factor [ZnT3 knockout (KO) mice] and an environmental factor (MIA). We induced MIA in pregnant female (dams) mice during mid-gestation, using polyinosinic:polycytidylic acid (polyI:C), which mimics a viral infection. Male and female ZnT3 KO and wild-type (WT) offspring were tested in five behavioral paradigms: Ultrasonic Vocalizations (USVs) at postnatal day 9 (P9), Open Field Test, Marble Burying Test, three-Chamber Social Test, and Pre-pulse Inhibition (PPI) in adulthood (P60–75). Our results indicate that loss of vesicular zinc does not result in enhanced ASD- and SZ-like phenotype compared to WT, nor does it show a more pronounced phenotype in male ZnT3 KO compared to female ZnT3 KO. Finally, MIA offspring demonstrated an ASD- and SZ-like phenotype only in specific behavioral tests: increased calls emitted in USVs and fewer marbles buried. Our results suggest that there is no interaction between the loss of vesicular zinc and MIA induction in the susceptibility to developing an ASD- and SZ-like phenotype.
Collapse
Affiliation(s)
- Katy Celina Sandoval
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Sarah E. Thackray
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Alison Wong
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Nicole Niewinski
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Colten Chipak
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Suhkjinder Rehal
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Richard H. Dyck
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
- *Correspondence: Richard H. Dyck,
| |
Collapse
|
22
|
Utilizing Genomically Targeted Molecular Data to Improve Patient-Specific Outcomes in Autism Spectrum Disorder. Int J Mol Sci 2022; 23:ijms23042167. [PMID: 35216282 PMCID: PMC8879068 DOI: 10.3390/ijms23042167] [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: 12/31/2021] [Revised: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
Molecular biology combined with genomics can be a powerful tool for developing potential intervention strategies for improving outcomes in children with autism spectrum disorders (ASD). Monogenic etiologies rarely cause autism. Instead, ASD is more frequently due to many polygenic contributing factors interacting with each other, combined with the epigenetic effects of diet, lifestyle, and environment. One limitation of genomics has been identifying ways of responding to each identified gene variant to translate the information to something clinically useful. This paper will illustrate how understanding the function of a gene and the effects of a reported variant on a molecular level can be used to develop actionable and targeted potential interventions for a gene variant or combinations of variants. For illustrative purposes, this communication highlights a specific genomic variant, SHANK3. The steps involved in developing molecularly genomically targeted actionable interventions will be demonstrated. Cases will be shared to support the efficacy of this strategy and to show how clinicians utilized these targeted interventions to improve ASD-related symptoms significantly. The presented approach demonstrates the utility of genomics as a part of clinical decision-making.
Collapse
|
23
|
Stanton JE, Malijauskaite S, McGourty K, Grabrucker AM. The Metallome as a Link Between the "Omes" in Autism Spectrum Disorders. Front Mol Neurosci 2021; 14:695873. [PMID: 34290588 PMCID: PMC8289253 DOI: 10.3389/fnmol.2021.695873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/14/2021] [Indexed: 12/26/2022] Open
Abstract
Metal dyshomeostasis plays a significant role in various neurological diseases such as Alzheimer's disease, Parkinson's disease, Autism Spectrum Disorders (ASD), and many more. Like studies investigating the proteome, transcriptome, epigenome, microbiome, etc., for years, metallomics studies have focused on data from their domain, i.e., trace metal composition, only. Still, few have considered the links between other "omes," which may together result in an individual's specific pathologies. In particular, ASD have been reported to have multitudes of possible causal effects. Metallomics data focusing on metal deficiencies and dyshomeostasis can be linked to functions of metalloenzymes, metal transporters, and transcription factors, thus affecting the proteome and transcriptome. Furthermore, recent studies in ASD have emphasized the gut-brain axis, with alterations in the microbiome being linked to changes in the metabolome and inflammatory processes. However, the microbiome and other "omes" are heavily influenced by the metallome. Thus, here, we will summarize the known implications of a changed metallome for other "omes" in the body in the context of "omics" studies in ASD. We will highlight possible connections and propose a model that may explain the so far independently reported pathologies in ASD.
Collapse
Affiliation(s)
- Janelle E Stanton
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland
| | - Sigita Malijauskaite
- Bernal Institute, University of Limerick, Limerick, Ireland.,Department of Chemical Sciences, University of Limerick, Limerick, Ireland
| | - Kieran McGourty
- Bernal Institute, University of Limerick, Limerick, Ireland.,Department of Chemical Sciences, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
24
|
Zieminska E, Ruszczynska A, Augustyniak J, Toczylowska B, Lazarewicz JW. Zinc and Copper Brain Levels and Expression of Neurotransmitter Receptors in Two Rat ASD Models. Front Mol Neurosci 2021; 14:656740. [PMID: 34267627 PMCID: PMC8277171 DOI: 10.3389/fnmol.2021.656740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Zinc and copper are important trace elements necessary for the proper functioning of neurons. Impaired zinc and/or copper metabolism and signaling are implicated in many brain diseases, including autism (ASD). In our studies, autistic-like behavior in rat offsprings was induced by application to pregnant mothers valproic acid or thalidomide. Zinc and copper contents were measured in serum and brain structures: hippocampus, cerebral cortex, and cerebellum. Our research shows no interconnections in the particular metal concentrations measured in autistic animal brains and their sera. Based on patient researches, we studied 26 genes belonging to disturbed neurotransmitter pathways. In the same brain regions, we examined the expression of genes encoding proteins of cholinergic, adrenergic, serotonin, and dopamine receptors. In both rats’ ASD models, 17 out of the tested gene expression were decreased. In the cerebellum and cerebral cortex, expression of genes encoding cholinergic, adrenergic, and dopaminergic receptors decreased, whereas in the hippocampus only expression of serotoninergic receptors genes was downregulated. The changes in metals content observed in the rat brain can be secondary phenomena, perhaps elements of mechanisms that compensate for neurotransmission dysfunctions.
