1
|
Hou Y, Zuo Y, Song S, Zhang T. Long-term variable photoperiod exposure impairs hippocampal synapse involving of the glutamate system and leads to memory deficits in male Wistar rats. Exp Neurol 2025; 387:115191. [PMID: 39971149 DOI: 10.1016/j.expneurol.2025.115191] [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: 12/25/2024] [Revised: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Excessive artificial light at night can induce the human circadian misalignment, potentially impairing memory consolidation and the rhythms of hippocampal clock genes. To investigate the impact of circadian misalignment on hippocampal function, we measured various field excitatory postsynaptic potentials (fEPSP) and golgi staining in the CA1 and dentate gyrus (DG) regions in Wistar rats. Our findings revealed that circadian misalignment resulted in a leftward shift in the input-output (I-O) curve within the CA1 region, decreased long-term potentiation (LTP), multi-time interval paired-pulse ratio (PPR), as well as dendritic spines and complexity across both CA1 and DG regions. Additionally, magnetic resonance spectroscopy (MRS) showed that circadian misalignment downregulated glutamate-related neurotransmitters (Glu + Gln) in the hippocampus, contributing to impaired synaptic function. Furthermore, disruptions to glutamate receptor subunits due to circadian misalignment led to reduced expression of AMPA receptor and NMDA receptor subunits in the hippocampus. In summary, our results suggest that memory impairments resulting from circadian misalignment are associated with diminished functionality within the glutamatergic system; this includes reductions in both Glx levels and availability of glutamate receptor subunits-key factors contributing to compromised synaptic function within the hippocampus.
Collapse
Affiliation(s)
- Yuanyuan Hou
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yao Zuo
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, 550004, China
| | - Shaofei Song
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, 610041, China
| | - Tong Zhang
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China; School of Rehabilitation, Capital Medical University, Beijing 100068, China.
| |
Collapse
|
2
|
Cuello AC, Giacobini E. A brief history of the CNS cholinergic transmission. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:1-10. [PMID: 40340055 DOI: 10.1016/b978-0-443-19088-9.00011-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
This chapter provides a concise narrative of the evolution of events that have led to the current widely accepted concept of cholinergic neurotransmission. It recounts the saga of a century of intense research, technologic innovations, and fundamental discoveries, highlighting the contributions of key figures who have provoked ongoing intellectual challenges and scientific disputes regarding the physiologic significance of cholinergic transmission. Additionally, the chapter describes the resolution of decades-old debates surrounding the hypothesis of electrical vs chemical neurotransmission-a debate that has shaped current fundamental concepts and garnered several Nobel Prizes.
Collapse
Affiliation(s)
- A Claudio Cuello
- Department of Pharmacology & Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada; Department of Pharmacology, Oxford University, Oxford, United Kingdom.
| | - Ezio Giacobini
- Department of Internal Medicine, Readaptation and Geriatrics, Faculty of Medicine, University of Geneva Hospitals, University of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
O'Shea MJ, Anversa RG, Ch'ng SS, Campbell EJ, Walker LC, Andrews ZB, Lawrence AJ, Brown RM. An Island of Reil excitation: Mapping glutamatergic (vGlut1+ and vGlut2+) connections in the medial insular cortex. Biochem Pharmacol 2024; 230:116637. [PMID: 39561925 DOI: 10.1016/j.bcp.2024.116637] [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: 05/06/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
The insular cortex is a multifunctional and richly connected region of the cerebral cortex, critical in the neural integration of external stimuli and internal signals. Well-served for this role by a large network of afferent and efferent connections, the mouse insula can be simplified into an anterior, medial and posterior portion. Here we focus on the medial subregion, a once over-looked area that has gained recent attention for its involvement in an array of behaviours. Although the connections of medial insular cortex neurons have been previously identified, their precise glutamatergic phenotype remains undefined (typically defined by the presence of the subtype of vesicular glutamate transporters). Hence, we combined Cre knock-in mouse lines and adeno-associated viral tracing to distinguish between the expression of the two major vesicular glutamate transporters, type 1 (vGlut1) and 2 (vGlut2), in the subregion's neuronal inputs and outputs. Our results determined that the medial insula has extensive glutamatergic efferents expressing both vGlut1 and vGlut2 throughout the neuraxis. In contrast, a more conservative number of glutamatergic inputs were observed, with exclusively vGlut2+ projections received from hypothalamic and thalamic regions. Taken together, we demonstrate that vGlut1- and vGlut2-expressing networks of this insular subdivision have distinct connectivity patterns, including a greater abundance of vGlut1+ fibres innervating hypothalamic regions and the extended amygdala. These findings provide insight into the distinct chemo-architecture of this region, which may facilitate further investigation into the role of the medial insula in complex behaviour.
Collapse
Affiliation(s)
- Mia Jessica O'Shea
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia
| | - Roberta Goncalves Anversa
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Sarah Sulaiman Ch'ng
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Erin Jane Campbell
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia; School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Brain Neuromodulation Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Leigh Clasina Walker
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Zane Bruce Andrews
- Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Andrew John Lawrence
- Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Robyn Mary Brown
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, VIC, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
4
|
Baytas O, Davidson SM, Kauer JA, Morrow EM. Loss of mitochondrial enzyme GPT2 leads to reprogramming of synaptic glutamate metabolism. Mol Brain 2024; 17:87. [PMID: 39604975 PMCID: PMC11600823 DOI: 10.1186/s13041-024-01154-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Recessive loss-of-function mutations in the mitochondrial enzyme Glutamate Pyruvate Transaminase 2 (GPT2) cause intellectual disability in children. Given this cognitive disorder, and because glutamate metabolism is tightly regulated to sustain excitatory neurotransmission, here we investigate the role of GPT2 in synaptic function. GPT2 catalyzes a reversible reaction interconverting glutamate and pyruvate with alanine and alpha-ketoglutarate, a TCA cycle intermediate; thereby, GPT2 may play an important role in linking mitochondrial tricarboxylic acid (TCA) cycle with synaptic transmission. In mouse brain, we find that GPT2 is enriched in mitochondria of synaptosomes (isolated synaptic terminals). Loss of Gpt2 in mouse appears to lead to reprogramming of glutamate and glutamine metabolism, and to decreased glutamatergic synaptic transmission. Whole-cell patch-clamp recordings in pyramidal neurons of CA1 hippocampal slices from Gpt2-null mice reveal decreased excitatory post-synaptic currents (mEPSCs) without changes in mEPSC frequency, or importantly, changes in inhibitory post-synaptic currents (mIPSCs). Additional evidence of defective glutamate release included reduced levels of glutamate released from Gpt2-null synaptosomes measured biochemically. Glutamate release from synaptosomes was rescued to wild-type levels by alpha-ketoglutarate supplementation. Additionally, we observed evidence of altered metabolism in isolated Gpt2-null synaptosomes: decreased TCA cycle intermediates, and increased glutamate dehydrogenase activity. Notably, alterations in the TCA cycle and the glutamine pool were alleviated by alpha-ketoglutarate supplementation. In conclusion, our data support a model whereby GPT2 mitochondrial activity may contribute to glutamate availability in pre-synaptic terminals, thereby highlighting potential interactions between pre-synaptic mitochondrial metabolism and synaptic transmission.
Collapse
Affiliation(s)
- Ozan Baytas
- Department of Molecular Biology, Cell Biology and Biochemistry, Laboratories for Molecular Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, 02912, USA
- Neuroscience Graduate Program, Brown University, Providence, RI, 02912, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Shawn M Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08540, USA
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Julie A Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94035, USA
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Laboratories for Molecular Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA.
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
5
|
McIntyre RS, Jain R. Glutamatergic Modulators for Major Depression from Theory to Clinical Use. CNS Drugs 2024; 38:869-890. [PMID: 39150594 PMCID: PMC11486832 DOI: 10.1007/s40263-024-01114-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Major depressive disorder (MDD) is a chronic, burdensome, highly prevalent disease that is characterized by depressed mood and anhedonia. MDD is especially burdensome as approved monoamine antidepressant treatments have weeks-long delays before clinical benefit and low remission rates. In the past 2 decades, a promising target emerged to improve patient outcomes in depression treatment: glutamatergic signaling. This narrative review provides a high-level overview of glutamate signaling in synaptogenesis and neural plasticity and the implications of glutamate dysregulation in depression. Based on this preclinical evidence implicating glutamate in depression and the rapid improvement of depression with ketamine treatment in a proof-of-concept trial, a range of N-methyl-D-aspartate (NMDA)-targeted therapies have been investigated. While an array of treatments has been investigated in registered phase 2 or 3 clinical trials, the development of most of these agents has been discontinued. Multiple glutamate-targeted antidepressants are actively in development, and two are approved. Nasal administration of esketamine (Spravato®) was approved by the US Food and Drug Administration (FDA) in 2019 to treat adults with treatment-resistant depression and in 2020 for adults with MDD with acute suicidal ideation or behavior. Oral combination dextromethorphan-bupropion (AXS-05, Auvelity® extended-release tablet) was FDA approved in 2022 for the treatment of MDD in adults. These approvals bolster the importance of glutamate in depression and represent an exciting breakthrough in contemporary psychiatry, providing new avenues of treatment for patients as first-line therapy or with either poor response or unacceptable side effects to monoaminergic antidepressants.
Collapse
Affiliation(s)
- Roger S McIntyre
- University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Rakesh Jain
- Department of Psychiatry, Texas Tech University School of Medicine-Permian Basin, Midland, TX, USA.
- Texas Tech University School of Medicine-Permian Basin, 2500 W William Cannon Drive, Suite 505, Austin, Texas, 78745, USA.
| |
Collapse
|
6
|
Hamanaka Y, Hasebe M, Shiga S. Neural mechanism of circadian clock-based photoperiodism in insects and snails. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2024; 210:601-625. [PMID: 37596422 PMCID: PMC11226556 DOI: 10.1007/s00359-023-01662-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/20/2023]
Abstract
The photoperiodic mechanism distinguishes between long and short days, and the circadian clock system is involved in this process. Although the necessity of circadian clock genes for photoperiodic responses has been demonstrated in many species, how the clock system contributes to photoperiodic mechanisms remains unclear. A comprehensive study, including the functional analysis of relevant genes and physiology of their expressing cells, is necessary to understand the molecular and cellular mechanisms. Since Drosophila melanogaster exhibits a shallow photoperiodism, photoperiodic mechanisms have been studied in non-model species, starting with brain microsurgery and neuroanatomy, followed by genetic manipulation in some insects. Here, we review and discuss the involvement of the circadian clock in photoperiodic mechanisms in terms of neural networks in insects. We also review recent advances in the neural mechanisms underlying photoperiodic responses in insects and snails, and additionally circadian clock systems in snails, whose involvement in photoperiodism has hardly been addressed yet. Brain neurosecretory cells, insulin-like peptide/diuretic hormone44-expressing pars intercerebralis neurones in the bean bug Riptortus pedestris and caudo-dorsal cell hormone-expressing caudo-dorsal cells in the snail Lymnaea stagnalis, both promote egg laying under long days, and their electrical excitability is attenuated under short and medium days, which reduces oviposition. The photoperiodic responses of the pars intercerebralis neurones are mediated by glutamate under the control of the clock gene period. Thus, we are now able to assess the photoperiodic response by neurosecretory cell activity to investigate the upstream mechanisms, that is, the photoperiodic clock and counter.
