1
|
Mercado-Ayón E, Talgo E, Flatley L, Coulman J, Lynch DR. Neurochemical alterations in the cerebellum of Friedreich's Ataxia mouse models. Exp Neurol 2025; 386:115176. [PMID: 39904419 DOI: 10.1016/j.expneurol.2025.115176] [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: 12/13/2024] [Revised: 01/29/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025]
Abstract
Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disorder caused by frataxin deficiency. Neurological deficits remain the ubiquitous feature of FRDA and include progressive ataxia and dysarthria, both of which are controlled to a large degree by the cerebellum. The precise impact of frataxin deficiency on the cerebellum including Purkinje cells remains unclear. In the present work, we examined the biochemical and structural properties of the cerebellum and Purkinje cells in the doxycycline-inducible (FRDAkd) and the Knock-in/Knockout (KIKO) mouse models of FRDA. Acute systemic knockdown of frataxin in FRDAkd mice and chronic frataxin deficiency in KIKO leads to a significant decrease in levels of AMPA receptors, particularly GluR2, and an increase in glial glutamate transporters. Significant astroglial accumulation occurred in KIKO cerebellum but not in FRDAkd mice. Purkinje cell dendritic arbors in the molecular layer did not change compared to wildtype in either model. The Purkinje cell postsynaptic receptor NMDAR1 significantly decreased only in the FRDAkd cerebellum while other NMDA receptor subunits, largely found in non-Purkinje cells, did not change. Overall, we observed dysregulated levels of glutamate receptors and transporters in the KIKO and FRDAkd mice models of Friedreich ataxia, suggesting the importance of frataxin in maintaining Purkinje cells and cerebellar integrity along with synaptic properties. These results point to conserved but not identical synaptic features between the models that may represent markers or conceivably targets in human FRDA.
Collapse
Affiliation(s)
- Elizabeth Mercado-Ayón
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA.
| | - Ellarie Talgo
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Liam Flatley
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Jennifer Coulman
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA.
| | - David R Lynch
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Einenkel AM, Salameh A. Selective vulnerability of hippocampal CA1 and CA3 pyramidal cells: What are possible pathomechanisms and should more attention be paid to the CA3 region in future studies? J Neurosci Res 2024; 102:e25276. [PMID: 38284845 DOI: 10.1002/jnr.25276] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/22/2023] [Accepted: 10/29/2023] [Indexed: 01/30/2024]
Abstract
Transient ischemia and reperfusion selectively damage neurons in brain, with hippocampal pyramidal cells being particularly vulnerable. Even within hippocampus, heterogeneous susceptibility is evident, with higher vulnerability of CA1 versus CA3 neurons described for several decades. Therefore, numerous studies have focused exclusively on CA1. Pediatric cardiac surgery is increasingly focusing on studies of hippocampal structures, and a negative impact of cardiopulmonary bypass on the hippocampus cannot be denied. Recent studies show a shift in selective vulnerability from neurons of CA1 to CA3. This review shows that cell damage is increased in CA3, sometimes stronger than in CA1, depending on several factors (method, species, age, observation period). Despite a highly variable pattern, several markers illustrate greater damage to CA3 neurons than previously assumed. Nevertheless, the underlying cellular mechanisms have not been fully deciphered to date. The complexity is reflected in possible pathomechanisms discussed here, with numerous factors (NMDA, kainate and AMPA receptors, intrinsic oxidative stress potential and various radicals, AKT isoforms, differences in vascular architecture, ratio of pro- and anti-apoptotic Bcl-2 factors, vulnerability of interneurons, mitochondrial dysregulation) contributing to either enhanced CA1 or CA3 vulnerability. Furthermore, differences in expressed genome, proteome, metabolome, and transcriptome in CA1 and CA3 appear to influence differential behavior after damaging stimuli, thus metabolomics-, transcriptomics-, and proteomics-based analyses represent a viable option to identify pathways of selective vulnerability in hippocampal neurons. These results emphasize that future studies should focus on the CA3 field in addition to CA1, especially with regard to improving therapeutic strategies after ischemic/hypoxic brain injury.