Collapse
Affiliation(s)
- Elzbieta Zieminska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Ruszczynska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Justyna Augustyniak
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Toczylowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Jerzy W Lazarewicz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
25
|
Zhang C, Maslar D, Minckley TF, LeJeune KD, Qin Y. Spontaneous, synchronous zinc spikes oscillate with neural excitability and calcium spikes in primary hippocampal neuron culture. J Neurochem 2021; 157:1838-1849. [PMID: 33638177 DOI: 10.1111/jnc.15334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/23/2021] [Indexed: 12/19/2022]
Abstract
Zinc has been suggested to act as an intracellular signaling molecule due to its regulatory effects on numerous protein targets including enzymes, transcription factors, ion channels, neurotrophic factors, and postsynaptic scaffolding proteins. However, intracellular zinc concentration is tightly maintained at steady levels under natural physiological conditions. Dynamic changes in intracellular zinc concentration have only been detected in certain types of cells that are exposed to pathologic stimuli or upon receptor ligand binding. Unlike calcium, the ubiquitous signaling metal ion that can oscillate periodically and spontaneously in various cells, spontaneous zinc oscillations have never been reported. In this work, we made the novel observation that the developing neurons generated spontaneous and synchronous zinc spikes in primary hippocampal cultures using a fluorescent zinc sensor, FluoZin-3. Blocking of glutamate receptor-dependent calcium influx depleted the zinc spikes, suggesting that these zinc spikes were driven by the glutamate-mediated spontaneous neural excitability and calcium spikes that have been characterized in early developing neurons. Simultaneous imaging of calcium or pH together with zinc, we uncovered that a downward pH spike was evoked with each zinc spike and this transient cellular acidification occurred downstream of calcium spikes but upstream of zinc spikes. Our results suggest that spontaneous, synchronous zinc spikes were generated through calcium influx-induced cellular acidification, which liberates zinc from intracellular zinc binding ligands. Given that changes in zinc concentration can modulate activities of proteins essential for synapse maturation and neuronal differentiation, these zinc spikes might act as important signaling roles in neuronal development.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Drew Maslar
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Taylor F Minckley
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kate D LeJeune
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Yan Qin
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| |
Collapse
|
26
|
Krall RF, Tzounopoulos T, Aizenman E. The Function and Regulation of Zinc in the Brain. Neuroscience 2021; 457:235-258. [PMID: 33460731 DOI: 10.1016/j.neuroscience.2021.01.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/31/2022]
Abstract
Nearly sixty years ago Fredrich Timm developed a histochemical technique that revealed a rich reserve of free zinc in distinct regions of the brain. Subsequent electron microscopy studies in Timm- stained brain tissue found that this "labile" pool of cellular zinc was highly concentrated at synaptic boutons, hinting a possible role for the metal in synaptic transmission. Although evidence for activity-dependent synaptic release of zinc would not be reported for another twenty years, these initial findings spurred decades of research into zinc's role in neuronal function and revealed a diverse array of signaling cascades triggered or regulated by the metal. Here, we delve into our current understanding of the many roles zinc plays in the brain, from influencing neurotransmission and sensory processing, to activating both pro-survival and pro-death neuronal signaling pathways. Moreover, we detail the many mechanisms that tightly regulate cellular zinc levels, including metal binding proteins and a large array of zinc transporters.
Collapse
Affiliation(s)
- Rebecca F Krall
- Department of Neurobiology, University of Pittsburgh School of Medicine, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, USA
| | - Thanos Tzounopoulos
- Department of Otolaryngology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA.
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, USA; Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, USA.
| |
Collapse
|
27
|
Imbalanced post- and extrasynaptic SHANK2A functions during development affect social behavior in SHANK2-mediated neuropsychiatric disorders. Mol Psychiatry 2021; 26:6482-6504. [PMID: 34021263 PMCID: PMC8760046 DOI: 10.1038/s41380-021-01140-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Mutations in SHANK genes play an undisputed role in neuropsychiatric disorders. Until now, research has focused on the postsynaptic function of SHANKs, and prominent postsynaptic alterations in glutamatergic signal transmission have been reported in Shank KO mouse models. Recent studies have also suggested a possible presynaptic function of SHANK proteins, but these remain poorly defined. In this study, we examined how SHANK2 can mediate electrophysiological, molecular, and behavioral effects by conditionally overexpressing either wild-type SHANK2A or the extrasynaptic SHANK2A(R462X) variant. SHANK2A overexpression affected pre- and postsynaptic targets and revealed a reversible, development-dependent autism spectrum disorder-like behavior. SHANK2A also mediated redistribution of Ca2+-permeable AMPA receptors between apical and basal hippocampal CA1 dendrites, leading to impaired synaptic plasticity in the basal dendrites. Moreover, SHANK2A overexpression reduced social interaction and increased the excitatory noise in the olfactory cortex during odor processing. In contrast, overexpression of the extrasynaptic SHANK2A(R462X) variant did not impair hippocampal synaptic plasticity, but still altered the expression of presynaptic/axonal signaling proteins. We also observed an attention-deficit/hyperactivity-like behavior and improved social interaction along with enhanced signal-to-noise ratio in cortical odor processing. Our results suggest that the disruption of pre- and postsynaptic SHANK2 functions caused by SHANK2 mutations has a strong impact on social behavior. These findings indicate that pre- and postsynaptic SHANK2 actions cooperate for normal neuronal function, and that an imbalance between these functions may lead to different neuropsychiatric disorders.
Collapse
|
28
|
Yasuda H, Tsutsui T, Suzuki K. Metallomics Analysis for Assessment of Toxic Metal Burdens in Infants/Children and Their Mothers: Early Assessment and Intervention Are Essential. Biomolecules 2020; 11:biom11010006. [PMID: 33374671 PMCID: PMC7822439 DOI: 10.3390/biom11010006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/19/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Accumulation of toxic metals in infants/children is of serious concern worldwide, from the viewpoint of their harmful effects on the normal growth and development. This metallomics study investigates the extent of toxic metal burdens in infants/children and the relationship to those in their mothers for 77 child/mother pair subjects. For mercury, its geometric mean concentration in infants/children was of similar level to that in their mothers, and a high-significant close correlation was observed between infants/children and their mothers (β = 0.758, r = 0.539, p < 0.0001). A significant but less intimate mother/child relationship was observed for arsenic (β = 0.301, r = 0.433), lead (β = 0.444, r = 0.471) and aluminum (β = 0.379, r = 0.451). Remarkably, the burden levels of lead, cadmium and aluminum in infants/children were approximately three times higher than those in their mothers (p < 0.0001), and the burden levels in some individuals were several tens of times higher than in the mothers. In contrast, some essential metal levels such as zinc, magnesium and calcium in infants/children were significantly lower than those in their mothers, and 29 individuals (37.7%) in the child subjects were estimated to be zinc-deficient. In addition, significant inverse correlations were observed between zinc and lead (r = −0.267, p = 0.019), and magnesium and arsenic (r = −0.514, p < 0.0001). These findings suggest that these toxic metal burdens and essential metal deficiencies in infants/children are of serious concern for their neurodevelopment, indicating that the early assessment and intervention are crucial. It is expected that larger epidemiological and intervention studies will provide a reasonable and essential pathway for intervention of neurodevelopment disorders.