Collapse
Affiliation(s)
- Yoshitaka Hamanaka
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Masaharu Hasebe
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Sakiko Shiga
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan.
| |
Collapse
|
7
|
Chowdhury KU, Holden ME, Wiley MT, Suppiramaniam V, Reed MN. Effects of Cannabis on Glutamatergic Neurotransmission: The Interplay between Cannabinoids and Glutamate. Cells 2024; 13:1130. [PMID: 38994982 PMCID: PMC11240741 DOI: 10.3390/cells13131130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
There has been a significant increase in the consumption of cannabis for both recreational and medicinal purposes in recent years, and its use can have long-term consequences on cognitive functions, including memory. Here, we review the immediate and long-term effects of cannabis and its derivatives on glutamatergic neurotransmission, with a focus on both the presynaptic and postsynaptic alterations. Several factors can influence cannabinoid-mediated changes in glutamatergic neurotransmission, including dosage, sex, age, and frequency of use. Acute exposure to cannabis typically inhibits glutamate release, whereas chronic use tends to increase glutamate release. Conversely, the postsynaptic alterations are more complicated than the presynaptic effects, as cannabis can affect the glutamate receptor expression and the downstream signaling of glutamate. All these effects ultimately influence cognitive functions, particularly memory. This review will cover the current research on glutamate-cannabis interactions, as well as the future directions of research needed to understand cannabis-related health effects and neurological and psychological aspects of cannabis use.
Collapse
Affiliation(s)
- Kawsar U. Chowdhury
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, GA 30144, USA
| | | | - Miles T. Wiley
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, GA 30144, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
8
|
Grønbæk-Thygesen M, Hartmann-Petersen R. Cellular and molecular mechanisms of aspartoacylase and its role in Canavan disease. Cell Biosci 2024; 14:45. [PMID: 38582917 PMCID: PMC10998430 DOI: 10.1186/s13578-024-01224-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/24/2024] [Indexed: 04/08/2024] Open
Abstract
Canavan disease is an autosomal recessive and lethal neurological disorder, characterized by the spongy degeneration of the white matter in the brain. The disease is caused by a deficiency of the cytosolic aspartoacylase (ASPA) enzyme, which catalyzes the hydrolysis of N-acetyl-aspartate (NAA), an abundant brain metabolite, into aspartate and acetate. On the physiological level, the mechanism of pathogenicity remains somewhat obscure, with multiple, not mutually exclusive, suggested hypotheses. At the molecular level, recent studies have shown that most disease linked ASPA gene variants lead to a structural destabilization and subsequent proteasomal degradation of the ASPA protein variants, and accordingly Canavan disease should in general be considered a protein misfolding disorder. Here, we comprehensively summarize the molecular and cell biology of ASPA, with a particular focus on disease-linked gene variants and the pathophysiology of Canavan disease. We highlight the importance of high-throughput technologies and computational prediction tools for making genotype-phenotype predictions as we await the results of ongoing trials with gene therapy for Canavan disease.
Collapse
Affiliation(s)
- Martin Grønbæk-Thygesen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| | - Rasmus Hartmann-Petersen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200N, Copenhagen, Denmark.
| |
Collapse
|
9
|
Witkin JM, Shafique H, Cerne R, Smith JL, Marini AM, Lipsky RH, Delery E. Mechanistic and therapeutic relationships of traumatic brain injury and γ-amino-butyric acid (GABA). Pharmacol Ther 2024; 256:108609. [PMID: 38369062 DOI: 10.1016/j.pharmthera.2024.108609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Traumatic brain injury (TBI) is a highly prevalent medical condition for which no medications specific for the prophylaxis or treatment of the condition as a whole exist. The spectrum of symptoms includes coma, headache, seizures, cognitive impairment, depression, and anxiety. Although it has been known for years that the inhibitory neurotransmitter γ-amino-butyric acid (GABA) is involved in TBI, no novel therapeutics based upon this mechanism have been introduced into clinical practice. We review the neuroanatomical, neurophysiological, neurochemical, and neuropharmacological relationships of GABA neurotransmission to TBI with a view toward new potential GABA-based medicines. The long-standing idea that excitatory and inhibitory (GABA and others) balances are disrupted by TBI is supported by the experimental data but has failed to invent novel methods of restoring this balance. The slow progress in advancing new treatments is due to the complexity of the disorder that encompasses multiple dynamically interacting biological processes including hemodynamic and metabolic systems, neurodegeneration and neurogenesis, major disruptions in neural networks and axons, frank brain lesions, and a multitude of symptoms that have differential neuronal and neurohormonal regulatory mechanisms. Although the current and ongoing clinical studies include GABAergic drugs, no novel GABA compounds are being explored. It is suggested that filling the gap in understanding the roles played by specific GABAA receptor configurations within specific neuronal circuits could help define new therapeutic approaches. Further research into the temporal and spatial delivery of GABA modulators should also be useful. Along with GABA modulation, research into the sequencing of GABA and non-GABA treatments will be needed.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | | | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA; Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA
| | - Ann M Marini
- Department of Neurology, Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert H Lipsky
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Elizabeth Delery
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.
| |
Collapse
|
10
|
Long Y, Zhao Z, Xie W, Shi J, Yang F, Zhu D, Jiang P, Tang Q, Ti Z, Jiang B, Yang X, Gao G, Qi W. Kallistatin leads to cognition impairment via downregulating glutamine synthetase. Pharmacol Res 2024; 202:107145. [PMID: 38492829 DOI: 10.1016/j.phrs.2024.107145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
In many neurodegenerative disorders, such as Alzheimer's disease (AD), glutamate-mediated neuronal excitotoxicity is considered the basis for cognitive impairment. The mRNA and protein expression of SERPINA4(Kallistatin) are higher in patients with AD. However, whether Kallistatin plays a regulatory role in glutamate-glutamine cycle homeostasis remains unclear. In this study, we identified impaired cognitive function in Kallistatin transgenic (KAL-TG) mice. Baseline glutamate levels were elevated and miniature excitatory postsynaptic current (mEPSC) frequency was increased in the hippocampus, suggesting the impairment of glutamate homeostasis in KAL-TG mice. Mechanistically, we demonstrated that Kallistatin promoted lysine acetylation and ubiquitination of glutamine synthetase (GS) and facilitated its degradation via the proteasome pathway, thereby downregulating GS. Fenofibrate improved cognitive memory in KAL-TG mice by downregulating serum Kallistatin. Collectively, our study findings provide insights the mechanism by which Kallistatin regulates cognitive impairment, and suggest the potential of fenofibrate to prevente and treat of AD patients with high levels of Kallistatin.
Collapse
Affiliation(s)
- Yanlan Long
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhen Zhao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wanting Xie
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinhui Shi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fengyu Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dan Zhu
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ping Jiang
- Department of Clinical Medical Laboratory, Guangzhou First People Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qilong Tang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhou Ti
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bin Jiang
- Guangdong Province Key Laboratory of Brain Function and Disease, School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong, China.
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products (Sun Yat-sen University), Guangzhou, China; Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, China.
| |
Collapse
|
11
|
Jacobs PS, Jee J, Fang L, Devlin E, Iannelli C, Thakuri D, Loughead J, Epperson CN, Wilson N, Roalf D, Reddy R, Nanga RPR. Application of glutamate weighted CEST in brain imaging of nicotine dependent participants in vivo at 7T. PLoS One 2024; 19:e0297310. [PMID: 38363747 PMCID: PMC10871471 DOI: 10.1371/journal.pone.0297310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 01/03/2024] [Indexed: 02/18/2024] Open
Abstract
INTRODUCTION With nicotine dependence being a significant healthcare issue worldwide there is a growing interest in developing novel therapies and diagnostic aids to assist in treating nicotine addiction. Glutamate (Glu) plays an important role in cognitive function regulation in a wide range of conditions including traumatic brain injury, aging, and addiction. Chemical exchange saturation transfer (CEST) imaging via ultra-high field MRI can image the exchange of certain saturated labile protons with the surrounding bulk water pool, making the technique a novel tool to investigate glutamate in the context of addiction. The aim of this work was to apply glutamate weighted CEST (GluCEST) imaging to study the dorsal anterior cingulate cortex (dACC) in a small population of smokers and non-smokers to determine its effectiveness as a biomarker of nicotine use. METHODS 2D GluCEST images were acquired on 20 healthy participants: 10 smokers (ages 29-50) and 10 non-smokers (ages 25-69), using a 7T MRI system. T1-weighted images were used to segment the GluCEST images into white and gray matter tissue and further into seven gray matter regions. Wilcoxon rank-sum tests were performed, comparing mean GluCEST contrast between smokers and non-smokers across brain regions. RESULTS GluCEST levels were similar between smokers and non-smokers; however, there was a moderate negative age dependence (R2 = 0.531) in smokers within the cingulate gyrus. CONCLUSION Feasibility of GluCEST imaging was demonstrated for in vivo investigation of smokers and non-smokers to assess glutamate contrast differences as a potential biomarker with a moderate negative age correlation in the cingulate gyrus suggesting reward network involvement.
Collapse
Affiliation(s)
- Paul S. Jacobs
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Joelle Jee
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Liu Fang
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Emily Devlin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Claudia Iannelli
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States of America
| | - Deepa Thakuri
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - James Loughead
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Cynthia Neill Epperson
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States of America
| | - Neil Wilson
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - David Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ravi Prakash Reddy Nanga
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
12
|
Rathod SS, Agrawal YO. Phytocannabinoids as Potential Multitargeting Neuroprotectants in Alzheimer's Disease. Curr Drug Res Rev 2024; 16:94-110. [PMID: 37132109 DOI: 10.2174/2589977515666230502104021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 05/04/2023]
Abstract
The Endocannabinoid System (ECS) is a well-studied system that influences a variety of physiological activities. It is evident that the ECS plays a significant role in metabolic activities and also has some neuroprotective properties. In this review, we emphasize several plant-derived cannabinoids such as β-caryophyllene (BCP), Cannabichromene (CBC), Cannabigerol (CBG), Cannabidiol (CBD), and Cannabinol (CBN), which are known to have distinctive modulation abilities of ECS. In Alzheimer's disease (AD), the activation of ECS may provide neuroprotection by modulating certain neuronal circuitry pathways through complex molecular cascades. The present article also discusses the implications of cannabinoid receptors (CB1 and CB2) as well as cannabinoid enzymes (FAAH and MAGL) modulators in AD. Specifically, CBR1 or CB2R modulations result in reduced inflammatory cytokines such as IL-2 and IL-6, as well as a reduction in microglial activation, which contribute to an inflammatory response in neurons. Furthermore, naturally occurring cannabinoid metabolic enzymes (FAAH and MAGL) inhibit the NLRP3 inflammasome complex, which may offer significant neuroprotection. In this review, we explored the multi-targeted neuroprotective properties of phytocannabinoids and their possible modulations, which could offer significant benefits in limiting AD.
Collapse
Affiliation(s)
- Sumit S Rathod
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, 425405, Maharashtra, India
- Shri Vile Parle Kelavani Mandal's, Institute of Pharmacy, Dhule, Dist. Dhule, 424001, Maharashtra, India
| | - Yogeeta O Agrawal
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, 425405, Maharashtra, India
| |
Collapse
|
13
|
Storm-Mathisen J, Johnston GAR. In memoriam: Frode Fonnum (1937-2023). J Neurochem 2023; 167:711-715. [PMID: 37859335 DOI: 10.1111/jnc.15973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 10/21/2023]
Abstract
Frode Fonnum died unexpectedly on 26th April 2023, at 86 years of age. He was a tower of strength-a primeval force-in neuroscience, neurochemistry and toxicology. His highly cited publications, comprised salient evidence for GABA and glutamate as brain neurotransmitters. He served as an expert, and as an organizer, including of European research cooperation and as President of the International Society for Neurochemistry (ISN). Photo credit: Per Kristian Opstad.