Collapse
Affiliation(s)
- Anne-Marie Einenkel
- Clinic for Pediatric Cardiology, University of Leipzig, Heart Centre, Leipzig, Germany
| | - Aida Salameh
- Clinic for Pediatric Cardiology, University of Leipzig, Heart Centre, Leipzig, Germany
| |
Collapse
|
3
|
Song Q, Huang W, Ye W, Yan H, Wang L, Yang Y, Cheng X, Zhang W, Zheng J, He P, He Y, Fang D, Han X. Neuroprotective Effects of Estrogen Through BDNF-Transient Receptor Potential Channels 6 Signaling Pathway in the Hippocampus in a Rat Model of Perimenopausal Depression. Front Aging Neurosci 2022; 14:869274. [PMID: 35875795 PMCID: PMC9305198 DOI: 10.3389/fnagi.2022.869274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/07/2022] [Indexed: 12/18/2022] Open
Abstract
Estradiol (E2) has been proven to be effective in treating perimenopausal depression (PD); however, the downstream signaling pathways have not been fully elucidated. Transient receptor potential channels 6 (TRPC6) plays a vital role in promoting neuronal development and the formation of excitatory synapses. At present, we found that the serum levels of E2 and brain-derived neurotrophic factor (BDNF) declined significantly in the women with PD compared to perimenopausal women, which was accompanied by a clear reduction in TRPC6 levels. To further reveal the effects of TRPC6 on neuronal survival and excitability, the PD-like rat model was established by the total removal of left ovary and 80% removal of right ovary followed by 21 days of the chronic unpredictable mild stress. Intragastric administration of E2 (2 mg/kg), intraperitoneal injection of BDNF/TrB signaling pathway inhibitor (K252a, 100 μg/kg) and TRPC6 agonist (OAG, 0.6 mg/kg), and intracerebroventricular infusion of anti-BDNF antibody for blocking BDNF (0.5 μg/24 μl/rat) daily for 21 days were conducted. The levels of BDNF and TRPC6 in rat serum were lower in PD rats compared to the control rats; the depression-like behavior was induced, the neuronal death rate in the hippocampus increased, and the thickness of postsynaptic density (PSD) and the number of asymmetric synapses decreased significantly in the PD group. E2 treatment greatly upregulated the serum levels of BDNF and TRPC6, the neuronal excitability indicated by an elevation in the PSD thickness and the numbers of asymmetric synapses, and these actions were reversed by K252a; co-administration of TRPC6 agonist and K252a improved neuronal degeneration and increased the neuronal excitability induced in the E2-treated PD rats. K252a or anti-BDNF antibody inhibited the increased neuronal BDNF and TRPC6 expression in E2-treated PD rats; co-treatment of TRPC6 agonist and anti-BDNF antibody reduced neuronal death and increased the BDNF and TRPC6 expression in the hippocampal CA1 neurons in the E2-treated PD rats. These results suggest that the neuroprotective role of E2 in PD is closely related to enhance the activity of BDNF/TRPC6 pathway and is helpful to provide new prevention and strategies.
Collapse
|
4
|
Li T, Xu G, Yi J, Huang Y. Intraoperative Hypothermia Induces Vascular Dysfunction in the CA1 Region of Rat Hippocampus. Brain Sci 2022; 12:brainsci12060692. [PMID: 35741578 PMCID: PMC9221322 DOI: 10.3390/brainsci12060692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Intraoperative hypothermia is very common and leads to memory decline. The hippocampus is responsible for memory formation. As a functional core area, the cornu ammonis 1 (CA1) region of the hippocampus contains abundant blood vessels and is susceptible to ischemia. The aim of the study was to explore vascular function and neuronal state in the CA1 region of rats undergoing intraoperative hypothermia. The neuronal morphological change and activity-regulated cytoskeleton-associated protein (Arc) expression were evaluated by haematoxylin-eosin staining and immunofluorescence respectively. Histology and immunohistochemistry were used to assess vascular function. Results showed that intraoperative hypothermia inhibited the expression of vascular endothelial growth factor and endothelial nitric oxide synthase, and caused reactive oxygen species accumulation. Additionally, the phenotype of vascular smooth muscle cells was transformed from contractile to synthetic, showing a decrease in smooth muscle myosin heavy chain and an increase in osteopontin. Ultimately, vascular dysfunction caused neuronal pyknosis in the CA1 region and reduced memory-related Arc expression. In conclusion, neuronal disorder in the CA1 region was caused by intraoperative hypothermia-related vascular dysfunction. This study could provide a novel understanding of the effect of intraoperative hypothermia in the hippocampus, which might identify a new research target and treatment strategy.