Collapse
Affiliation(s)
- Hiroshi Yasuda
- La Belle Vie Research Laboratory, Tokyo 103-0006, Japan;
- Correspondence: (H.Y.); (K.S.)
| | | | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
- Correspondence: (H.Y.); (K.S.)
| |
Collapse
|
29
|
Popovitz J, Mysore SP, Adwanikar H. Neural Markers of Vulnerability to Anxiety Outcomes after Traumatic Brain Injury. J Neurotrauma 2020; 38:1006-1022. [PMID: 33050836 DOI: 10.1089/neu.2020.7320] [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] [Indexed: 11/12/2022] Open
Abstract
Anxiety outcomes after traumatic brain injury (TBI) are complex, and the underlying neural mechanisms are poorly understood. Here, we developed a multi-dimensional behavioral profiling approach to investigate anxiety-like outcomes in mice that takes into account individual variability. Departing from the tradition of comparing outcomes in TBI versus sham groups, we identified a subgroup within the TBI group that is vulnerable to anxiety dysfunction, and present increased exploration of the anxiogenic zone compared to sham controls or resilient injured animals, by applying dimensionality reduction, clustering, and post hoc validation to behavioral data obtained from multiple assays for anxiety at several post-injury time points. These vulnerable animals expressed distinct molecular profiles in the corticolimbic network, with downregulation in gamma-aminobutyric acid and glutamate and upregulation in neuropeptide Y markers. Indeed, among vulnerable animals, not resilient or sham controls, severity of anxiety-related outcomes correlated strongly with expression of molecular markers. Our results establish a foundational approach, with predictive power, for reliably identifying maladaptive anxiety outcomes after TBI and uncovering neural signatures of vulnerability to anxiety.
Collapse
Affiliation(s)
- Juliana Popovitz
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shreesh P Mysore
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hita Adwanikar
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Granzotto A, Canzoniero LMT, Sensi SL. A Neurotoxic Ménage-à-trois: Glutamate, Calcium, and Zinc in the Excitotoxic Cascade. Front Mol Neurosci 2020; 13:600089. [PMID: 33324162 PMCID: PMC7725690 DOI: 10.3389/fnmol.2020.600089] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Fifty years ago, the seminal work by John Olney provided the first evidence of the neurotoxic properties of the excitatory neurotransmitter glutamate. A process hereafter termed excitotoxicity. Since then, glutamate-driven neuronal death has been linked to several acute and chronic neurological conditions, like stroke, traumatic brain injury, Alzheimer’s, Parkinson’s, and Huntington’s diseases, and Amyotrophic Lateral Sclerosis. Mechanisms linked to the overactivation of glutamatergic receptors involve an aberrant cation influx, which produces the failure of the ionic neuronal milieu. In this context, zinc, the second most abundant metal ion in the brain, is a key but still somehow underappreciated player of the excitotoxic cascade. Zinc is an essential element for neuronal functioning, but when dysregulated acts as a potent neurotoxin. In this review, we discuss the ionic changes and downstream effects involved in the glutamate-driven neuronal loss, with a focus on the role exerted by zinc. Finally, we summarize our work on the fascinating distinct properties of NADPH-diaphorase neurons. This neuronal subpopulation is spared from excitotoxic insults and represents a powerful tool to understand mechanisms of resilience against excitotoxic processes.
Collapse
Affiliation(s)
- Alberto Granzotto
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States.,Center for Advanced Sciences and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences (DNISC), Laboratory of Molecular Neurology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | | | - Stefano L Sensi
- Center for Advanced Sciences and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences (DNISC), Laboratory of Molecular Neurology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
31
|
Krzywoszyńska K, Witkowska D, Świątek-Kozłowska J, Szebesczyk A, Kozłowski H. General Aspects of Metal Ions as Signaling Agents in Health and Disease. Biomolecules 2020; 10:biom10101417. [PMID: 33036384 PMCID: PMC7600656 DOI: 10.3390/biom10101417] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
This review focuses on the current knowledge on the involvement of metal ions in signaling processes within the cell, in both physiological and pathological conditions. The first section is devoted to the recent discoveries on magnesium and calcium-dependent signal transduction-the most recognized signaling agents among metals. The following sections then describe signaling pathways where zinc, copper, and iron play a key role. There are many systems in which changes in intra- and extra-cellular zinc and copper concentrations have been linked to important downstream events, especially in nervous signal transduction. Iron signaling is mostly related with its homeostasis. However, it is also involved in a recently discovered type of programmed cell death, ferroptosis. The important differences in metal ion signaling, and its disease-leading alterations, are also discussed.