Collapse
Affiliation(s)
| | - Graham A R Johnston
- Sydney Pharmacy School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Silva ARR, Silva PV, Soares AR, González-Alcaraz MN, van Gestel CAM, Roelofs D, Moura G, Soares AMVM, Loureiro S. Daphnia magna Multigeneration Exposure to Carbendazim: Gene Transcription Responses. TOXICS 2023; 11:918. [PMID: 37999570 PMCID: PMC10674461 DOI: 10.3390/toxics11110918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023]
Abstract
The world population is experiencing colossal growth and thus demand for food, leading to an increase in the use of pesticides. Persistent pesticide contamination, such as carbendazim, remains a pressing environmental concern, with potentially long-term impacts on aquatic ecosystems. In the present study, Daphnia magna was exposed to carbendazim (5 µg L-1) for 12 generations, with the aim of assessing gene transcription alterations induced by carbendazim (using a D. magna custom microarray). The results showed that carbendazim caused changes in genes involved in the response to stress, DNA replication/repair, neurotransmission, ATP production, and lipid and carbohydrate metabolism at concentrations already found in the environment. These outcomes support the results of previous studies, in which carbendazim induced genotoxic effects and reproduction impairment (increasing the number of aborted eggs with the decreasing number of neonates produced). The exposure of daphnids to carbendazim did not cause a stable change in gene transcription between generations, with more genes being differentially expressed in the F0 generation than in the F12 generation. This could show some possible daphnid acclimation after 12 generations and is aligned with previous multigenerational studies where few ecotoxicological effects at the individual and populational levels and other subcellular level effects (e.g., biochemical biomarkers) were found.
Collapse
Affiliation(s)
- Ana Rita R. Silva
- Centre for Environmental and Marine Studies (CESAM) & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (P.V.S.); (A.M.V.M.S.); (S.L.)
| | - Patrícia V. Silva
- Centre for Environmental and Marine Studies (CESAM) & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (P.V.S.); (A.M.V.M.S.); (S.L.)
| | - Ana Raquel Soares
- Department of Medical Sciences & Institute for Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (A.R.S.); (G.M.)
| | - M. Nazaret González-Alcaraz
- Department of Agricultural Engineering of the E.T.S.I.A., Technical University of Cartagena, 30203 Cartagena, Spain;
| | - Cornelis A. M. van Gestel
- Amsterdam Institute for Life and Environment (A-LIFE), Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.A.M.v.G.); (D.R.)
| | - Dick Roelofs
- Amsterdam Institute for Life and Environment (A-LIFE), Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.A.M.v.G.); (D.R.)
- Keygene N.V., Agro Business Park 90, 6708 PW Wageningen, The Netherlands
| | - Gabriela Moura
- Department of Medical Sciences & Institute for Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal; (A.R.S.); (G.M.)
| | - Amadeu M. V. M. Soares
- Centre for Environmental and Marine Studies (CESAM) & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (P.V.S.); (A.M.V.M.S.); (S.L.)
| | - Susana Loureiro
- Centre for Environmental and Marine Studies (CESAM) & Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (P.V.S.); (A.M.V.M.S.); (S.L.)
| |
Collapse
|
15
|
Andrade López JM, Pani AM, Wu M, Gerhart J, Lowe CJ. Molecular characterization of nervous system organization in the hemichordate acorn worm Saccoglossus kowalevskii. PLoS Biol 2023; 21:e3002242. [PMID: 37725784 PMCID: PMC10508912 DOI: 10.1371/journal.pbio.3002242] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/11/2023] [Indexed: 09/21/2023] Open
Abstract
Hemichordates are an important group for investigating the evolution of bilaterian nervous systems. As the closest chordate outgroup with a bilaterally symmetric adult body plan, hemichordates are particularly informative for exploring the origins of chordates. Despite the importance of hemichordate neuroanatomy for testing hypotheses on deuterostome and chordate evolution, adult hemichordate nervous systems have not been comprehensively described using molecular techniques, and classic histological descriptions disagree on basic aspects of nervous system organization. A molecular description of hemichordate nervous system organization is important for both anatomical comparisons across phyla and for attempts to understand how conserved gene regulatory programs for ectodermal patterning relate to morphological evolution in deep time. Here, we describe the basic organization of the adult hemichordate Saccoglossus kowalevskii nervous system using immunofluorescence, in situ hybridization, and transgenic reporters to visualize neurons, neuropil, and key neuronal cell types. Consistent with previous descriptions, we found the S. kowalevskii nervous system consists of a pervasive nerve plexus concentrated in the anterior, along with nerve cords on both the dorsal and ventral side. Neuronal cell types exhibited clear anteroposterior and dorsoventral regionalization in multiple areas of the body. We observed spatially demarcated expression patterns for many genes involved in synthesis or transport of neurotransmitters and neuropeptides but did not observe clear distinctions between putatively centralized and decentralized portions of the nervous system. The plexus shows regionalized structure and is consistent with the proboscis base as a major site for information processing rather than the dorsal nerve cord. In the trunk, there is a clear division of cell types between the dorsal and ventral cords, suggesting differences in function. The absence of neural processes crossing the basement membrane into muscle and extensive axonal varicosities suggest that volume transmission may play an important role in neural function. These data now facilitate more informed neural comparisons between hemichordates and other groups, contributing to broader debates on the origins and evolution of bilaterian nervous systems.
Collapse
Affiliation(s)
- José M. Andrade López
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Ariel M. Pani
- Departments of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, Unites States of America
| | - Mike Wu
- Department of Molecular and Cell Biology, University of California, Berkeley, California, Unites States of America
| | - John Gerhart
- Department of Molecular and Cell Biology, University of California, Berkeley, California, Unites States of America
| | - Christopher J. Lowe
- Department of Biology, Stanford University, Stanford, California, United States of America
| |
Collapse
|
16
|
Rudd MT, Manley PJ, Hanney B, Meng Z, Shu Y, de Leon P, Frie JL, Han Y, Wai JMC, Yang ZQ, Perkins JJ, Hurzy DM, Manikowski JJ, Zhu H, Bungard CJ, Converso A, Meissner RS, Cosden ML, Hayashi I, Ma L, O’Brien J, Uebele VN, Schachter JB, Bhandari N, Ward GJ, Fillgrove KL, Lu B, Liang Y, Dubost DC, Puri V, Eddins DM, Vardigan JD, Drolet RE, Kern JT, Uslaner JM. Discovery of MK-8768, a Potent and Selective mGluR2 Negative Allosteric Modulator. ACS Med Chem Lett 2023; 14:1088-1094. [PMID: 37583812 PMCID: PMC10424309 DOI: 10.1021/acsmedchemlett.3c00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/29/2023] [Indexed: 08/17/2023] Open
Abstract
Glutamate plays a key role in cognition and mood, and it has been shown that inhibiting ionotropic glutamate receptors disrupts cognition, while enhancing ionotropic receptor activity is pro-cognitive. One approach to elevating glutamatergic tone has been to antagonize presynaptic metabotropic glutamate receptor 2 (mGluR2). A desire for selectivity over the largely homologous mGluR3 motivated a strategy to achieve selectivity through the identification of mGluR2 negative allosteric modulators (NAMs). Extensive screening and optimization efforts led to the identification of a novel series of 4-arylquinoline-2-carboxamides. This series was optimized for mGluR2 NAM potency, clean off-target activity, and desirable physical properties, which resulted in the identification of improved C4 and C7 substituents. The initial lead compound from this series was Ames-positive in a single strain with metabolic activation, indicating that a reactive metabolite was likely responsible for the genetic toxicity. Metabolic profiling and Ames assessment across multiple analogs identified key structure-activity relationships associated with Ames positivity. Further optimization led to the Ames-negative mGluR2 negative allosteric modulator MK-8768.
Collapse
Affiliation(s)
- Michael T. Rudd
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Peter J. Manley
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Barbara Hanney
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Zhaoyang Meng
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Youheng Shu
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Pablo de Leon
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Jessica L. Frie
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Yongxin Han
- External
Discovery Chemistry, Merck & Co., Inc, Boston, Massachusetts 02115, United States
| | - Jenny Miu-Chun Wai
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Zhi-Qiang Yang
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - James J. Perkins
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Danielle M. Hurzy
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Jesse J. Manikowski
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Hong Zhu
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Christopher J. Bungard
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Antonella Converso
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Robert S. Meissner
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Mali L. Cosden
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Ikuo Hayashi
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Lei Ma
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Julie O’Brien
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Victor N. Uebele
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Joel B. Schachter
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Neetesh Bhandari
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Gwendolyn J. Ward
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Kerry L. Fillgrove
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Bing Lu
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Yuexia Liang
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - David C. Dubost
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Vanita Puri
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Donnie M. Eddins
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Joshua D. Vardigan
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Robert E. Drolet
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Jonathan T. Kern
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Jason M. Uslaner
- Departments
of Discovery Chemistry, Neuroscience Biology Discovery, Pharmacology, Nonclinical Dug
Safety, Pharmacokinetics, Discovery Pharmaceutical Sciences, and In Vivo Pharmacology, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| |
Collapse
|
17
|
Tian L, Yu T. An integrated deep learning framework for the interpretation of untargeted metabolomics data. Brief Bioinform 2023; 24:bbad244. [PMID: 37369636 DOI: 10.1093/bib/bbad244] [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: 02/07/2023] [Revised: 06/02/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Untargeted metabolomics is gaining widespread applications. The key aspects of the data analysis include modeling complex activities of the metabolic network, selecting metabolites associated with clinical outcome and finding critical metabolic pathways to reveal biological mechanisms. One of the key roadblocks in data analysis is not well-addressed, which is the problem of matching uncertainty between data features and known metabolites. Given the limitations of the experimental technology, the identities of data features cannot be directly revealed in the data. The predominant approach for mapping features to metabolites is to match the mass-to-charge ratio (m/z) of data features to those derived from theoretical values of known metabolites. The relationship between features and metabolites is not one-to-one since some metabolites share molecular composition, and various adduct ions can be derived from the same metabolite. This matching uncertainty causes unreliable metabolite selection and functional analysis results. Here we introduce an integrated deep learning framework for metabolomics data that take matching uncertainty into consideration. The model is devised with a gradual sparsification neural network based on the known metabolic network and the annotation relationship between features and metabolites. This architecture characterizes metabolomics data and reflects the modular structure of biological system. Three goals can be achieved simultaneously without requiring much complex inference and additional assumptions: (1) evaluate metabolite importance, (2) infer feature-metabolite matching likelihood and (3) select disease sub-networks. When applied to a COVID metabolomics dataset and an aging mouse brain dataset, our method found metabolic sub-networks that were easily interpretable.