Collapse
|
5
|
Influence of Topiramate on the Synaptic Endings of the Temporal Lobe Neocortex in an Experimental Model of Hyperthermia-Induced Seizures: An Ultrastructural Study. Brain Sci 2021; 11:brainsci11111433. [PMID: 34827435 PMCID: PMC8615765 DOI: 10.3390/brainsci11111433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 11/17/2022] Open
Abstract
The objective of this pioneering study was to assess potentially neuroprotective properties of topiramate (TPM), a broad spectrum and newer-generation antiepileptic used against damage to synaptic endings of the temporal lobe neocortex in experimental hyperthermia-induced seizures (HS). TPM (80 mg/kg b.m.) was administered in young male Wistar rats with an intragastric tube before and immediately after HS. Specimens (1 mm3) collected from the neocortex, fixed via transcardial perfusion with paraformaldehyde and glutaraldehyde solution, were routinely processed for transmission-electron microscopic study, i.e., for descriptive and morphometric analysis. The ultrastructure of neocortical neuropil components affected by hyperthermic stress showed distinct swelling of pre and post-synaptic axodendritic and axospinal endings, including total disintegration. Mitochondria were markedly damaged in synaptic structures. Axoplasm of presynaptic boutons contained a decreased number of synaptic vesicles. Synaptic junctions showed active zone-shortening. Preventive administration of TPM before HS induction demonstrated neuroprotective effects against synaptic damage in approximately 1/4 of these structures. Interestingly, beneficial effects on synapsis morphology were more common in perivascular zones close to well-preserved capillaries. They were demonstrated by smaller swelling of both presynaptic and postsynaptic parts, well-preserved mitochondria, an increased number and regular distribution of synaptic vesicles within axoplasm, and a significantly increased synaptic active zones. However, topiramate used directly after HS was ineffective in the prevention of hyperthermia-evoked synaptic injury. Our findings support the hypothesis that topiramate applied before HS can protect some neocortical synapses via the vascular factor by enhancing blood-brain barrier components and improving the blood supply of gray matter in the temporal lobe, which may be significant in febrile seizure-prevention in children.
Collapse
|
6
|
Park MTM, Jeon P, Khan AR, Dempster K, Chakravarty MM, Lerch JP, MacKinley M, Théberge J, Palaniyappan L. Hippocampal neuroanatomy in first episode psychosis: A putative role for glutamate and serotonin receptors. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110297. [PMID: 33691200 DOI: 10.1016/j.pnpbp.2021.110297] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/01/2021] [Accepted: 03/02/2021] [Indexed: 02/01/2023]
Abstract
Disrupted serotonergic and glutamatergic signaling interact and contribute to the pathophysiology of schizophrenia, which is particularly relevant for the hippocampus where diverse expression of serotonin receptors is noted. Hippocampal atrophy is a well-established feature of schizophrenia, with select subfields hypothesized as particularly vulnerable due to variation in glutamate receptor densities. We investigated hippocampal anomalies in first-episode psychosis (FEP) in relation to receptor distributions by leveraging 4 sources of data: (1) ultra high-field (7-Tesla) structural neuroimaging, and (2) proton magnetic resonance spectroscopy (1H-MRS) of glutamate from 27 healthy and 41 FEP subjects, (3) gene expression data from the Allen Human Brain Atlas and (4) atlases of the serotonin receptor system. Automated methods delineated the hippocampus to map receptor density across subfields. We used gene expression data to correlate serotonin and glutamate receptor genes across the hippocampus. Measures of individual hippocampal shape-receptor alignment were derived through normative modelling and correlations to receptor distributions, termed Receptor-Specific Morphometric Signatures (RSMS). We found reduced hippocampal volumes in FEP, while CA4-dentate gyrus showed greatest reductions. Gene expression indicated 5-HT1A and 5-HT4 to correlate with AMPA and NMDA expression, respectively. Magnitudes of subfield volumetric reduction in FEP correlated most with 5-HT1A (R = 0.64, p = 4.09E-03) and 5-HT4 (R = 0.54, p = 0.02) densities as expected, and replicated using previously published data from two FEP studies. Right-sided 5-HT4-RSMS was correlated with MRS glutamate (R = 0.357, p = 0.048). We demonstrate a putative glutamate-driven hippocampal variability in FEP through a serotonin receptor-density gated mechanism, thus outlining a mechanistic interplay between serotonin and glutamate in determining the hippocampal morphology in schizophrenia.
Collapse
Affiliation(s)
- Min Tae M Park
- Department of Psychiatry, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Peter Jeon
- Department of Medical Biophysics, Western University, London, Canada
| | - Ali R Khan
- Department of Medical Biophysics, Western University, London, Canada; Robarts Research Institute, Western University, London, Canada
| | - Kara Dempster
- Department of Psychiatry, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - M Mallar Chakravarty
- Departments of Psychiatry and Biological and Biomedical Engineering, McGill University, Montreal, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Canada
| | - Jason P Lerch
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Jean Théberge
- Department of Psychiatry, Schulich School of Medicine and Dentistry, Western University, London, Canada; Department of Medical Biophysics, Western University, London, Canada; Lawson Health Research Institute, London, Canada
| | - Lena Palaniyappan
- Department of Psychiatry, Schulich School of Medicine and Dentistry, Western University, London, Canada; Robarts Research Institute, Western University, London, Canada; Lawson Health Research Institute, London, Canada.