Collapse
Affiliation(s)
- Karolina Krzywoszyńska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Correspondence: (K.K.); (D.W.); Tel.: +48-77-44-23-549 (K.K); +48-77-44-23-548 (D.W.)
| | - Danuta Witkowska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Correspondence: (K.K.); (D.W.); Tel.: +48-77-44-23-549 (K.K); +48-77-44-23-548 (D.W.)
| | - Jolanta Świątek-Kozłowska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
| | - Agnieszka Szebesczyk
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
| | - Henryk Kozłowski
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Faculty of Chemistry, University of Wrocław, 14 F. Joliot-Curie St., 50-383 Wrocław, Poland
| |
Collapse
|
32
|
Dolleman-van der Weel MJ, Witter MP. The thalamic midline nucleus reuniens: potential relevance for schizophrenia and epilepsy. Neurosci Biobehav Rev 2020; 119:422-439. [PMID: 33031816 DOI: 10.1016/j.neubiorev.2020.09.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 09/03/2020] [Accepted: 09/28/2020] [Indexed: 01/08/2023]
Abstract
Anatomical, electrophysiological and behavioral studies in rodents have shown that the thalamic midline nucleus reuniens (RE) is a crucial link in the communication between hippocampal formation (HIP, i.e., CA1, subiculum) and medial prefrontal cortex (mPFC), important structures for cognitive and executive functions. A common feature in neurodevelopmental and neurodegenerative brain diseases is a dysfunctional connectivity/communication between HIP and mPFC, and disturbances in the cognitive domain. Therefore, it is assumed that aberrant functioning of RE may contribute to behavioral/cognitive impairments in brain diseases characterized by cortico-thalamo-hippocampal circuit dysfunctions. In the human brain the connections of RE are largely unknown. Yet, recent studies have found important similarities in the functional connectivity of HIP-mPFC-RE in humans and rodents, making cautious extrapolating experimental findings from animal models to humans justifiable. The focus of this review is on a potential involvement of RE in schizophrenia and epilepsy.
Collapse
Affiliation(s)
- M J Dolleman-van der Weel
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU Norwegian University of Science and Technology, Trondheim NO-7491, Norway.
| | - M P Witter
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU Norwegian University of Science and Technology, Trondheim NO-7491, Norway.
| |
Collapse
|
33
|
Pratt EPS, Damon LJ, Anson KJ, Palmer AE. Tools and techniques for illuminating the cell biology of zinc. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118865. [PMID: 32980354 DOI: 10.1016/j.bbamcr.2020.118865] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Zinc (Zn2+) is an essential micronutrient that is required for a wide variety of cellular processes. Tools and methods have been instrumental in revealing the myriad roles of Zn2+ in cells. This review highlights recent developments fluorescent sensors to measure the labile Zn2+ pool, chelators to manipulate Zn2+ availability, and fluorescent tools and proteomics approaches for monitoring Zn2+-binding proteins in cells. Finally, we close with some highlights on the role of Zn2+ in regulating cell function and in cell signaling.
Collapse
Affiliation(s)
- Evan P S Pratt
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America
| | - Leah J Damon
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America
| | - Kelsie J Anson
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America
| | - Amy E Palmer
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO 80303, United States of America.
| |
Collapse
|
34
|
Kocyła A, Tran JB, Krężel A. Galvanization of Protein-Protein Interactions in a Dynamic Zinc Interactome. Trends Biochem Sci 2020; 46:64-79. [PMID: 32958327 DOI: 10.1016/j.tibs.2020.08.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/10/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
The presence of Zn2+ at protein-protein interfaces modulates complex function, stability, and introduces structural flexibility/complexity, chemical selectivity, and reversibility driven in a Zn2+-dependent manner. Recent studies have demonstrated that dynamically changing Zn2+ affects numerous cellular processes, including protein-protein communication and protein complex assembly. How Zn2+-involved protein-protein interactions (ZPPIs) are formed and dissociate and how their stability and reactivity are driven in a zinc interactome remain poorly understood, mostly due to experimental obstacles. Here, we review recent research advances on the role of Zn2+ in the formation of interprotein sites, their architecture, function, and stability. Moreover, we underline the importance of zinc networks in intersystemic communication and highlight bioinformatic and experimental challenges required for the identification and investigation of ZPPIs.
Collapse
Affiliation(s)
- Anna Kocyła
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Józef Ba Tran
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Artur Krężel
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland.
| |
Collapse
|
35
|
Disruption of zinc transporter ZnT3 transcriptional activity and synaptic vesicular zinc in the brain of Huntington's disease transgenic mouse. Cell Biosci 2020; 10:106. [PMID: 32944220 PMCID: PMC7488477 DOI: 10.1186/s13578-020-00459-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/05/2020] [Indexed: 12/27/2022] Open
Abstract
Background Huntington’s disease (HD) is a neurodegenerative disease that involves a complex combination of psychiatric, cognitive and motor impairments. Synaptic dysfunction has been implicated in HD pathogenesis. However, the mechanisms have not been clearly delineated. Synaptic vesicular zinc is closely linked to modulating synaptic transmission and maintaining cognitive ability. It is significant to assess zinc homeostasis for further revealing the pathogenesis of synaptic dysfunction and cognitive impairment in HD. Results Histochemical staining by autometallography indicated that synaptic vesicular zinc was decreased in the hippocampus, cortex and striatum of N171-82Q HD transgenic mice. Analyses by immunohistochemistry, Western blot and RT-PCR found that the expression of zinc transporter 3 (ZnT3) required for transport of zinc into synaptic vesicles was obviously reduced in these three brain regions of the HD mice aged from 14 to 20 weeks and BHK cells expressing mutant huntingtin. Significantly, dual-luciferase reporter gene and chromatin immunoprecipitation assays demonstrated that transcription factor Sp1 could activate ZnT3 transcription via its binding to the GC boxes in ZnT3 promoter. Moreover, mutant huntingtin was found to inhibit the binding of Sp1 to the promoter of ZnT3 and down-regulate ZnT3 expression, and the decline in ZnT3 expression could be ameliorated through overexpression of Sp1. Conclusions This is first study to reveal a significant loss of synaptic vesicular zinc and a decline in ZnT3 transcriptional activity in the HD transgenic mice. Our work sheds a novel mechanistic insight into pathogenesis of HD that mutant huntingtin down-regulates expression of ZnT3 through inhibiting binding of Sp1 to the promoter of ZnT3 gene, causing disruption of synaptic vesicular zinc homeostasis. Disrupted vesicular zinc ultimately leads to early synaptic dysfunction and cognitive deficits in HD. It is also suggested that maintaining normal synaptic vesicular zinc concentration is a potential therapeutic strategy for HD.