Collapse
Affiliation(s)
- Leqi Tian
- School of Data Science, The Chinese University of Hong Kong - Shenzhen, Guangdong, China
- Shenzhen Research Institute of Big Data, Guangdong, China
| | - Tianwei Yu
- School of Data Science, The Chinese University of Hong Kong - Shenzhen, Guangdong, China
- Shenzhen Research Institute of Big Data, Guangdong, China
- Guangdong Provincial Key Laboratory of Big Data Computing, Guangdong, China
| |
Collapse
|
18
|
Purushotham SS, Buskila Y. Astrocytic modulation of neuronal signalling. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1205544. [PMID: 37332623 PMCID: PMC10269688 DOI: 10.3389/fnetp.2023.1205544] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023]
Abstract
Neuronal signalling is a key element in neuronal communication and is essential for the proper functioning of the CNS. Astrocytes, the most prominent glia in the brain play a key role in modulating neuronal signalling at the molecular, synaptic, cellular, and network levels. Over the past few decades, our knowledge about astrocytes and their functioning has evolved from considering them as merely a brain glue that provides structural support to neurons, to key communication elements. Astrocytes can regulate the activity of neurons by controlling the concentrations of ions and neurotransmitters in the extracellular milieu, as well as releasing chemicals and gliotransmitters that modulate neuronal activity. The aim of this review is to summarise the main processes through which astrocytes are modulating brain function. We will systematically distinguish between direct and indirect pathways in which astrocytes affect neuronal signalling at all levels. Lastly, we will summarize pathological conditions that arise once these signalling pathways are impaired focusing on neurodegeneration.
Collapse
Affiliation(s)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- The MARCS Institute, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
19
|
Hsu SK, Lu CW, Chiu KM, Lee MY, Lin TY, Wang SJ. Mangiferin depresses vesicular glutamate release in synaptosomes from the rat cerebral cortex by decreasing synapsin I phosphorylation. Eur J Pharmacol 2023; 950:175772. [PMID: 37146708 DOI: 10.1016/j.ejphar.2023.175772] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
Mangiferin is a glucosyl xanthone that has been shown to be a neuroprotective agent against brain disorders involving excess glutamate. However, the effect of mangiferin on the function of the glutamatergic system has not been investigated. In this study, we used synaptosomes from the rat cerebral cortex to investigate the effect of mangiferin on glutamate release and identify the possible underlying mechanism. We observed that mangiferin produced a concentration-dependent reduction in the release of glutamate elicited by 4-aminopyridine with an IC50 value of 25 μM. Inhibition of glutamate release was blocked by removing extracellular calcium and by treatment with the vacuolar-type H+-ATPase inhibitor bafilomycin A1, which prevents the uptake and storage of glutamate in vesicles. Moreover, we showed that mangiferin decreased the 4-aminopyridine-elicited FM1-43 release and synaptotagmin 1 luminal domain antibody (syt1-L ab) uptake from synaptosomes, which correlated with decreased synaptic vesicle exocytosis. Transmission electron microscopy in synaptosomes also showed that mangiferin attenuated the 4-aminopyridine-elicited decrease in the number of synaptic vesicles. In addition, antagonism of Ca2+/calmodulin-dependent kinase II (CaMKII) and protein kinase A (PKA) counteracted mangiferin's effect on glutamate release. Mangiferin also decreased the phosphorylation of CaMKII, PKA, and synapsin I elicited by 4-aminopyridine treatment. Our data suggest that mangiferin reduces PKA and CaMKII activation and synapsin I phosphorylation, which could decrease synaptic vesicle availability and lead to a subsequent reduction in vesicular glutamate release from synaptosomes.
Collapse
Affiliation(s)
- Szu-Kai Hsu
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Department of Neurosurgery, Cathay General Hospital, Taipei, 106438, Taiwan
| | - Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Electrical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan
| | - Ming-Yi Lee
- Department of Medical Research, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, 22060, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan, 32003, Taiwan.
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, 33303, Taiwan.
| |
Collapse
|
20
|
Schmitt LI, David C, Steffen R, Hezel S, Roos A, Schara-Schmidt U, Kleinschnitz C, Leo M, Hagenacker T. Spinal astrocyte dysfunction drives motor neuron loss in late-onset spinal muscular atrophy. Acta Neuropathol 2023; 145:611-635. [PMID: 36930296 PMCID: PMC10119066 DOI: 10.1007/s00401-023-02554-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023]
Abstract
Spinal muscular atrophy (SMA) is a progressive neuromuscular disorder caused by a loss of the survival of motor neuron 1 (SMN1) gene, resulting in a loss of spinal motor neurons (MNs), leading to muscle weakness and wasting. The pathogenesis of MN loss in SMA and the selective vulnerability in different cellular populations are not fully understood. To investigate the role of spinal astrocytes in the pathogenesis of late-onset SMA, we used a mouse model in addition to in vitro approaches. Immunostaining, Western blot analysis, small interfering ribonucleic acid (siRNA) transfections, functional assays, enzyme-linked immunosorbent assay (ELISA), behavioral tests, and electrophysiological measurements were performed. Early activation of spinal astrocytes and a reduction of the excitatory amino acid transporter 1 (EAAT1) on postnatal day (P) 20 preceded the loss of spinal MNs in SMA mice occurring on P42. EAAT1 reduction resulted in elevated glutamate levels in the spinal cord of SMA mice at P20 and P42. SMA-like astrocytes generated by siRNA and an ex vivo model of glutamate excitotoxicity involving organotypic spinal cord slice cultures revealed the critical role of glutamate homeostasis in the degeneration of MNs. The pre-emptive administration of arundic acid (AA), as an inhibitor of astrocyte activation, to SMA mice prior to the loss of motor neurons (P28) resulted in elevated EAAT1 protein levels compared to vehicle-treated SMA mice and prevented the increase of glutamate in the spinal cord and the loss of spinal MNs. Furthermore, AA preserved motor functions during behavioral experiments, the electrophysiological properties, and muscle alteration of SMA mice. In a translational approach, we transfected healthy human fibroblasts with SMN1 siRNA, resulting in reduced EAAT1 expression and reduced uptake but increased glutamate release. These findings were verified by detecting elevated glutamate levels and reduced levels of EAAT1 in cerebrospinal fluid of untreated SMA type 2 and 3 patients. In addition, glutamate was elevated in serum samples, while EAAT1 was not detectable. Our data give evidence for the crucial role of spinal astrocytes in the pathogenesis of late-onset SMA, a potential driving force for MN loss by glutamate excitotoxicity caused by EAAT1 reduction as an early pathophysiological event. Furthermore, our study introduces EAAT1 as a potential therapeutic target for additional SMN-independent therapy strategies to complement SMN-enhancing drugs.
Collapse
Affiliation(s)
- Linda-Isabell Schmitt
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Christina David
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Rebecca Steffen
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Stefanie Hezel
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Andreas Roos
- Department of Pediatrics 1, Division of Neuropediatrics, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatrics 1, Division of Neuropediatrics, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Markus Leo
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Tim Hagenacker
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| |
Collapse
|
21
|
Ping Y, Ohata K, Kikushima K, Sakamoto T, Islam A, Xu L, Zhang H, Chen B, Yan J, Eto F, Nakane C, Takao K, Miyakawa T, Kabashima K, Watanabe M, Kahyo T, Yao I, Fukuda A, Ikegami K, Konishi Y, Setou M. Tubulin Polyglutamylation by TTLL1 and TTLL7 Regulate Glutamate Concentration in the Mice Brain. Biomolecules 2023; 13:biom13050784. [PMID: 37238654 DOI: 10.3390/biom13050784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
As an important neurotransmitter, glutamate acts in over 90% of excitatory synapses in the human brain. Its metabolic pathway is complicated, and the glutamate pool in neurons has not been fully elucidated. Tubulin polyglutamylation in the brain is mainly mediated by two tubulin tyrosine ligase-like (TTLL) proteins, TTLL1 and TTLL7, which have been indicated to be important for neuronal polarity. In this study, we constructed pure lines of Ttll1 and Ttll7 knockout mice. Ttll knockout mice showed several abnormal behaviors. Matrix-assisted laser desorption/ionization (MALDI) Imaging mass spectrometry (IMS) analyses of these brains showed increases in glutamate, suggesting that tubulin polyglutamylation by these TTLLs acts as a pool of glutamate in neurons and modulates some other amino acids related to glutamate.
Collapse
Affiliation(s)
- Yashuang Ping
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kenji Ohata
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kenji Kikushima
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takumi Sakamoto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Ariful Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hengsen Zhang
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Bin Chen
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Jing Yan
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Fumihiro Eto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Chiho Nakane
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Keizo Takao
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan
- Genetic Engineering and Functional Genomics Unit, Frontier Technology Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Miyakawa
- Genetic Engineering and Functional Genomics Unit, Frontier Technology Center, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Institute for Comprehensive Medical Science Division of Systems Medicine, Fujita Health University, Aichi 470-1192, Japan
| | - Katsuya Kabashima
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Ikuko Yao
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo 669-1330, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Koji Ikegami
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Hiroshima 734-8553, Japan
| | - Yoshiyuki Konishi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Applied Chemistry and Biotechnology, University of Fukui, 3-9-1 Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
22
|
Xu X, Wang J, Du S, Shen X, Lian J, Zhou J, Wang M, Feng W, Lv Z, Zhu J, Jin L, Sun H, Wu L, Wang X, Qiu H, Wang W, Teng H, Wang Y, Huang Z. Yes-associated protein regulates glutamate homeostasis through promoting the expression of excitatory amino acid transporter-2 in astrocytes via β-catenin signaling. Glia 2023; 71:1197-1216. [PMID: 36617748 DOI: 10.1002/glia.24332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 01/10/2023]
Abstract
The homeostasis of glutamate is mainly regulated by the excitatory amino acid transporters (EAATs), especially by EAAT2 in astrocytes. Excessive glutamate in the synaptic cleft caused by dysfunction or dysregulation of EAAT2 can lead to excitotoxicity, neuronal death and cognitive dysfunction. However, it remains unclear about the detailed regulation mechanism of expression and function of astrocytic EAAT2. In this study, first, we found increased neuronal death and impairment of cognitive function in YAPGFAP -CKO mice (conditionally knock out Yes-associated protein [YAP] in astrocytes), and identified EAAT2 as a downstream target of YAP through RNA sequencing. Second, the expression of EAAT2 was decreased in cultured YAP-/- astrocytes and the hippocampus of YAPGFAP -CKO mice, and glutamate uptake was reduced in YAP-/- astrocytes, but increased in YAP-upregulated astrocytes. Third, further investigation of the mechanism showed that the mRNA and protein levels of β-catenin were decreased in YAP-/- astrocytes and increased in YAP-upregulated astrocytes. Wnt3a activated YAP signaling and up-regulated EAAT2 through β-catenin. Furthermore, over-expression or activation of β-catenin partially restored the downregulation of EAAT2, the impairment of glutamate uptake, neuronal death and cognitive decline that caused by YAP deletion. Finally, activation of EAAT2 also rescued neuronal death and cognitive decline in YAPGFAP -CKO mice. Taken together, our study identifies an unrecognized role of YAP signaling in the regulation of glutamate homeostasis through the β-catenin/EAAT2 pathway in astrocytes, which may provide novel insights into the pathogenesis of brain diseases that closely related to the dysfunction or dysregulation of EAAT2, and promote the development of clinical strategy.