| |
Collapse
|
7
|
Zeng H, Han X, Zhu Z, Yu S, Mei S, Cheng X, Zhang W, Zhang G, Fang D. Increased uterine NLRP3 inflammasome and leucocyte infiltration in a rat model of preeclampsia. Am J Reprod Immunol 2021; 86:e13493. [PMID: 34375018 DOI: 10.1111/aji.13493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/09/2021] [Accepted: 08/04/2021] [Indexed: 11/30/2022] Open
Abstract
The disruption of the inflammatory microenvironment in the uterus affects pregnancy outcome. However, the exact quantification and distribution of leukocyte subpopulations in the uterus in preeclampsia (PE) have not been clearly characterized. Inflammasomes promote the release of proinflammatory cytokines interleukin (IL)-β and IL-18. A higher expression of NLRP3 inflammasome in placentas contributes to excessive inflammation in PE. However, related studies on the uterus are scarce. We aimed to investigate changes in the infiltration of leukocyte subpopulations in decidual and uterine tissues, and explore the role of activation of uterine NLRP3 inflammasomes in PE. Decidual tissues were collected from normotensive pregnant women and preeclamptic women. A PE-like model was established via administration of lipopolysaccharide to normal pregnant rats. Uterine and decidual tissues were collected from all experimental groups. It was found that the number of leukocytes was significantly elevated in decidual and uterine tissues in PE patients compared to normal controls. The leukocytes (predominantly macrophages and NK cells) particularly infiltrated into the decidua and uterine decidua in PE-like rats, and these were sparse in the myometrium. The NLRP3 immunoreactivity in the uterus was extremely little in control rats, its immunoreactivity and caspase-1 immunoreactivity were significantly elevated in the PE-like rats; the mRNA expression results also indicated an upward trend in the activation of NLRP3 inflammasomes. These results support that leucocyte infiltration in the decidua and uterine deciduas, and the activation of NLRP3 inflammasome in the uterus, which participate in the pathogenesis, are responsible for the excessive inflammation at the maternal-fetal interface during PE. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Huiqian Zeng
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Xinjia Han
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Zhiqin Zhu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Shengjun Yu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Shanshan Mei
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Xi Cheng
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Weiqiang Zhang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Guanglan Zhang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| | - Dajun Fang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University
| |
Collapse
|
8
|
VEGF Modulates the Neural Dynamics of Hippocampal Subregions in Chronic Global Cerebral Ischemia Rats. Neuromolecular Med 2021; 23:416-427. [PMID: 33398803 DOI: 10.1007/s12017-020-08642-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022]
Abstract
Theta and gamma rhythms in hippocampus are important to cognitive performance. The cognitive impairments following cerebral ischemia is linked with the dysfunction of theta and gamma oscillations. As the primary mechanism for learning and memory, synaptic plasticity is in connection with these neural oscillations. Although vascular endothelial growth factor (VEGF) is thought to protect synaptic function in the ischemia rats to relieve cognitive impairment, little has been done on its effect of neural dynamics with this process. The present study investigated whether the alternation of neural oscillations in the hippocampus of ischemia rats is one of the potential neuroprotective mechanisms of VEGF. Rats were treated with the intranasal administration of VEGF at 72 h following chronic global cerebral ischemia procedure. Then local field potentials (LFPs) in hippocampal CA1 and CA3 regions were recorded and analyzed. Our results showed that VEGF can improve the power of theta and gamma rhythms in CA1 region after ischemia. Chronic global cerebral ischemia reduced the theta-gamma phase-amplitude coupling (PAC) not only within CA1 area but also in the pathway from CA3 to CA1, while VEGF alleviated the decreased coupling strength. Despite these notable differences, there were no obvious changes in the PAC within CA3 region. Surprisingly, the ischemia state did not affect the phase-phase interaction of hippocampus. In conclusion, our findings demonstrated that VEGF enhanced the theta-gamma PAC strength of CA3-CA1 pathway in ischemia rats, which may futher improve the information transmission within the hippocampus. These results illustrated the potential electrophysiologic mechanism of VEGF on cognitive improvement.