Collapse
|
36
|
Vyas Y, Lee K, Jung Y, Montgomery JM. Influence of maternal zinc supplementation on the development of autism-associated behavioural and synaptic deficits in offspring Shank3-knockout mice. Mol Brain 2020; 13:110. [PMID: 32758248 PMCID: PMC7409418 DOI: 10.1186/s13041-020-00650-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/30/2020] [Indexed: 11/10/2022] Open
Abstract
Autism Spectrum Disorders (ASD) are characterised by deficits in social interactions and repetitive behaviours. Multiple ASD-associated mutations have been identified in the Shank family of proteins that play a critical role in the structure and plasticity of glutamatergic synapses, leading to impaired synapse function and the presentation of ASD-associated behavioural deficits in mice. Shank proteins are highly regulated by zinc, where zinc binds the Shank SAM domain to drive synaptic protein recruitment and synaptic maturation. Here we have examined the influence of maternal dietary zinc supplementation during pregnancy and lactation on the development of ASD-associated behavioural and synaptic changes in the offspring Shank3 knockout (Shank3−/−) mice. Behavioural and electrophysiological experiments were performed in juvenile and adult Shank3−/− and wildtype littermate control mice born from mothers fed control (30 ppm, ppm) or supplemented (150 ppm) dietary zinc. We observed that the supplemented maternal zinc diet prevented ASD-associated deficits in social interaction and normalised anxiety behaviours in Shank3−/− offspring mice. These effects were maintained into adulthood. Repetitive grooming was also prevented in adult Shank3−/− offspring mice. At the synaptic level, maternal zinc supplementation altered postsynaptic NMDA receptor-mediated currents and presynaptic function at glutamatergic synapses onto medium spiny neurons in the cortico-striatal pathway of the Shank3−/− offspring mice. These data show that increased maternal dietary zinc during pregnancy and lactation can alter the development of ASD-associated changes at the synaptic and the behavioural levels, and that zinc supplementation from the beginning of brain development can prevent ASD-associated deficits in Shank3−/− mice long term.
Collapse
Affiliation(s)
- Yukti Vyas
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Kevin Lee
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Yewon Jung
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| |
Collapse
|
37
|
Breen MS, Browne A, Hoffman GE, Stathopoulos S, Brennand K, Buxbaum JD, Drapeau E. Transcriptional signatures of participant-derived neural progenitor cells and neurons implicate altered Wnt signaling in Phelan-McDermid syndrome and autism. Mol Autism 2020; 11:53. [PMID: 32560742 PMCID: PMC7304190 DOI: 10.1186/s13229-020-00355-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) is a rare genetic disorder with high risk of autism spectrum disorder (ASD), intellectual disability, and language delay, and is caused by 22q13.3 deletions or mutations in the SHANK3 gene. To date, the molecular and pathway changes resulting from SHANK3 haploinsufficiency in PMS remain poorly understood. Uncovering these mechanisms is critical for understanding pathobiology of PMS and, ultimately, for the development of new therapeutic interventions. METHODS We developed human-induced pluripotent stem cell (hiPSC)-based models of PMS by reprogramming peripheral blood samples from individuals with PMS (n = 7) and their unaffected siblings (n = 6). For each participant, up to three hiPSC clones were generated and differentiated into induced neural progenitor cells (hiPSC-NPCs; n = 39) and induced forebrain neurons (hiPSC-neurons; n = 41). Genome-wide RNA-sequencing was applied to explore transcriptional differences between PMS probands and unaffected siblings. RESULTS Transcriptome analyses identified 391 differentially expressed genes (DEGs) in hiPSC-NPCs and 82 DEGs in hiPSC-neurons, when comparing cells from PMS probands and unaffected siblings (FDR < 5%). Genes under-expressed in PMS were implicated in Wnt signaling, embryonic development, and protein translation, while over-expressed genes were enriched for pre- and postsynaptic density genes, regulation of synaptic plasticity, and G-protein-gated potassium channel activity. Gene co-expression network analysis identified two modules in hiPSC-neurons that were over-expressed in PMS, implicating postsynaptic signaling and GDP binding, and both modules harbored a significant enrichment of genetic risk loci for developmental delay and intellectual disability. Finally, PMS-associated genes were integrated with other ASD hiPSC transcriptome findings and several points of convergence were identified, indicating altered Wnt signaling and extracellular matrix. LIMITATIONS Given the rarity of the condition, we could not carry out experimental validation in independent biological samples. In addition, functional and morphological phenotypes caused by loss of SHANK3 were not characterized here. CONCLUSIONS This is the largest human neural sample analyzed in PMS. Genome-wide RNA-sequencing in hiPSC-derived neural cells from individuals with PMS revealed both shared and distinct transcriptional signatures across hiPSC-NPCs and hiPSC-neurons, including many genes implicated in risk for ASD, as well as specific neurobiological pathways, including the Wnt pathway.
Collapse
Affiliation(s)
- Michael S Breen
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew Browne
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sofia Stathopoulos
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Kristen Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - Elodie Drapeau
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
38
|
Balduit A, Mangogna A, Agostinis C, Zito G, Romano F, Ricci G, Bulla R. Zinc Oxide Exerts Anti-Inflammatory Properties on Human Placental Cells. Nutrients 2020; 12:nu12061822. [PMID: 32570911 PMCID: PMC7353449 DOI: 10.3390/nu12061822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/07/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Background: An aberrant and persistent inflammatory state at the fetal-maternal interface is considered as a key contributor in compromised pregnancies. Decidual endothelial cells (DECs) play a pivotal role in the control of the local decidual inflammation. The aim of the current study was to determine whether dietary supplement with zinc oxide (ZnO), due to its very low adverse effects, may be useful for modulating the inflammatory response in the first trimester of pregnancy. Methods: The anti-inflammatory properties of ZnO in pregnancy were evaluated by in vitro tests on endothelial cells isolated from normal deciduas and on a trophoblast cell line (HTR8/Svneo). The effects of this treatment were analyzed in terms of adhesion molecule expression and inflammatory cytokine secretion, by real time-quantitative PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Results: Our data showed that ZnO was able to reduce the inflammatory response of DECs, in terms of vascular cell adhesion molecule-1 (VCAM-1), interleukin (IL)-8, IL-6, tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) expression induced by TNF-α stimulation. This compound exerted no effect on intracellular adhesion molecule-1 (ICAM-1) exocytosis induced by TNF-α on stimulated trophoblast cells, but significantly reduced their IL-6 expression. Conclusion: According to these results, it can be suggested that the ZnO supplement, through its modulation of the pro-inflammatory response of DECs, can be used in pregnancy for the prevention of local decidual inflammation.