Collapse
Affiliation(s)
- Xingxing Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiaojiao Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Siyu Du
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xiya Shen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiashu Lian
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Zhou
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mianxian Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenjin Feng
- Zhejiang Sinogen Medical Equipment Co., Ltd., Wenzhou, China
| | - Zhaoting Lv
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Junzhe Zhu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Lingting Jin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huankun Sun
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Lihao Wu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Xiaoning Wang
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Haoyu Qiu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Honglin Teng
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Wang
- Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihui Huang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
23
|
Pidoplichko VI, Figueiredo TH, Braga MFM, Pan H, Marini AM. Alpha-linolenic acid enhances the facilitation of GABAergic neurotransmission in the BLA and CA1. Exp Biol Med (Maywood) 2023; 248:596-604. [PMID: 37208920 PMCID: PMC10350796 DOI: 10.1177/15353702231165010] [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: 01/31/2023] [Accepted: 03/02/2023] [Indexed: 05/21/2023] Open
Abstract
Hyperexcitability is a major mechanism implicated in several neuropsychiatric disorders, such as organophosphate-induced status epilepticus (SE), primary epilepsy, stroke, spinal cord injury, traumatic brain injury, schizophrenia, and autism spectrum disorders. Underlying mechanisms are diverse, but a functional impairment and loss of GABAergic inhibitory neurons are common features in many of these disorders. While novel therapies abound to correct for the loss of GABAergic inhibitory neurons, it has been difficult at best to improve the activities of daily living for the majority of patients. Alpha-linolenic acid (ALA) is an essential omega-3 polyunsaturated fatty acid found in plants. ALA exerts pleiotropic effects in the brain that attenuate injury in chronic and acute brain disease models. However, the effect of ALA on GABAergic neurotransmission in hyperexcitable brain regions involved in neuropsychiatric disorders, such as the basolateral amygdala (BLA) and CA1 subfield of the hippocampus, is unknown. Administration of a single dose of ALA (1500 nmol/kg) subcutaneously increased the charge transfer of inhibitory postsynaptic potential currents mediated by GABAA receptors in pyramidal neurons by 52% in the BLA and by 92% in the CA1 compared to vehicle animals a day later. Similar results were obtained in pyramidal neurons from the BLA and CA1 when ALA was bath-applied in slices from naïve animals. Importantly, pretreatment with the high-affinity, selective TrkB inhibitor, k252, completely abolished the ALA-induced increase in GABAergic neurotransmission in the BLA and CA1, suggesting a brain-derived neurotrophic factor (BDNF)-mediated mechanism. Addition of mature BDNF (20 ng/mL) significantly increased GABAA receptor inhibitory activity in the BLA and CA1 pyramidal neurons similar to the results obtained with ALA. ALA may be an effective treatment for neuropsychiatric disorders where hyperexcitability is a major feature.
Collapse
Affiliation(s)
- Volodymir I Pidoplichko
- Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Maria FM Braga
- Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Hongna Pan
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Ann M Marini
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
24
|
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
|
25
|
Conti F, Pietrobon D. Astrocytic Glutamate Transporters and Migraine. Neurochem Res 2023; 48:1167-1179. [PMID: 36583835 DOI: 10.1007/s11064-022-03849-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/31/2022]
Abstract
Glutamate levels and lifetime in the brain extracellular space are dinamically regulated by a family of Na+- and K+-dependent glutamate transporters, which thereby control numerous brain functions and play a role in numerous neurological and psychiatric diseases. Migraine is a neurological disorder characterized by recurrent attacks of typically throbbing and unilateral headache and by a global dysfunction in multisensory processing. Familial hemiplegic migraine type 2 (FHM2) is a rare monogenic form of migraine with aura caused by loss-of-function mutations in the α2 Na/K ATPase (α2NKA). In the adult brain, this pump is expressed almost exclusively in astrocytes where it is colocalized with glutamate transporters. Knockin mouse models of FHM2 (FHM2 mice) show a reduced density of glutamate transporters in perisynaptic astrocytic processes (mirroring the reduced expression of α2NKA) and a reduced rate of glutamate clearance at cortical synapses during neuronal activity and sensory stimulation. Here we review the migraine-relevant alterations produced by the astrocytic glutamate transport dysfunction in FHM2 mice and their underlying mechanisms, in particular regarding the enhanced brain susceptibility to cortical spreading depression (the phenomenon that underlies migraine aura and can also initiate the headache mechanisms) and the enhanced algesic response to a migraine trigger.
Collapse
Affiliation(s)
- Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.
| | - Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center (PNC), University of Padova, 35131, Padua, Italy.
- CNR Institute of Neuroscience, 35131, Padua, Italy.
| |
Collapse
|
26
|
Maková M, Kašparová S, Tvrdík T, Noguera M, Belovičová K, Csatlosová K, Dubovický M. Mirtazapine modulates Glutamate and GABA levels in the animal model of maternal depression. MRI and 1H MRS study in female rats. Behav Brain Res 2023; 442:114296. [PMID: 36641082 DOI: 10.1016/j.bbr.2023.114296] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
We aimed to determine, using in vivo magnetic resonance, whether maternal depression induced by chronic unpredictable stress (CUS) in the pre-gestational period in female rats would be evidenced by structural or neurometabolic changes in the hippocampal region of the brain. At the same time, appropriate behavioral tests were also administered after a relatively long two-month period of a stress paradigm. The objective of the study was not only to study an animal model of CUS using magnetic resonance imaging (MRI) and proton magnetic resonance spectroscopy (1H MRS) focused on the hippocampus, but also to use this technique to verify the effectiveness of mirtazapine antidepressant treatment. In the group with CUS, we found a significant decrease in the relative concentration of γ-aminobutyric acid (GABA/tCr) and glutamate+glutamine (Glx/tCr) compared to the control group, while we did not observe any statistically significant change in hippocampal volumes. Moreover, the forced swim test revealed an increase in depression-like behavior. The most important finding was the return of GABA/tCr and Glx/tCr levels to control levels during mirtazapine treatment; however, behavioral tests did not demonstrate any effects from mirtazapine treatment. In vivo1H MRS confirmed mirtazapine modulation of CUS in an animal model more robustly than behavioral tests.
Collapse
Affiliation(s)
- Marianna Maková
- Slovak University of Technology in Bratislava, Central Laboratory of Faculty of Food and Chemical Technology, Radlinského 9, Bratislava 81237, Slovak Republic.
| | - Svatava Kašparová
- Slovak University of Technology in Bratislava, Central Laboratory of Faculty of Food and Chemical Technology, Radlinského 9, Bratislava 81237, Slovak Republic.
| | - Tomáš Tvrdík
- Slovak University of Technology in Bratislava, Central Laboratory of Faculty of Food and Chemical Technology, Radlinského 9, Bratislava 81237, Slovak Republic; Department of Radiology, Faculty of Medicine of Comenius University in Bratislava, Slovak Medical University and University Hospital Bratislava, Limbová 12, Bratislava 83303, Slovak Republic.
| | - Mireia Noguera
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy Sciences, Dúbravská cesta 9, Bratislava 84104, Slovak Republic.
| | - Kristína Belovičová
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy Sciences, Dúbravská cesta 9, Bratislava 84104, Slovak Republic.
| | - Kristína Csatlosová
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy Sciences, Dúbravská cesta 9, Bratislava 84104, Slovak Republic.
| | - Michal Dubovický
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy Sciences, Dúbravská cesta 9, Bratislava 84104, Slovak Republic.
| |
Collapse
|
27
|
Yin C, Harms AC, Hankemeier T, Kindt A, de Lange ECM. Status of Metabolomic Measurement for Insights in Alzheimer's Disease Progression-What Is Missing? Int J Mol Sci 2023; 24:ijms24054960. [PMID: 36902391 PMCID: PMC10003384 DOI: 10.3390/ijms24054960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease, leading to the progressive loss of memory and other cognitive functions. As there is still no cure for AD, the growth in the number of susceptible individuals represents a major emerging threat to public health. Currently, the pathogenesis and etiology of AD remain poorly understood, while no efficient treatments are available to slow down the degenerative effects of AD. Metabolomics allows the study of biochemical alterations in pathological processes which may be involved in AD progression and to discover new therapeutic targets. In this review, we summarized and analyzed the results from studies on metabolomics analysis performed in biological samples of AD subjects and AD animal models. Then this information was analyzed by using MetaboAnalyst to find the disturbed pathways among different sample types in human and animal models at different disease stages. We discuss the underlying biochemical mechanisms involved, and the extent to which they could impact the specific hallmarks of AD. Then we identify gaps and challenges and provide recommendations for future metabolomics approaches to better understand AD pathogenesis.
Collapse
Affiliation(s)
- Chunyuan Yin
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Amy C. Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Alida Kindt
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Elizabeth C. M. de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Correspondence:
| |
Collapse
|
28
|
Fontana IC, Souza DG, Souza DO, Gee A, Zimmer ER, Bongarzone S. A Medicinal Chemistry Perspective on Excitatory Amino Acid Transporter 2 Dysfunction in Neurodegenerative Diseases. J Med Chem 2023; 66:2330-2346. [PMID: 36787643 PMCID: PMC9969404 DOI: 10.1021/acs.jmedchem.2c01572] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The excitatory amino acid transporter 2 (EAAT2) plays a key role in the clearance and recycling of glutamate - the major excitatory neurotransmitter in the mammalian brain. EAAT2 loss/dysfunction triggers a cascade of neurodegenerative events, comprising glutamatergic excitotoxicity and neuronal death. Nevertheless, our current knowledge regarding EAAT2 in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD), is restricted to post-mortem analysis of brain tissue and experimental models. Thus, detecting EAAT2 in the living human brain might be crucial to improve diagnosis/therapy for ALS and AD. This perspective article describes the role of EAAT2 in physio/pathological processes and provides a structure-activity relationship of EAAT2-binders, bringing two perspectives: therapy (activators) and diagnosis (molecular imaging tools).
Collapse
Affiliation(s)
- Igor C Fontana
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom.,Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 - Neo floor seventh, 141 83 Stockholm, Sweden
| | - Débora G Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Av. Ipiranga, 6681 Porto Alegre, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil
| | - Antony Gee
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Eduardo R Zimmer
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Av. Sarmento Leite 500, sala, 90035-003 Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry (PPGBioq), and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Av. Sarmento Leite 500, sala, 305 Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Av. Ipiranga, 6681 Porto Alegre, Brazil.,McGill University Research Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Salvatore Bongarzone
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| |
Collapse
|
29
|
Mi Z, Abrahamson EE, Ryu AY, Malek-Ahmadi M, Kofler JK, Fish KN, Sweet RA, Villemagne VL, Schneider JA, Mufson EJ, Ikonomovic MD. Vesicular Glutamate Transporter Changes in the Cortical Default Mode Network During the Clinical and Pathological Progression of Alzheimer's Disease. J Alzheimers Dis 2023; 94:227-246. [PMID: 37212097 PMCID: PMC10994206 DOI: 10.3233/jad-221063] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
BACKGROUND Altered glutamatergic neurotransmission may contribute to impaired default mode network (DMN) function in Alzheimer's disease (AD). Among the DMN hub regions, frontal cortex (FC) was suggested to undergo a glutamatergic plasticity response in prodromal AD, while the status of glutamatergic synapses in the precuneus (PreC) during clinical-neuropathological AD progression is not known. OBJECTIVE To quantify vesicular glutamate transporter VGluT1- and VGluT2-containing synaptic terminals in PreC and FC across clinical stages of AD. METHODS Unbiased sampling and quantitative confocal immunofluorescence of cortical VGluT1- and VGluT2-immunoreactive profiles and spinophilin-labeled dendritic spines were performed in cases with no cognitive impairment (NCI), mild cognitive impairment (MCI), mild-moderate AD (mAD), or moderate-severe AD (sAD). RESULTS In both regions, loss of VGluT1-positive profile density was seen in sAD compared to NCI, MCI, and mAD. VGluT1-positive profile intensity in PreC did not differ across groups, while in FC it was greater in MCI, mAD, and sAD compared to NCI. VGluT2 measures were stable in PreC while FC had greater VGluT2-positive profile density in MCI compared to sAD, but not NCI or mAD. Spinophilin measures in PreC were lower in mAD and sAD compared to NCI, while in FC they were stable across groups. Lower VGluT1 and spinophilin measures in PreC, but not FC, correlated with greater neuropathology. CONCLUSION Frank loss of VGluT1 in advanced AD relative to NCI occurs in both DMN regions. In FC, an upregulation of VGluT1 protein content in remaining glutamatergic terminals may contribute to this region's plasticity response in AD.