Collapse
|
9
|
Evaluation of Altered Glutamatergic Activity in a Piglet Model of Hypoxic-Ischemic Brain Damage Using 1H-MRS. DISEASE MARKERS 2020; 2020:8850816. [PMID: 33029259 PMCID: PMC7532412 DOI: 10.1155/2020/8850816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/05/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
Abstract
Methods Twenty-five newborn piglets were selected and then randomly assigned to the control group (n = 5) and the model group (n = 20) subjected to HI. HI was induced by blocking bilateral carotid blood flow under simultaneous inhalation of a 6% oxygen mixture. 1H-MRS data were acquired from the basal ganglia at the following time points after HI: 6, 12, 24, and 72 h. Changes in protein levels of EAAT2 and GluR2 were determined by immunohistochemical analysis. Correlations among metabolite concentrations, metabolite ratios, and the protein levels of EAAT2 and GluR2 were investigated. Results The Glu level sharply increased after HI, reached a transient low level of depletion that approached the normal level in the control group, and subsequently increased again. Negative correlations were found between concentrations of Glu and EAAT2 protein levels (R s = -0.662, P < 0.001) and between the Glu/creatine (Cr) ratio and EAAT2 protein level (R s = -0.664, P < 0.001). Moreover, changes in GluR2 protein level were significantly and negatively correlated with those in Glu level (the absolute Glu concentration, R s = -0.797, P < 0.001; Glu/Cr, R s = -0.567, P = 0.003). Conclusions Changes in Glu level measured by 1H-MRS were inversely correlated with those in EAAT2 and GluR2 protein levels following HI, and the results demonstrated that 1H-MRS can reflect the early changes of glutamatergic activity in vivo.
Collapse
|
10
|
Zhang Y, Fu T, Han S, Ding Y, Wang J, Zheng J, Li J. Monocular Deprivation Affects Visual Cortex Plasticity Through cPKCγ-Modulated GluR1 Phosphorylation in Mice. Invest Ophthalmol Vis Sci 2020; 61:44. [PMID: 32343785 PMCID: PMC7401946 DOI: 10.1167/iovs.61.4.44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose To determine how visual cortex plasticity changes after monocular deprivation (MD) in mice and whether conventional protein kinase C gamma (cPKCγ) plays a role in visual cortex plasticity. Methods cPKCγ membrane translocation levels were quantified by using immunoblotting to explore the effects of MD on cPKCγ activation. Electrophysiology was used to record field excitatory postsynaptic potential (fEPSP) amplitude with the goal of observing changes in visual cortex plasticity after MD. Immunoblotting was also used to determine the phosphorylation levels of GluR1 at Ser831. Light transmission was analyzed using electroretinography to examine the effects of MD and cPKCγ on mouse retinal function. Results Membrane translocation levels of cPKCγ significantly increased in the contralateral visual cortex of MD mice compared to wild-type (WT) mice (P < 0.001). In the contralateral visual cortex, long-term potentiation (LTP) and the phosphorylation levels of GluR1 at Ser 831 were increased in cPKCγ+/+ mice after MD. Interestingly, these levels could be downregulated by cPKCγ knockout compared to cPKCγ+/++MD mice (P < 0.001). Compared to the right eyes of WT mice, the amplitudes of a-waves and b-waves declined in deprived right eyes of mice after MD (P < 0.001). There were no significant differences when comparing cPKCγ+/+ and cPKCγ−/− mice with MD. Conclusions cPKCγ participates in the plasticity of the visual cortex after MD, which is characterized by increased LTP in the contralateral visual cortex, which may be a result of cPKCγ-mediated phosphorylation of GluR1 at Ser 831.
Collapse
|
11
|
Han X, Zhou N, Hu H, Li X, Liu H. Nicotine Alleviates Cortical Neuronal Injury by Suppressing Neuroinflammation and Upregulating Neuronal PI3K-AKT Signaling in an Eclampsia-Like Seizure Model. Neurotox Res 2020; 38:665-681. [PMID: 32767216 DOI: 10.1007/s12640-020-00265-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
Our previous studies showed that treatment with alpha7 nicotinic acetylcholine receptor (α7nAChR) agonist nicotine could alleviate systemic inflammation and reduce neuronal loss in the hippocampus and seizure severity in eclampsia. In this study, we further investigated whether there is also neuronal damage in the cortex after eclamptic seizure, elucidated the potential mechanisms underlying the neuroprotective roles of nicotine in eclampsia. Retrospective analysis of MRI data of severe preeclampsia (SPE) patients was conducted. A preeclampsia model was established by lipopolysaccharide injection (PE group), and pentylenetetrazol was used to induce eclamptic seizure (E group). α7nAChR agonist nicotine and its antagonist (α-BGT) and PI3K inhibitor wortmannin were used for drug administration. Neuronal damage was detected by Nissl staining, and changes in neuroinflammation, neuronal apoptosis, α7nAChR expression, and PI3K-AKT signaling on cortical neurons were detected by immunohistochemistry and western blotting. MRI images showed that most abnormal signals from the brain of SPE patients were located in the cortex. The neuron survival ratio was lower in the cortex than in the hippocampus within the E group; such ratios in the cortex were significantly lower in the E and PE groups compared with those of the control group. Nicotine markedly decreased the production of inflammatory cytokines and microglial activation in the cortex of the E group. Moreover, nicotine increased p-AKT levels and decreased cleaved caspase-3 levels in cortical neurons. Treatment with α-BGT reversed effects of nicotine. Wortmannin also blocked the anti-neuronal apoptosis action of nicotine. Our results suggest that nicotine protects against neuronal injury in the cortex following eclampsia possibly by inhibiting neuroinflammation and activating neuronal PI3K-AKT pathway.