Collapse
Affiliation(s)
- Andrea Balduit
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (A.M.); (R.B.)
| | - Alessandro Mangogna
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (A.M.); (R.B.)
| | - Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34134 Trieste, Italy; (G.Z.); (F.R.); (G.R.)
- Correspondence: ; Tel.: +39-04055-88646
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34134 Trieste, Italy; (G.Z.); (F.R.); (G.R.)
| | - Federico Romano
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34134 Trieste, Italy; (G.Z.); (F.R.); (G.R.)
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34134 Trieste, Italy; (G.Z.); (F.R.); (G.R.)
- Department of Medical, Surgical and Health Science, University of Trieste, 34129 Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (A.B.); (A.M.); (R.B.)
| |
Collapse
|
39
|
Hippocampal Metabolite Profiles in Two Rat Models of Autism: NMR-Based Metabolomics Studies. Mol Neurobiol 2020; 57:3089-3105. [PMID: 32468248 PMCID: PMC7320041 DOI: 10.1007/s12035-020-01935-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/13/2020] [Indexed: 12/22/2022]
Abstract
Autism spectrum disorders (ASDs) are increasingly being diagnosed. Hypotheses link ASD to genetic, epigenetic, or environmental factors. The role of oxidative stress and the imbalance between excitatory and inhibitory neurotransmission in the pathogenesis of ASD has been suggested. Rats in which ASD symptoms are induced by valproate (VPA) or thalidomide (THAL) application in utero are useful models in ASD studies. Our study investigated whether rats in ASD models show changes in metabolite levels in the brain consistent with the hypothetical pathomechanisms of ASD. Female rats were fed one dose of 800 mg/kg VPA or 500 mg/kg THAL orally on the 11th day of gestation, and 1-month offspring were used for the experiments. Metabolic profiles from proton nuclear magnetic resonance spectroscopy of hydrophilic and hydrophobic extracts of rat hippocampi were subjected to OPLS-DA statistical analysis. Large differences between both models in the content of several metabolites in the rat hippocampus were noticed. The following metabolic pathways were identified as being disturbed in both ASD models: steroid hormone biosynthesis; fatty acid biosynthesis; the synthesis and degradation of ketone bodies; glycerophospholipid metabolism; cholesterol metabolism; purine metabolism; arginine and proline metabolism; valine, leucine, and isoleucine biosynthesis and degradation. These results indicate disorders of energy metabolism, altered structure of cell membranes, changes in neurotransmission, and the induction of oxidative stress in the hippocampus. Our data, consistent with hypotheses of ASD pathomechanisms, may be useful in future ASD studies, especially for the interpretation of the results of metabolomics analysis of body fluids in rat ASD models.
Collapse
|
40
|
Liu X, Zhong S, Li Z, Chen J, Wang Y, Lai S, Miao H, Jia Y. Serum copper and zinc levels correlate with biochemical metabolite ratios in the prefrontal cortex and lentiform nucleus of patients with major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109828. [PMID: 31778759 DOI: 10.1016/j.pnpbp.2019.109828] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 11/18/2019] [Accepted: 11/23/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Previous studies have demonstrated that copper and zinc metabolism are associated with the development of major depressive disorder (MDD). Abnormal copper and zinc levels may be related to neurotransmission and biochemical metabolism in the brains of MDD patients, especially in the prefrontal cortex (PFC) and lentiform nucleus (LN). However, the mechanism of how copper and zinc levels contribute to neural metabolism in MDD patients remains to be deciphered. This study aimed to correlate copper and zinc levels with biochemical metabolite ratios in the PFC and LN of MDD patients. METHOD Twenty-nine MDD patients and thirty-two healthy control (HC) volunteers were enrolled in this study. Proton magnetic resonance spectroscopy (1H-MRS) was used to determine the levels of the N-acetylaspartate (NAA), choline (Cho) and creatine (Cr) in the brain, and specifically in the PFC and LN regions. Serum copper and zinc levels were measured using atomic emission spectrometry (AES). Afterwards, copper and zinc levels were correlated with biochemical metabolite ratios in the PFC and LN regions of the brain. RESULTS Higher serum copper and lower serum zinc levels with higher copper/zinc ratios were observed in MDD patients. NAA/Cr ratios in the PFC of MDD patients were lower compared to HC volunteers. In MDD patients, serum copper levels were negatively correlated with NAA/Cr ratios in the right PFC and right LN, while copper/zinc ratios were negatively correlated with NAA/Cr ratios in the right LN. No significant differences in serum copper and zinc levels with NAA/Cr ratios in the left PFC and left LN were observed in MDD patients. CONCLUSION Our findings suggest that higher serum copper and lower serum zinc levels may contribute to neuronal impairment by affecting neuronal biochemical metabolite ratios in the right PFC and right LN of MDD patients. Abnormal copper and zinc levels may play an important role in the pathophysiology of MDD.
Collapse
Affiliation(s)
- Xuanjun Liu
- Department of Neurology, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuming Zhong
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhinan Li
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Psychiatry, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510631, China
| | | | - Ying Wang
- Medical Imaging Center of The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shunkai Lai
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | | | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
41
|
Qureshi F, Adams J, Coleman D, Quig D, Hahn J. Urinary Essential Elements of Young Children with Autism Spectrum Disorder and their Mothers. RESEARCH IN AUTISM SPECTRUM DISORDERS 2020; 72:101518. [PMID: 32382316 PMCID: PMC7205186 DOI: 10.1016/j.rasd.2020.101518] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
BACKGROUND Even though the cause of autism spectrum disorders (ASD) remains unknown, the current understanding points towards complex interactions between environmental and genetic factors. One important environmental factor to consider is intake of toxic and essential elements, and their role in metabolism. Essential elements have received considerably less attention in the literature than the presence of toxins in urine. METHOD The purpose of this investigation is to comprehensively assess the association between urinary element compositions of 28 mothers who had young children with ASD and 29 mothers who had young typically developing (TD) children, and in a subset of their children (21 with ASD and 26 TD). RESULTS The results show that there are significant differences between the ASD and TD children cohorts' concentrations for four specific elements (sulfur, phosphorous, molybdenum, and tin). Utilizing multivariate statistical techniques (Fisher's discriminant analysis and support vector machines), it was possible to distinguish the ASD from the TD children groups with an 81% accuracy after cross-validation utilizing the four significantly different elements. However, among the mother cohorts assessed, there were no significant differences between those that had children with ASD and those with TD children. There was a significant correlation of levels of phosphorus and sulfur in the children with ASD (r = 0.63, p = 3.0E-3) and in the TD children (r = 0.47, p = 0.02). CONCLUSIONS Notable differences were observed between the elemental concentration in urine of children with ASD and their TD peers. Analyzing cellular pathways related to these elements are promising areas of future research.