Collapse
Affiliation(s)
- Zhiping Mi
- Department of Neurology, University of Pittsburgh School of
Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA
Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Eric E. Abrahamson
- Department of Neurology, University of Pittsburgh School of
Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA
Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Angela Y. Ryu
- Department of Neurology, University of Pittsburgh School of
Medicine, Pittsburgh, PA, USA
| | - Michael Malek-Ahmadi
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
- Department of Biomedical Informatics, University of Arizona
College of Medicine, Phoenix, AZ, USA
| | - Julia K. Kofler
- Department of Pathology, University of Pittsburgh School of
Medicine, Pittsburgh, PA, USA
| | - Kenneth N. Fish
- Department of Psychiatry, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA
| | - Robert A. Sweet
- Department of Neurology, University of Pittsburgh School of
Medicine, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA
| | - Victor L. Villemagne
- Department of Psychiatry, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Rush University
Medical Center, Chicago, IL, USA
| | - Elliott J. Mufson
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
- Departments of Translational Neurosciences and Neurology,
Barrow Neurological Institute, Phoenix, AZ, USA
| | - Milos D. Ikonomovic
- Department of Neurology, University of Pittsburgh School of
Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA
Pittsburgh Healthcare System, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Holton K. The potential role of dietary intervention for the treatment of neuroinflammation. TRANSLATIONAL NEUROIMMUNOLOGY, VOLUME 7 2023:239-266. [DOI: 10.1016/b978-0-323-85841-0.00022-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
31
|
Halaris A, Cook J. The Glutamatergic System in Treatment-Resistant Depression and Comparative Effectiveness of Ketamine and Esketamine: Role of Inflammation? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:487-512. [PMID: 36949323 DOI: 10.1007/978-981-19-7376-5_21] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The glutamatergic system is the primary excitatory pathway within the CNS and is responsible for cognition, memory, learning, emotion, and mood. Because of its significant importance in widespread nervous system function, it is tightly regulated through multiple mechanisms, such as glutamate recycling, microglial interactions, and inflammatory pathways. Imbalance within the glutamatergic system has been implicated in a wide range of pathological conditions including neurodegenerative conditions, neuromuscular conditions, and mood disorders including depression. Major depressive disorder (MDD) is the most common mood disorder worldwide, has a high prevalence rate, and afflicts approximately 280 million people. While there are numerous treatments for the disease, 30-40% of patients are unresponsive to treatment and deemed treatment resistant; approximately another third experience only partial improvement (World Health Organization, Depression fact sheet [Internet], 2020). Esketamine, the S-enantiomer of ketamine, was approved by the Food and Drug Administration for treatment-resistant depression (TRD) in 2019 and has offered new hope to patients. It is the first treatment targeting the glutamatergic system through a complex mechanism. Numerous studies have implicated imbalance in the glutamatergic system in depression and treatment resistance. Esketamine and ketamine principally work through inhibition of the NMDA receptor, though more recent studies have implicated numerous other mechanisms mediating the antidepressant efficacy of these agents. These mechanisms include increase in brain-derived neurotrophic factor (BDNF), activation of mammalian target of the rapamycin complex (mTORC), and reduction in inflammation. Esketamine and ketamine have been shown to decrease inflammation in numerous ways principally through reducing pro-inflammatory cytokines (e.g., TNF-α, IL-6) (Loix et al., Acta Anaesthesiol Belg 62(1):47-58, 2011; Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021). This anti-inflammatory effect has also been shown to be involved in the antidepressive properties of both ketamine and esketamine (Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021).
Collapse
Affiliation(s)
- Angelos Halaris
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA.
| | - John Cook
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
32
|
Bakhshi S, Tehrani-Doost M, Batouli SAH. Evaluation of fronto-cerebellar neurometabolites in youth with ADHD compared to the healthy group and their associations with cognitive and behavioral characteristics: A proton magnetic spectroscopy study. Int J Psychophysiol 2022; 182:190-199. [DOI: 10.1016/j.ijpsycho.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
|
33
|
Romero G, Park J, Koehler F, Pralle A, Anikeeva P. Modulating cell signalling in vivo with magnetic nanotransducers. NATURE REVIEWS. METHODS PRIMERS 2022; 2:92. [PMID: 38111858 PMCID: PMC10727510 DOI: 10.1038/s43586-022-00170-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/15/2022] [Indexed: 12/20/2023]
Abstract
Weak magnetic fields offer nearly lossless transmission of signals within biological tissue. Magnetic nanomaterials are capable of transducing magnetic fields into a range of biologically relevant signals in vitro and in vivo. These nanotransducers have recently enabled magnetic control of cellular processes, from neuronal firing and gene expression to programmed apoptosis. Effective implementation of magnetically controlled cellular signalling relies on careful tailoring of magnetic nanotransducers and magnetic fields to the responses of the intended molecular targets. This primer discusses the versatility of magnetic modulation modalities and offers practical guidelines for selection of appropriate materials and field parameters, with a particular focus on applications in neuroscience. With recent developments in magnetic instrumentation and nanoparticle chemistries, including those that are commercially available, magnetic approaches promise to empower research aimed at connecting molecular and cellular signalling to physiology and behaviour in untethered moving subjects.
Collapse
Affiliation(s)
- Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Jimin Park
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Florian Koehler
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnd Pralle
- Department of Physics, University at Buffalo, the State University of New York, Buffalo, NY, USA
| | - Polina Anikeeva
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
34
|
Li SH, Abd-Elrahman KS, Ferguson SS. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacol Ther 2022; 239:108275. [DOI: 10.1016/j.pharmthera.2022.108275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
|
35
|
Bartesaghi R. Brain circuit pathology in Down syndrome: from neurons to neural networks. Rev Neurosci 2022; 34:365-423. [PMID: 36170842 DOI: 10.1515/revneuro-2022-0067] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/28/2022] [Indexed: 11/15/2022]
Abstract
Down syndrome (DS), a genetic pathology caused by triplication of chromosome 21, is characterized by brain hypotrophy and impairment of cognition starting from infancy. While studies in mouse models of DS have elucidated the major neuroanatomical and neurochemical defects of DS, comparatively fewer investigations have focused on the electrophysiology of the DS brain. Electrical activity is at the basis of brain functioning. Therefore, knowledge of the way in which brain circuits operate in DS is fundamental to understand the causes of behavioral impairment and devise targeted interventions. This review summarizes the state of the art regarding the electrical properties of the DS brain, starting from individual neurons and culminating in signal processing in whole neuronal networks. The reported evidence derives from mouse models of DS and from brain tissues and neurons derived from individuals with DS. EEG data recorded in individuals with DS are also provided as a key tool to understand the impact of brain circuit alterations on global brain activity.
Collapse
Affiliation(s)
- Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
36
|
Clock gene-dependent glutamate dynamics in the bean bug brain regulate photoperiodic reproduction. PLoS Biol 2022; 20:e3001734. [PMID: 36067166 PMCID: PMC9447885 DOI: 10.1371/journal.pbio.3001734] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022] Open
Abstract
Animals adequately modulate their physiological status and behavior according to the season. Many animals sense photoperiod for seasonal adaptation, and the circadian clock is suggested to play an essential role in photoperiodic time measurement. However, circadian clock-driven neural signals in the brain that convey photoperiodic information remain unclear. Here, we focused on brain extracellular dynamics of a classical neurotransmitter glutamate, which is widely used for brain neurotransmission, and analyzed its involvement in photoperiodic responses using the bean bug Riptortus pedestris that shows clear photoperiodism in reproduction. Extracellular glutamate levels in the whole brain were significantly higher under short-day conditions, which cause a reproductive diapause, than those under long-day conditions. The photoperiodic change in glutamate levels was clearly abolished by knockdown of the clock gene period. We also demonstrated that genetic modulation of glutamate dynamics by knockdown of glutamate-metabolizing enzyme genes, glutamate oxaloacetate transaminase (got) and glutamine synthetase (gs), attenuated photoperiodic responses in reproduction. Further, we investigated glutamate-mediated photoperiodic modulations at a cellular level, focusing on the pars intercerebralis (PI) neurons that photoperiodically change their neural activity and promote oviposition. Electrophysiological analyses showed that L-Glutamate acts as an inhibitory signal to PI neurons via glutamate-gated chloride channel (GluCl). Additionally, combination of electrophysiology and genetics revealed that knockdown of got, gs, and glucl disrupted cellular photoperiodic responses of the PI neurons, in addition to reproductive phenotypes. Our results reveal that the extracellular glutamate dynamics are photoperiodically regulated depending on the clock gene and play an essential role in the photoperiodic control of reproduction via inhibitory pathways.
Collapse
|
37
|
Martinez-Lozada Z, Hewett SJ, Zafra F, Ortega A. Editorial: The known, the unknown, and the future of glutamate transporters. Front Cell Neurosci 2022; 16:1005834. [PMID: 36060278 PMCID: PMC9433117 DOI: 10.3389/fncel.2022.1005834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Zila Martinez-Lozada
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
- *Correspondence: Zila Martinez-Lozada
| | - Sandra J. Hewett
- Program in Neuroscience, Department of Biology, Syracuse University, Syracuse, NY, United States
| | - Francisco Zafra
- Center of Molecular Biology Severo Ochoa, School of Science, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz — IdiPAZ, Madrid, Spain
| | - Arturo Ortega
- Department of Toxicology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
38
|
Chen J, Zhang J, Yang DD, Li ZC, Zhao B, Chen Y, He Z. Clonidine ameliorates cerebral ischemia-reperfusion injury by up-regulating the GluN3 subunits of NMDA receptor. Metab Brain Dis 2022; 37:1829-1841. [PMID: 35727521 DOI: 10.1007/s11011-022-01028-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/04/2022] [Indexed: 10/18/2022]
Abstract
This study aimed to investigate the protective effects of the alpha-2 adrenergic receptor (α2-AR) agonist, clonidine, on the cerebral ischemia-reperfusion (I/R) injury and elaborate the underlying mechanisms. Cerebral I/R model was established by middle cerebral artery occlusion (MCAO) for 2 h followed by reperfusion for 4 h in adult male SD rats. Saline, clonidine and yohimbine (an α2-AR antagonist) were intraperitoneally administered each day for one week before surgery. Neurological deficit was evaluated just before decapitation. TTC staining was applied for correlation of cerebral infarction volume. HE staining was performed to observe the neuron morphology. Immunohistochemical staining was performed to detect the localization and expression of GluN3 proteins. Western blot analysis also was used to detect the expression levels of GluN3 proteins. Our data showed that clonidine ameliorated neurological deficit and reduced the cerebral infarction volume of the rats with cerebral I/R. It is worth noting that treatment with clonidine up-regulated the protein expression of GluN3 in the rats with the cerebral I/R, especially in the cell membrane. Moreover, clonidine also up-regulated the transposition from cytoplasm to cell membrane of GluN3 after cerebral I/R. In addition, yohimbine abolished the neuroprotective effects of clonidine. The results indicated that clonidine played a protective role in cerebral I/R injury through regulation of the protein expression of GluN3 subunits of N-methyl-D-aspartate (NMDA) receptor.