Collapse
Affiliation(s)
- Xinjia Han
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, China
| | - Ning Zhou
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huiping Hu
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, China
| | - Xin Li
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Hefei, China
| | - Huishu Liu
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, 510623, China.
| |
Collapse
|
12
|
Turovskaya MV, Gaidin SG, Vedunova MV, Babaev AA, Turovsky EA. BDNF Overexpression Enhances the Preconditioning Effect of Brief Episodes of Hypoxia, Promoting Survival of GABAergic Neurons. Neurosci Bull 2020; 36:733-760. [PMID: 32219700 PMCID: PMC7340710 DOI: 10.1007/s12264-020-00480-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/17/2019] [Indexed: 12/18/2022] Open
Abstract
Hypoxia causes depression of synaptic plasticity, hyperexcitation of neuronal networks, and the death of specific populations of neurons. However, brief episodes of hypoxia can promote the adaptation of cells. Hypoxic preconditioning is well manifested in glutamatergic neurons, while this adaptive mechanism is virtually suppressed in GABAergic neurons. Here, we show that brain-derived neurotrophic factor (BDNF) overexpression in neurons enhances the preconditioning effect of brief episodes of hypoxia. The amplitudes of the NMDAR- and AMPAR-mediated Ca2+ responses of glutamatergic and GABAergic neurons gradually decreased after repetitive brief hypoxia/reoxygenation cycles in cell cultures transduced with the (AAV)-Syn-BDNF-EGFP virus construct. In contrast, the amplitudes of the responses of GABAergic neurons increased in non-transduced cultures after preconditioning. The decrease of the amplitudes in GABAergic neurons indicated the activation of mechanisms of hypoxic preconditioning. Preconditioning suppressed apoptotic or necrotic cell death. This effect was most pronounced in cultures with BDNF overexpression. Knockdown of BDNF abolished the effect of preconditioning and promoted the death of GABAergic neurons. Moreover, the expression of the anti-apoptotic genes Stat3, Socs3, and Bcl-xl substantially increased 24 h after hypoxic episodes in the transduced cultures compared to controls. The expression of genes encoding the pro-inflammatory cytokines IL-10 and IL-6 also increased. In turn, the expression of pro-apoptotic (Bax, Casp-3, and Fas) and pro-inflammatory (IL-1β and TNFα) genes decreased after hypoxic episodes in cultures with BDNF overexpression. Inhibition of vesicular BDNF release abolished its protective action targeting inhibition of the oxygen-glucose deprivation (OGD)-induced [Ca2+]i increase in GABAergic and glutamatergic neurons, thus promoting their death. Bafilomycin A1, Brefeldin A, and tetanus toxin suppressed vesicular release (including BDNF) and shifted the gene expression profile towards excitotoxicity, inflammation, and apoptosis. These inhibitors of vesicular release abolished the protective effects of hypoxic preconditioning in glutamatergic neurons 24 h after hypoxia/reoxygenation cycles. This finding indicates a significant contribution of vesicular BDNF release to the development of the mechanisms of hypoxic preconditioning. Thus, our results demonstrate that BDNF plays a pivotal role in the activation and enhancement of the preconditioning effect of brief episodes of hypoxia and promotes tolerance of the most vulnerable populations of GABAergic neurons to hypoxia/ischemia.