Collapse
Affiliation(s)
- Fatir Qureshi
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - James Adams
- School of Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| | - Devon Coleman
- School of Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| | - David Quig
- Doctor's Data, 3755 Illinois Avenue, St. Charles, IL United States
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
42
|
Sanford L, Palmer AE. Dissociated Hippocampal Neurons Exhibit Distinct Zn 2+ Dynamics in a Stimulation-Method-Dependent Manner. ACS Chem Neurosci 2020; 11:508-514. [PMID: 32013397 PMCID: PMC7251562 DOI: 10.1021/acschemneuro.0c00006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Ionic Zn2+ has increasingly been recognized as an important neurotransmitter and signaling ion in glutamatergic neuron pathways. Intracellular Zn2+ transiently increases as a result of neuronal excitation, and this Zn2+ signal is essential for neuron plasticity, but the source and regulation of the signal is still unclear. In this study, we rigorously quantified Zn2+, Ca2+, and pH dynamics in dissociated mouse hippocampal neurons stimulated with bath application of high KCl or glutamate. While both stimulation methods yielded Zn2+ signals, Ca2+ influx, and acidification, glutamate stimulation induced more sustained high intracellular Ca2+ and a larger increase in intracellular Zn2+. However, the stimulation-induced pH change was similar between conditions, indicating that a different cellular change is responsible for the stimulation-dependent difference in Zn2+ signal. This work provides the first robust quantification of Zn2+ dynamics in neurons using different methods of stimulation.
Collapse
Affiliation(s)
- Lynn Sanford
- Department of Biochemistry, BioFrontiers Institute , University of Colorado Boulder , Boulder , Colorado 80309 , United States
| | - Amy E Palmer
- Department of Biochemistry, BioFrontiers Institute , University of Colorado Boulder , Boulder , Colorado 80309 , United States
| |
Collapse
|
43
|
Bhandari R, Paliwal JK, Kuhad A. Neuropsychopathology of Autism Spectrum Disorder: Complex Interplay of Genetic, Epigenetic, and Environmental Factors. ADVANCES IN NEUROBIOLOGY 2020; 24:97-141. [PMID: 32006358 DOI: 10.1007/978-3-030-30402-7_4] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Autism spectrum disorder (ASD) is a complex heterogeneous consortium of pervasive development disorders (PDD) which ranges from atypical autism, autism, and Asperger syndrome affecting brain in the developmental stage. This debilitating neurodevelopmental disorder results in both core as well as associated symptoms. Core symptoms observed in autistic patients are lack of social interaction, pervasive, stereotyped, and restricted behavior while the associated symptoms include irritability, anxiety, aggression, and several comorbid disorders.ASD is a polygenic disorder and is multifactorial in origin. Copy number variations (CNVs) of several genes that regulate the synaptogenesis and signaling pathways are one of the major factors responsible for the pathogenesis of autism. The complex integration of various CNVs cause mutations in the genes which code for molecules involved in cell adhesion, voltage-gated ion-channels, scaffolding proteins as well as signaling pathways (PTEN and mTOR pathways). These mutated genes are responsible for affecting synaptic transmission by causing plasticity dysfunction responsible, in turn, for the expression of ASD.Epigenetic modifications affecting DNA transcription and various pre-natal and post-natal exposure to a variety of environmental factors are also precipitating factors for the occurrence of ASD. All of these together cause dysregulation of glutamatergic signaling as well as imbalance in excitatory: inhibitory pathways resulting in glial cell activation and release of inflammatory mediators responsible for the aberrant social behavior which is observed in autistic patients.In this chapter we review and provide insight into the intricate integration of various genetic, epigenetic, and environmental factors which play a major role in the pathogenesis of this disorder and the mechanistic approach behind this integration.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Jyoti K Paliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India.
| |
Collapse
|
44
|
Noninvasive Brain Stimulation Enhances Memory Acquisition and Is Associated with Synaptoneurosome Modification in the Rat Hippocampus. eNeuro 2019; 6:ENEURO.0311-19.2019. [PMID: 31699891 PMCID: PMC6900464 DOI: 10.1523/eneuro.0311-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/15/2019] [Accepted: 10/31/2019] [Indexed: 01/11/2023] Open
Abstract
Transcranial direct-current stimulation (tDCS) is a non-invasive brain stimulation approach previously shown to enhance memory acquisition, but more studies are needed to elucidate the underlying mechanisms. Here, we examined the effects of anodal tDCS (0.25 mA for 30 min) on the memory performance of male Sprague Dawley rats in the passive avoidance test (PAT) and the associated modifications to the hippocampal proteomes. Results indicate anodal tDCS applied before the acquisition period significantly enhanced memory performance in the PAT. Following PAT, synaptoneurosomes were biochemically purified from the hippocampi of tDCS-treated or sham-treated rats and individual protein abundances were determined by bottom-up liquid chromatography mass spectrometry analysis. Proteomic analysis identified 184 differentially expressed hippocampal proteins when comparing the sham to the tDCS before memory acquisition treatment group. Ingenuity pathway analysis (IPA) showed anodal tDCS before memory acquisition significantly enhanced pathways associated with memory, cognition, learning, transmission, neuritogenesis, and long-term potentiation (LTP). IPA identified significant upstream regulators including bdnf, shank3, and gsk3b. Protein-protein interaction (PPI) and protein sequence similarity (PSS) networks show that glutamate receptor pathways, ion channel activity, memory, learning, cognition, and long-term memory were significantly associated with anodal tDCS. Centrality measures from both networks identified key proteins including dlg, shank, grin, and gria that were significantly modified by tDCS applied before the acquisition period. Together, our results provide descriptive molecular evidence that anodal tDCS enhances memory performance in the PAT by modifying hippocampal synaptic plasticity related proteins.