Collapse
Affiliation(s)
- Jing Chen
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Juan Zhang
- The First People's Hospital of Yichang, Yichang, 443000, People's Republic of China
| | - Dan-Dan Yang
- The Second People's Hospital of China Three Gorges University, Yichang, 443000, People's Republic of China
| | - Zi-Cheng Li
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Bo Zhao
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Yue Chen
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Zhi He
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China.
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China.
| |
Collapse
|
39
|
Rodrigues T, Piccirillo S, Magi S, Preziuso A, Dos Santos Ramos V, Serfilippi T, Orciani M, Maciel Palacio Alvarez M, Luis Dos Santos Tersariol I, Amoroso S, Lariccia V. Control of Ca 2+ and metabolic homeostasis by the Na +/Ca 2+ exchangers (NCXs) in health and disease. Biochem Pharmacol 2022; 203:115163. [PMID: 35803319 DOI: 10.1016/j.bcp.2022.115163] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022]
Abstract
Spatial and temporal control of calcium (Ca2+) levels is essential for the background rhythms and responses of living cells to environmental stimuli. Whatever other regulators a given cellular activity may have, localized and wider scale Ca2+ events (sparks, transients, and waves) are hierarchical determinants of fundamental processes such as cell contraction, excitability, growth, metabolism and survival. Different cell types express specific channels, pumps and exchangers to efficiently generate and adapt Ca2+ patterns to cell requirements. The Na+/Ca2+ exchangers (NCXs) in particular contribute to Ca2+ homeostasis by buffering intracellular Ca2+ loads according to the electrochemical gradients of substrate ions - i.e., Ca2+ and sodium (Na+) - and under a dynamic control of redundant regulatory processes. An interesting feature of NCX emerges from the strict relationship that connects transporter activity with cell metabolism: on the one hand NCX operates under constant control of ATP-dependent regulatory processes, on the other hand the ion fluxes generated through NCX provide mechanistic support for the Na+-driven uptake of glutamate and Ca2+ influx to fuel mitochondrial respiration. Proof of concept evidence highlights therapeutic potential of preserving a timed and balanced NCX activity in a growing rate of diseases (including excitability, neurodegenerative, and proliferative disorders) because of an improved ability of stressed cells to safely maintain ion gradients and mitochondrial bioenergetics. Here, we will summarize and review recent works that have focused on the pathophysiological roles of NCXs in balancing the two-way relationship between Ca2+ signals and metabolism.
Collapse
Affiliation(s)
- Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil.
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vyctória Dos Santos Ramos
- Interdisciplinary Center for Biochemistry Investigation (CIIB), University of Mogi das Cruzes (UMC), Mogi das Cruzes, SP, Brazil
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Histology, University "Politecnica delle Marche", Ancona, Italy.
| | - Marcela Maciel Palacio Alvarez
- Department of Biochemistry, São Paulo School of Medicine, Federal University of São Paulo (Unifesp) São Paulo, SP, Brazil
| | | | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| |
Collapse
|
40
|
A Peptide Aptamer Based Electrochemical Amperometric Sensor for Sensitive L-Glutamate Detection. Bioelectrochemistry 2022; 146:108165. [DOI: 10.1016/j.bioelechem.2022.108165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 01/05/2023]
|
41
|
Zhang C, He J, Wang X, Su R, Huang Q, Qiao F, Qin C, Qin J, Chen L. Dietary gamma-aminobutyric acid (GABA) improves non-specific immunity and alleviates lipopolysaccharide (LPS)-induced immune overresponse in juvenile Chinese mitten crab (Eriocheir sinensis). FISH & SHELLFISH IMMUNOLOGY 2022; 124:480-489. [PMID: 35489590 DOI: 10.1016/j.fsi.2022.04.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
Inhibitory neurotransmitter gamma-aminobutyric acid (GABA) is an immunomodulator to inhibit immune-mediated pro-inflammatory response and has been used to treat various immune-related diseases in mammals. However, the immunoregulatory effect of GABA in crustaceans has not been reported. This study evaluates the regulatory effect of dietary GABA supplementation on the innate immune status and immunoregulatory potential in lipopolysaccharide (LPS)-induced immune response in juvenile Eriocheir sinensis. Juvenile crabs were fed with six diets supplemented with graded GABA levels (0, 40, 80, 160, 320 and 640 mg/kg dry matter) for 8 weeks and then 24 h LPS challenge test was carried out. The results showed that dietary GABA supplementation significantly decreased mortality at 4 and 8 weeks. Moreover, the hemocyanin content, acid phosphatase, and alkaline phosphatase activities significantly increased in the crabs fed GABA supplementation compared with the control. On the contrary, the alanine aminotransferase and alanine aminotransferase activities in serum decreased significantly in the GABA supplementation groups compared with the control. Similarly, superoxide dismutase activity, glutathione content, and the transcriptional expression of the antioxidant-related genes and immune-related genes were significantly higher in the GABA supplementation groups than in the control. In addition, the mRNA expressions of anti-lipopolysaccharide factors (ALF 1, ALF 2, ALF 3) and inflammatory signaling pathways related genes (TLR, Myd88, Relish, LITAF, P38-MAPK, ADAM17) were significantly up-regulated in LPS stimulation groups compared with PBS treatment. Meanwhile, pro-apoptosis-related genes' mRNA expressions were significantly up-regulated, and anti-apoptosis-related genes were significantly down-regulated under LPS stimulation compared with PBS treatment. However, GABA pretreatment effectively alleviated LPS-induced immune overresponse and apoptosis. Therefore, this study demonstrates that dietary GABA supplementation could be used as an immunomodulator to improve the non-specific immunity and antioxidant capacity and alleviate the immune-mediated immune overresponse of juvenile E. sinensis.
Collapse
Affiliation(s)
- Cong Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Jiaqi He
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Xiaodan Wang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China.
| | - Ruiying Su
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Qincheng Huang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Sichuan, 641100, PR China
| | - Jianguang Qin
- College of Science and Engineering, Flinders University, Adelaide, SA, 5001, Australia
| | - Liqiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China.
| |
Collapse
|
42
|
Chiu KM, Lin TY, Lee MY, Lu CW, Wang SJ. Lappaconitine inhibits glutamate release from rat cerebrocortical nerve terminals by suppressing Ca2+ influx and protein kinase A cascade. Neurotoxicology 2022; 91:218-227. [DOI: 10.1016/j.neuro.2022.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 05/02/2022] [Accepted: 05/20/2022] [Indexed: 10/18/2022]
|
43
|
Shi B, Zhao X, Chai Y, Qin P, Qu W, Lin Q, Zhang Y. Detection of L‐Aspartic Acid and L‐Glutamic Acid in Water Using a Fluorescent Nanoparticle Constructed by Pillar[5]arene‐Based Molecular Recognition. ChemistrySelect 2022. [DOI: 10.1002/slct.202200757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Bingbing Shi
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education Key Laboratory of Eco-environmental Polymer Materials of Gansu Province College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou Gansu 730070 P. R. China
| | - Xing‐Xing Zhao
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education Key Laboratory of Eco-environmental Polymer Materials of Gansu Province College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou Gansu 730070 P. R. China
| | - Yongping Chai
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education Key Laboratory of Eco-environmental Polymer Materials of Gansu Province College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou Gansu 730070 P. R. China
| | - Peng Qin
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education Key Laboratory of Eco-environmental Polymer Materials of Gansu Province College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou Gansu 730070 P. R. China
| | - Wen‐Juan Qu
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education Key Laboratory of Eco-environmental Polymer Materials of Gansu Province College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou Gansu 730070 P. R. China
| | - Qi Lin
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education Key Laboratory of Eco-environmental Polymer Materials of Gansu Province College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou Gansu 730070 P. R. China
| | - You‐Ming Zhang
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education Key Laboratory of Eco-environmental Polymer Materials of Gansu Province College of Chemistry and Chemical Engineering Northwest Normal University Lanzhou Gansu 730070 P. R. China
- Gansu Natural Energy Research Institute Lanzhou Gansu 730046 P. R. China
| |
Collapse
|
44
|
Romano AD, Villani R, Sangineto M, Cassano T, Serviddio G. The GLP-1 receptor agonist Exendin-4 modulates hippocampal NMDA-receptor signalling in aged rats and improves cognitive impairment in diabetic elderly patients. JOURNAL OF GERONTOLOGY AND GERIATRICS 2022. [DOI: 10.36150/2499-6564-n474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
45
|
Neuparth T, Alves N, Machado AM, Pinheiro M, Montes R, Rodil R, Barros S, Ruivo R, Castro LFC, Quintana JB, Santos MM. Neuroendocrine pathways at risk? Simvastatin induces inter and transgenerational disruption in the keystone amphipod Gammarus locusta. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 244:106095. [PMID: 35121565 DOI: 10.1016/j.aquatox.2022.106095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 06/14/2023]
Abstract
The primary focus of environmental toxicological studies is to address the direct effects of chemicals on exposed organisms (parental generation - F0), mostly overlooking effects on subsequent non-exposed generations (F1 and F2 - intergenerational and F3 transgenerational, respectively). Here, we addressed the effects of simvastatin (SIM), one of the most widely prescribed human pharmaceuticals for the primary treatment of hypercholesterolemia, using the keystone crustacean Gammarus locusta. We demonstrate that SIM, at environmentally relevant concentrations, has significant inter and transgenerational (F1 and F3) effects in key signaling pathways involved in crustaceans' neuroendocrine regulation (Ecdysteroids, Catecholamines, NO/cGMP/PKG, GABAergic and Cholinergic signaling pathways), concomitantly with changes in apical endpoints, such as depressed reproduction and growth. These findings are an essential step to improve hazard and risk assessment of biological active compounds, such as SIM, and highlight the importance of studying the transgenerational effects of environmental chemicals in animals' neuroendocrine regulation.