Collapse
Affiliation(s)
- M V Turovskaya
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia
| | - S G Gaidin
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia
| | - M V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - A A Babaev
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - E A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia.
| |
Collapse
|
13
|
Wang C, Wei Y, Yuan Y, Yu Y, Xie K, Dong B, Shi Y, Wang G. The role of PI3K-mediated AMPA receptor changes in post-conditioning of propofol in brain protection. BMC Neurosci 2019; 20:51. [PMID: 31570094 PMCID: PMC6771103 DOI: 10.1186/s12868-019-0532-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We aimed to study the role of amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) glutamate receptor 2 (GluR2) subunit trafficking, and activity changes in short-term neuroprotection provided by propofol post-conditioning. We also aimed to determine the role of phosphoinositide-3-kinase (PI3K) in the regulation of these processes. METHODS Rats underwent 1 h of focal cerebral ischemia followed by 23 h of reperfusion were randomly divided into 6 groups (n = 36 per group): sham- operation (S), ischemia-reperfusion (IR), propofol (P group, propofol 20 mg/kg/h at the onset of reperfusion for 2 h after 60 min of occlusion), and LY294002 (PI3K non-selective antagonist) + sham (L + S, LY294002 of 1.5 mg/kg was infused 30 min before sham operation), LY294002+ ischemia-reperfusion (L + IR, LY294002 of 1.5 mg/kg was infused 30 min before middle cerebral artery occlusion), LY294002 + IR + propofol (L + P, LY294002 of 1.5 mg/kg was infused 30 min before middle cerebral artery occlusion and propofol 20 mg/kg/h at the onset of reperfusion for 2 h after 60 min of occlusion). RESULTS Compared with group IR, rats in group P had significant lower neurologic defect scores and infarct volume. Additionally, consistent with enhanced expression of PI3K-AMPAR GluR2 subunit complex substances in ipsilateral hippocampus, GluR2 subunits showed increased levels in both the plasma and postsynaptic membranes of neurons, while pGluR2 expression was reduced in group P. Furthermore, LY294002, the PI3K non-selective antagonist, blocked those effects. CONCLUSION These observations demonstrated that propofol post-conditioning revealed acute neuroprotective role against transient MCAO in rats. The short-term neuroprotective effect was contributed by enhanced GluR2 subunits trafficking to membrane and postsynaptic membranes of neurons, as well as down-regulated the expression of pGluR2 in damaged hippocampus. Finally, the above-mentioned protective mechanism might be contributed by increased combination of PI3K to AMPAR GluR2 subunit, thus maintained the expression and activation of AMPAR GluR2 in the ipsilateral hippocampus.
Collapse
Affiliation(s)
- Chenxu Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Ying Wei
- Department of Anesthesiology, Tianjin People’s Hospital, Tianjin Union Medical Center, Tianjin, 300191 China
| | - Yuan Yuan
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Beibei Dong
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Yuan Shi
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| |
Collapse
|
14
|
Sun Y, Feng X, Ding Y, Li M, Yao J, Wang L, Gao Z. Phased Treatment Strategies for Cerebral Ischemia Based on Glutamate Receptors. Front Cell Neurosci 2019; 13:168. [PMID: 31105534 PMCID: PMC6499003 DOI: 10.3389/fncel.2019.00168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/08/2019] [Indexed: 11/23/2022] Open
Abstract
Extracellular glutamate accumulation following cerebral ischemia leads to overactivation of glutamate receptors, thereby resulting in intracellular Ca2+ overload and excitotoxic neuronal injury. Multiple attempts have been made to counteract such effects by reducing glutamate receptor function, but none have been successful. In this minireview, we present the available evidence regarding the role of all types of ionotropic and metabotropic glutamate receptors in cerebral ischemia and propose phased treatment strategies based on glutamate receptors in both the acute and post-acute phases of cerebral ischemia, which may help realize the clinical application of glutamate receptor antagonists.