Collapse
|
45
|
Wang M, Liu X, Hou Y, Zhang H, Kang J, Wang F, Zhao Y, Chen J, Liu X, Wang Y, Wu S. Decrease of GSK-3β Activity in the Anterior Cingulate Cortex of Shank3b -/- Mice Contributes to Synaptic and Social Deficiency. Front Cell Neurosci 2019; 13:447. [PMID: 31749684 PMCID: PMC6843030 DOI: 10.3389/fncel.2019.00447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/18/2019] [Indexed: 12/22/2022] Open
Abstract
Social deficiency is one of the core syndromes of autism spectrum disorders (ASD), for which the underlying developmental mechanism still remains elusive. Anterior cingulate cortex (ACC) plays a key role in integrating social information and regulating social behavior. Recent studies have indicated that synaptic dysfunction in ACC is essential for ASD social defects. In the present study, we investigated the development of synapses and the roles of glycogen synthase kinase 3β (GSK-3β), which mediates multiple synaptic signaling pathways in ACC by using Shank3b−/− mice (a widely used ASD mouse model). Our data revealed that Shank3b mutation abolished the social induced c-Fos expression in ACC. From 4 weeks post-birth, neurons in Shank3b−/− ACC exhibited an obvious decrease in spine density and stubby spines. The length and thickness of post-synaptic density (PSD), the expression of vesicular glutamate transporter 2 (vGlut2) and glutamate receptor 2 (GluR2), and the frequency of miniature excitatory post-synaptic currents (mEPSCs) were significantly reduced in Shank3b−/−ACC. Interestingly, the levels of phosphorylated GSK-3β (Ser9), which inhibits the activity of GSK-3β, decreased along the same time course as the levels of GluR2 increased in ACC during development. Shank3b mutation leads to a dramatic increase of pGSK-3β (Ser9), and decrease of pPSD95 (a substrate of GSK-3β) and GluR2. Local delivery of AAV expressing constitutively active GSK-3β restored the expression of GluR2, increased the spine density and the number of mature spines. More importantly, active GSK-3β significantly promoted the social activity of Shank3b−/− mice. These data, in together, indicate that decrease of GSK-3β activity in ACC may contribute to the synaptic and social defects of Shank3b−/− mice. Enhancing GSK-3β activity may be utilized to treat ASD in the future.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xinyan Liu
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yilin Hou
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.,Department of Military Psychology, Fourth Military Medical University, Xi'an, China
| | - Haifeng Zhang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Junjun Kang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fei Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Youyi Zhao
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research, Department of Anethesiology, Center for Dental Materials and Advanced Manufacture, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Jing Chen
- Department of Anatomy, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xufeng Liu
- Department of Military Psychology, Fourth Military Medical University, Xi'an, China
| | - Yazhou Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shengxi Wu
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
46
|
Yennawar M, White RS, Jensen FE. AMPA Receptor Dysregulation and Therapeutic Interventions in a Mouse Model of CDKL5 Deficiency Disorder. J Neurosci 2019; 39:4814-4828. [PMID: 30952813 PMCID: PMC6561688 DOI: 10.1523/jneurosci.2041-18.2019] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 03/13/2019] [Accepted: 03/27/2019] [Indexed: 01/28/2023] Open
Abstract
Pathogenic mutations in cyclin-dependent kinase-like 5 (CDKL5) result in CDKL5 deficiency disorder (CDD), a rare disease marked by early-life seizures, autistic behaviors, and intellectual disability. Although mouse models of CDD exhibit dendritic instability and alterations in synaptic scaffolding proteins, studies of glutamate receptor levels and function are limited. Here we used a novel mouse model of CDD, the Cdkl5R59X knock-in mouse (R59X), to investigate changes in synaptic glutamate receptor subunits and functional consequences. Male mice were used for all experiments to avoid the confounding effects of X-inactivation that would be present in female heterozygous mice. We showed that adult male R59X mice recapitulated the behavioral outcomes observed in other mouse models of CDD, including social deficits and memory and learning impairments, and exhibited decreased latency to seizure upon pentylenetetrazol administration. Furthermore, we observed a specific increase in GluA2-lacking α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid)-type glutamate receptors (AMPARs) in the adult R59X hippocampus, which is accompanied electrophysiologically by increased rectification ratio of AMPAR EPSCs and elevated early-phase long term potentiation (LTP). Finally, we showed that acute treatment with the GluA2-lacking AMPAR blocker IEM-1460 decreased AMPAR currents, and rescued social deficits, working memory impairments, and seizure behavior latency in R59X mice.SIGNIFICANCE STATEMENT CDKL5 deficiency disorder (CDD) is a rare disease marked by autistic-like behaviors, intellectual disability, and seizures. While synaptic dysfunction has been observed in mouse models of CDD, there is limited information on how synaptic alterations contribute to behavioral and functional changes in CDD. Here we reveal elevated hippocampal GluA2-lacking AMPAR expression in a novel mouse model of CDD that is accompanied by changes in synaptic AMPAR function and plasticity. We also show, for the first time, that acutely targeting GluA2-lacking AMPAR dysregulation rescues core synaptic and neurobehavioral deficits in CDD.
Collapse
MESH Headings
- Adult
- Animals
- Behavior, Animal
- Child, Preschool
- Disease Models, Animal
- Epileptic Syndromes/drug therapy
- Epileptic Syndromes/genetics
- Epileptic Syndromes/psychology
- Excitatory Postsynaptic Potentials/genetics
- Female
- Gene Knock-In Techniques
- Humans
- Learning Disabilities/genetics
- Learning Disabilities/psychology
- Male
- Memory Disorders/genetics
- Memory Disorders/psychology
- Mice
- Mice, Inbred C57BL
- Mice, Neurologic Mutants
- Mutation/genetics
- Protein Serine-Threonine Kinases/deficiency
- Protein Serine-Threonine Kinases/genetics
- Psychomotor Performance
- Receptors, AMPA/deficiency
- Receptors, AMPA/drug effects
- Receptors, AMPA/genetics
- Seizures/chemically induced
- Seizures/physiopathology
- Social Behavior
- Spasms, Infantile/drug therapy
- Spasms, Infantile/genetics
- Spasms, Infantile/psychology
Collapse
Affiliation(s)
| | - Rachel S White
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Frances E Jensen
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|