Collapse
Affiliation(s)
- T Neuparth
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal.
| | - N Alves
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
| | - A M Machado
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
| | - M Pinheiro
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
| | - R Montes
- Department of Analytical Chemistry, Nutrition and Food Sciences, IAQBUS - Institute of Research on Chemical and Biological Analysis, Universidade de Santiago de Compostela, R. Constantino Candeira S/N, 15782 Santiago de Compostela, Spain
| | - R Rodil
- Department of Analytical Chemistry, Nutrition and Food Sciences, IAQBUS - Institute of Research on Chemical and Biological Analysis, Universidade de Santiago de Compostela, R. Constantino Candeira S/N, 15782 Santiago de Compostela, Spain
| | - S Barros
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, Quinta de Prados - Ed. Blocos Laboratoriais C1.10, 5000-801, Vila Real, Portugal
| | - R Ruivo
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| | - L Filipe C Castro
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
| | - J B Quintana
- Department of Analytical Chemistry, Nutrition and Food Sciences, IAQBUS - Institute of Research on Chemical and Biological Analysis, Universidade de Santiago de Compostela, R. Constantino Candeira S/N, 15782 Santiago de Compostela, Spain
| | - M M Santos
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; FCUP - Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
46
|
McGrath T, Baskerville R, Rogero M, Castell L. Emerging Evidence for the Widespread Role of Glutamatergic Dysfunction in Neuropsychiatric Diseases. Nutrients 2022; 14:nu14050917. [PMID: 35267893 PMCID: PMC8912368 DOI: 10.3390/nu14050917] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
The monoamine model of depression has long formed the basis of drug development but fails to explain treatment resistance or associations with stress or inflammation. Recent animal research, clinical trials of ketamine (a glutamate receptor antagonist), neuroimaging research, and microbiome studies provide increasing evidence of glutamatergic dysfunction in depression and other disorders. Glutamatergic involvement across diverse neuropathologies including psychoses, neurodevelopmental, neurodegenerative conditions, and brain injury forms the rationale for this review. Glutamate is the brain's principal excitatory neurotransmitter (NT), a metabolic and synthesis substrate, and an immune mediator. These overlapping roles and multiple glutamate NT receptor types complicate research into glutamate neurotransmission. The glutamate microcircuit comprises excitatory glutamatergic neurons, astrocytes controlling synaptic space levels, through glutamate reuptake, and inhibitory GABA interneurons. Astroglia generate and respond to inflammatory mediators. Glutamatergic microcircuits also act at the brain/body interface via the microbiome, kynurenine pathway, and hypothalamus-pituitary-adrenal axis. Disruption of excitatory/inhibitory homeostasis causing neuro-excitotoxicity, with neuronal impairment, causes depression and cognition symptoms via limbic and prefrontal regions, respectively. Persistent dysfunction reduces neuronal plasticity and growth causing neuronal death and tissue atrophy in neurodegenerative diseases. A conceptual overview of brain glutamatergic activity and peripheral interfacing is presented, including the common mechanisms that diverse diseases share when glutamate homeostasis is disrupted.
Collapse
Affiliation(s)
- Thomas McGrath
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| | - Richard Baskerville
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
- Correspondence:
| | - Marcelo Rogero
- School of Public Health, University of Sao Paulo, Sao Paulo 01246-904, Brazil;
| | - Linda Castell
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| |
Collapse
|
47
|
Xia X, Wang Y, Qin Y, Zhao S, Zheng JC. Exosome: A novel neurotransmission modulator or non-canonical neurotransmitter? Ageing Res Rev 2022; 74:101558. [PMID: 34990846 DOI: 10.1016/j.arr.2021.101558] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/13/2021] [Accepted: 12/30/2021] [Indexed: 02/08/2023]
Abstract
Neurotransmission is the electrical impulse-triggered propagation of signals between neurons or between neurons and other cell types such as skeletal muscle cells. Recent studies point out the involvement of exosomes, a type of small bilipid layer-enclosed extracellular vesicles, in regulating neurotransmission. Through horizontally transferring proteins, lipids, and nucleic acids, exosomes can modulate synaptic activities rapidly by controlling neurotransmitter release or progressively by regulating neural plasticity including synapse formation, neurite growth & removal, and axon guidance & elongation. In this review, we summarize the similarities and differences between exosomes and synaptic vesicles in their biogenesis, contents, and release. We also highlight the recent progress made in demonstrating the biological roles of exosome in regulating neurotransmission, and propose a modified model of neurotransmission, in which exosomes act as novel neurotransmitters. Lastly, we provide a comprehensive discussion of the enlightenment of the current knowledge on neurotransmission to the future directions of exosome research.
Collapse
|
48
|
Xia L, Liu L, Cai Y, Zhang Y, Tong F, Wang Q, Ding J, Wang X. Inhibition of Gasdermin D-Mediated Pyroptosis Attenuates the Severity of Seizures and Astroglial Damage in Kainic Acid-Induced Epileptic Mice. Front Pharmacol 2022; 12:751644. [PMID: 35153737 PMCID: PMC8831916 DOI: 10.3389/fphar.2021.751644] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 12/27/2021] [Indexed: 01/11/2023] Open
Abstract
Objective: Our study aimed to explore whether gasdermin D (GSDMD)-mediated pyroptosis is involved in the mechanism of kainic acid-induced seizures. Methods: C57BL/6 mice were randomly divided into sham and epilepsy groups. The epilepsy group was intrahippocampally injected with kainic acid to induce status epilepticus (SE), and the sham group was injected with an equal volume of saline. Dimethyl fumarate (DMF) was used as the GSDMD N-terminal fragments (GSDMD-N) inhibitor and suspended in 0.5% sodium carboxymethyl cellulose (CMC) for orally administration. The epilepsy group was divided into SE + CMC and SE + DMF groups. In the SE + DMF group, DMF was orally administered for 1 week before SE induction and was continued until the end of the experiment. An equal volume of CMC was administered to the sham and SE + CMC groups. Recurrent spontaneous seizures (SRSs) were monitored for 21 days after SE. Western blot analysis and immunofluorescent staining was performed. Results: The expression of GSDMD increased at 7–21 days post-SE, and GSDMD-N expression was significantly elevated 7 days after SE in both ipsilateral and contralateral hippocampus. GSDMD-positive cells were co-labeled with astrocytes, but not neurons or microglia. Astroglial damage occurs following status epilepticus (SE). Damaged astrocytes showed typical clasmatodendrosis in the CA1 region containing strong GSDMD expression at 7–21 days post-SE, accompanied by activated microglia. In the SE + DMF group, the expression of GSDMD-N was significantly inhibited compared to that in the SE + CMC group. After administration of DMF, SRSs at 7–21 days after SE were significantly decreased, and the number of clasmatodendritic astrocytes, microglia, and the expression of inflammatory factors such as IL-1β and IL-18 were significantly attenuated. Conclusion: GSDMD-mediated pyroptosis is involved in the mechanism of kainic acid-induced seizures. Our study provides a new potential therapeutic target for seizure control.
Collapse
Affiliation(s)
- Lu Xia
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Liu
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiying Cai
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiying Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fangchao Tong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiang Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Jing Ding, ; Xin Wang,
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of the State Key Laboratory of Medical Neurobiology, The institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
- *Correspondence: Jing Ding, ; Xin Wang,
| |
Collapse
|
49
|
Rapid Regulation of Glutamate Transport: Where Do We Go from Here? Neurochem Res 2022; 47:61-84. [PMID: 33893911 PMCID: PMC8542062 DOI: 10.1007/s11064-021-03329-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 01/03/2023]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system (CNS). A family of five Na+-dependent transporters maintain low levels of extracellular glutamate and shape excitatory signaling. Shortly after the research group of the person being honored in this special issue (Dr. Baruch Kanner) cloned one of these transporters, his group and several others showed that their activity can be acutely (within minutes to hours) regulated. Since this time, several different signals and post-translational modifications have been implicated in the regulation of these transporters. In this review, we will provide a brief introduction to the distribution and function of this family of glutamate transporters. This will be followed by a discussion of the signals that rapidly control the activity and/or localization of these transporters, including protein kinase C, ubiquitination, glutamate transporter substrates, nitrosylation, and palmitoylation. We also include the results of our attempts to define the role of palmitoylation in the regulation of GLT-1 in crude synaptosomes. In some cases, the mechanisms have been fairly well-defined, but in others, the mechanisms are not understood. In several cases, contradictory phenomena have been observed by more than one group; we describe these studies with the goal of identifying the opportunities for advancing the field. Abnormal glutamatergic signaling has been implicated in a wide variety of psychiatric and neurologic disorders. Although recent studies have begun to link regulation of glutamate transporters to the pathogenesis of these disorders, it will be difficult to determine how regulation influences signaling or pathophysiology of glutamate without a better understanding of the mechanisms involved.
Collapse
|
50
|
Kumar M, Singh S, Rana P, Kumar P, Sekhri T, Kanwar R, D'Souza M, Khushu S. Neurometabolite Changes in Hyperthyroid Patients Before and After Antithyroid Treatment: An in vivo 1H MRS Study. Front Hum Neurosci 2021; 15:739917. [PMID: 34899214 PMCID: PMC8662363 DOI: 10.3389/fnhum.2021.739917] [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: 07/14/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: Patients with hyperthyroidism have frequent neuropsychiatric symptoms such as lack of attention, concentration, poor memory, impaired executive functions, depression, and anxiety. These neurocognitive impairments such as memory, attention, and executive functions appear to be associated with dysfunction in brain regions. This study was conducted to investigate the metabolic changes in the brain subcortical regions, i.e., posterior parietal cortex and dorsolateral prefrontal cortex (DLPFC), in patients with hyperthyroidism before and after antithyroid treatment using proton magnetic resonance spectroscopy (1H MRS). Materials and Methods: We collected neuropsychological and 1H MRS data from posterior parietal cortex and DLPFC, in both control (N = 30) and hyperthyroid (N = 30) patients. In addition, follow-up data were available for 19 patients treated with carbimazole for 30 weeks. The relative ratios of the neurometabolites were calculated using the Linear Combination Model (LCModel). Analysis of co-variance using Bonferroni correction was performed between healthy controls and hyperthyroid patients, and a paired t-test was applied in patients at baseline and follow-up. Spearman's rank-order correlation was used to analyze bivariate associations between thyroid hormone levels and metabolite ratios, and the partial correlation analysis was performed between neuropsychological scores and metabolite ratios, with age and sex as covariates, in the patients before and after treatment. Results: Our results revealed a significant decrease in choline/creatine [glycerophosphocholine (GPC) + phosphocholine (PCh)/creatine (tCr)] in both the posterior parietal cortex and DLPFC in hyperthyroid patients, and these changes were reversible after antithyroid treatment. The posterior parietal cortex also showed significantly reduced glutamate/creatine (Glu/tCr), (glutamate + glutamine)/creatine (Glx/tCr), and increased glutathione/creatine (GSH/tCr) ratios in the hyperthyroid patients over control subjects. In DLPFC, only (N-acetyl aspartate + N-acetyl aspartyl-glutamate)/creatine (NAA + NAAG)/tCr was increased in the hyperthyroid patients. After antithyroid treatment, (GPC + PCh)/tCr increased, and Glx/tCr decreased in both brain regions in the patients at follow-up. Gln/tCr in the posterior parietal cortex was decreased in patients at follow-up. Interestingly, (GPC + PCh)/tCr in DLPFC showed a significantly inverse correlation with free tri-iodothyronine (fT3) in hyperthyroid patients at baseline, whereas NAA/tCr showed positive correlations with fT3 and free thyroxine (fT4) in hyperthyroid patients before and after antithyroid treatment, in the posterior parietal cortex. In DLPFC, only (NAA + NAAG)/tCr showed positive correlations with fT3 and fT4 in the patients before treatment. Conclusion: The overall findings suggest that all the brain metabolite changes were not completely reversed in the hyperthyroid patients after antithyroid treatment, even after achieving euthyroidism.
Collapse
Affiliation(s)
- Mukesh Kumar
- NMR Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India
| | - Sadhana Singh
- NMR Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India.,Centre for Ayurveda Biology and Holistic Nutrition (CABHN), The University of Trans-Disciplinary Health Sciences and Technology, Bengaluru, India
| | - Poonam Rana
- NMR Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India
| | - Pawan Kumar
- NMR Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India
| | - Tarun Sekhri
- Thyroid Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India
| | - Ratnesh Kanwar
- Thyroid Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India
| | - Maria D'Souza
- NMR Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India
| | - Subash Khushu
- NMR Research Center, Institute of Nuclear Medicine and Allied Sciences (INMAS), New Delhi, India.,Centre for Ayurveda Biology and Holistic Nutrition (CABHN), The University of Trans-Disciplinary Health Sciences and Technology, Bengaluru, India
| |
Collapse
|