Collapse
Affiliation(s)
- Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, China.,Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Xue Feng
- Hebei University of Science and Technology, Shijiazhuang, China
| | - Yue Ding
- Shijiazhuang Vocational College of Technology and Information, Shijiazhuang, China
| | - Mengting Li
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, China
| | - Jun Yao
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, China
| | - Long Wang
- Department of Family and Consumer Sciences, California State University, Long Beach, CA, United States
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, China.,State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Shijiazhuang, China
| |
Collapse
|
15
|
Interaction of DCF1 with ATP1B1 induces impairment in astrocyte structural plasticity via the P38 signaling pathway. Exp Neurol 2018; 302:214-229. [DOI: 10.1016/j.expneurol.2018.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/16/2017] [Accepted: 01/08/2018] [Indexed: 12/18/2022]
|
16
|
Zaric M, Drakulic D, Stojanovic IG, Mitrovic N, Grkovic I, Martinovic J. Regional-specific effects of cerebral ischemia/reperfusion and dehydroepiandrosterone on synaptic NMDAR/PSD-95 complex in male Wistar rats. Brain Res 2018; 1688:73-80. [PMID: 29577884 DOI: 10.1016/j.brainres.2018.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 10/17/2022]
Abstract
Excessive glutamate efflux and N-methyl-D-aspartate receptor (NMDAR) over-activation represent well-known hallmarks of cerebral ischemia/reperfusion (I/R) injury, still, expression of proteins involved in this aspect of I/R pathophysiology show inconsistent data. Neurosteroid dehydroepiandrosterone (DHEA) has been proposed as potent NMDAR modulator, but its influence on I/R-induced changes up to date remains questionable. Therefore, I/R-governed alteration of vesicular glutamate transporter 1 (vGluT1), synaptic NMDAR subunit composition, postsynaptic density protein 95 (PSD-95) and neuronal morphology alone or following DHEA treatment were examined. For that purpose, adult male Wistar rats were treated with a single dose of vehicle or DHEA (20 mg/kg i.p.) 4 h following sham operation or 15 min bilateral common carotid artery occlusion. Western blot was used for analyses of synaptic protein expressions in hippocampus and prefrontal cortex, while neuronal morphology was assessed using Nissl staining. Regional-specific postischemic changes were detected on protein level i.e. signs of neuronal damage in CA1 area was accompanied with hippocampal vGluT1, NR1, NR2B enhancement and PSD-95 decrement, while histological changes observed in layer III were associated with decreased NR1 subunit in prefrontal cortex. Under physiological conditions DHEA had no effect on protein and histological appearance, while in ischemic milieu it restored hippocampal PSD-95 and NR1 in prefrontal cortex to the control level. Along with intact neurons, ones characterized by morphology observed in I/R group were also present. Future studies involving NMDAR-related intracellular signaling and immunohistochemical analysis will reveal precise effects of I/R and DHEA treatment in selected brain regions.
Collapse
Affiliation(s)
- Marina Zaric
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Dunja Drakulic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Ivana Gusevac Stojanovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Natasa Mitrovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Ivana Grkovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Jelena Martinovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
17
|
Shi Z, Zhu L, Li T, Tang X, Xiang Y, Han X, Xia L, Zeng L, Nie J, Huang Y, Tsang CK, Wang Y, Lei Z, Xu Z, So KF, Ruan Y. Neuroprotective Mechanisms of Lycium barbarum Polysaccharides Against Ischemic Insults by Regulating NR2B and NR2A Containing NMDA Receptor Signaling Pathways. Front Cell Neurosci 2017; 11:288. [PMID: 29021742 PMCID: PMC5623723 DOI: 10.3389/fncel.2017.00288] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/04/2017] [Indexed: 01/03/2023] Open
Abstract
Glutamate excitotoxicity plays an important role in neuronal death after ischemia. However, all clinical trials using glutamate receptor inhibitors have failed. This may be related to the evidence that activation of different subunit of NMDA receptor will induce different effects. Many studies have shown that activation of the intrasynaptic NR2A subunit will stimulate survival signaling pathways, whereas upregulation of extrasynaptic NR2B will trigger apoptotic pathways. A Lycium barbarum polysaccharide (LBP) is a mixed compound extracted from Lycium barbarum fruit. Recent studies have shown that LBP protects neurons against ischemic injury by anti-oxidative effects. Here we first reported that the effect of LBP against ischemic injury can be achieved by regulating NR2B and NR2A signaling pathways. By in vivo study, we found LBP substantially reduced CA1 neurons from death after transient global ischemia and ameliorated memory deficit in ischemic rats. By in vitro study, we further confirmed that LBP increased the viability of primary cultured cortical neurons when exposed to oxygen-glucose deprivation (OGD) for 4 h. Importantly, we found that LBP antagonized increase in expression of major proteins in the NR2B signal pathway including NR2B, nNOS, Bcl-2-associated death promoter (BAD), cytochrome C (cytC) and cleaved caspase-3, and also reduced ROS level, calcium influx and mitochondrial permeability after 4 h OGD. In addition, LBP prevented the downregulation in the expression of NR2A, pAkt and pCREB, which are important cell survival pathway components. Furthermore, LBP attenuated the effects of a NR2B co-agonist and NR2A inhibitor on cell mortality under OGD conditions. Taken together, our results demonstrated that LBP is neuroprotective against ischemic injury by its dual roles in activation of NR2A and inhibition of NR2B signaling pathways, which suggests that LBP may be a superior therapeutic candidate for targeting glutamate excitotoxicity for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zhongshan Shi
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| | - Lihui Zhu
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| | - Tingting Li
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xiaoya Tang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Yonghui Xiang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xinjia Han
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Luoxing Xia
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Ling Zeng
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Junhua Nie
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Yongxia Huang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Chi Kwan Tsang
- Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying Wang
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Zhigang Lei
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zaocheng Xu
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kwok-Fai So
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China.,Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Yiwen Ruan
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| |
Collapse
|