1
|
Cheng J, Koch ET, Ramandi D, Mackay JP, O'Leary TP, Rees-Jones W, Raymond LA. Synaptic modulation of glutamate in striatum of the YAC128 mouse model of Huntington disease. Neurobiol Dis 2025; 205:106774. [PMID: 39716682 DOI: 10.1016/j.nbd.2024.106774] [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: 10/02/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Altered balance between striatal direct and indirect pathways contributes to early motor, cognitive and psychiatric symptoms in Huntington disease (HD). While degeneration of striatal D2-type dopamine receptor (D2)-expressing indirect pathway medium spiny neurons (iMSNs) occurs prior to that of D1-type dopamine receptor (D1)-expressing direct pathway neurons, altered corticostriatal synaptic function precedes degeneration. D2-mediated signaling on iMSNs reduces their excitability and promotes endocannabinoid (eCB) synthesis, suppressing glutamate release from cortical afferents. D2 receptors are also expressed on glutamatergic cortical terminals, cholinergic interneurons, and dopaminergic terminals from substantia nigra where they suppress release of glutamate, acetylcholine and dopamine, respectively, and these cell types may contribute to early striatal dysfunction in HD. Thus, we used corticostriatal brain slices and optogenetic probes to directly investigate neuromodulatory signaling in the transgenic YAC128 HD mouse model. RESULTS Low-dose D2 agonist quinpirole reduced cortically-evoked glutamate release in dorsal striatum of premanifest YAC128 slices but not WT, and blocking type 1 cannabinoid receptors mitigated this effect. YAC128 corticostriatal brain slices also showed increased evoked dopamine and reduced evoked eCB release compared to WT, while acetylcholine signaling patterns remained relatively intact. CONCLUSIONS These findings suggest that YAC128 corticostriatal slices show increased D2 sensitivity that is eCB-dependent, and that dopamine and eCB release are altered at an early disease stage. We provide evidence for impaired neuromodulatory signaling in early HD, guiding therapeutic efforts prior to the onset of overt motor symptoms later on.
Collapse
Affiliation(s)
- Judy Cheng
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Ellen T Koch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Daniel Ramandi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Cell and Developmental Biology, University of British Columbia, Vancouver, BC, Canada
| | - James P Mackay
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Timothy P O'Leary
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - William Rees-Jones
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
2
|
Chang CP, Wu CW, Chern Y. Metabolic dysregulation in Huntington's disease: Neuronal and glial perspectives. Neurobiol Dis 2024; 201:106672. [PMID: 39306013 DOI: 10.1016/j.nbd.2024.106672] [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: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a mutant huntingtin protein with an abnormal CAG/polyQ expansion in the N-terminus of HTT exon 1. HD is characterized by progressive neurodegeneration and metabolic abnormalities, particularly in the brain, which accounts for approximately 20 % of the body's resting metabolic rate. Dysregulation of energy homeostasis in HD includes impaired glucose transporters, abnormal functions of glycolytic enzymes, changes in tricarboxylic acid (TCA) cycle activity and enzyme expression in the basal ganglia and cortical regions of both HD mouse models and HD patients. However, current understanding of brain cell behavior during energy dysregulation and its impact on neuron-glia crosstalk in HD remains limited. This review provides a comprehensive summary of the current understanding of the differences in glucose metabolism between neurons and glial cells in HD and how these differences contribute to disease development compared with normal conditions. We also discuss the potential impact of metabolic shifts on neuron-glia communication in HD. A deeper understanding of these metabolic alterations may reveal potential therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Wen Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
3
|
Robbins E, Wong B, Pwint MY, Salavatian S, Mahajan A, Cui XT. Improving Sensitivity and Longevity of In Vivo Glutamate Sensors with Electrodeposited NanoPt. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40570-40580. [PMID: 39078097 PMCID: PMC11310907 DOI: 10.1021/acsami.4c06692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
In vivo glutamate sensing has provided valuable insight into the physiology and pathology of the brain. Electrochemical glutamate biosensors, constructed by cross-linking glutamate oxidase onto an electrode and oxidizing H2O2 as a proxy for glutamate, are the gold standard for in vivo glutamate measurements for many applications. While glutamate sensors have been employed ubiquitously for acute measurements, there are almost no reports of long-term, chronic glutamate sensing in vivo, despite demonstrations of glutamate sensors lasting for weeks in vitro. To address this, we utilized a platinum electrode with nanometer-scale roughness (nanoPt) to improve the glutamate sensors' sensitivity and longevity. NanoPt improved the GLU sensitivity by 67.4% and the sensors were stable in vitro for 3 weeks. In vivo, nanoPt glutamate sensors had a measurable signal above a control electrode on the same array for 7 days. We demonstrate the utility of the nanoPt sensors by studying the effect of traumatic brain injury on glutamate in the rat striatum with a flexible electrode array and report measurements of glutamate taken during the injury itself. We also show the flexibility of the nanoPt platform to be applied to other oxidase enzyme-based biosensors by measuring γ-aminobutyric acid in the porcine spinal cord. NanoPt is a simple, effective way to build high sensitivity, robust biosensors harnessing enzymes to detect neurotransmitters in vivo.
Collapse
Affiliation(s)
- Elaine
M. Robbins
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Benjamin Wong
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, United States
| | - May Yoon Pwint
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Center
for Neural Basis of Cognition, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Siamak Salavatian
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, United States
| | - Aman Mahajan
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, United States
| | - Xinyan Tracy Cui
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Center
for Neural Basis of Cognition, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United
States
| |
Collapse
|
4
|
Ye C, Zhou T, Deng Y, Wu S, Zeng T, Yang J, Shi YS, Yin Y, Li G. Enhanced performance of enzymes confined in biocatalytic hydrogen-bonded organic frameworks for sensing of glutamate in the central nervous system. Biosens Bioelectron 2024; 247:115963. [PMID: 38147717 DOI: 10.1016/j.bios.2023.115963] [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: 10/22/2023] [Revised: 12/07/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Glutamate (Glu) is a key excitatory neurotransmitter associated with various neurological disorders in the central nervous system, so its measurement is vital to both basic research and biomedical application. In this work, we propose the first example of using biocatalytic hydrogen-bonded organic frameworks (HOFs) as the hosting matrix to encapsulate glutamate oxidase (GLOD) via a de novo approach, fabricating a cascaded-enzyme nanoreactor for Glu biosensing. In this design, the ferriporphyrin ligands can assemble to form Fe-HOFs with high catalase-like activity, while offering a scaffold for the in-situ immobilization of GLOD. Moreover, the formed GLOD@Fe-HOFs are favorable for the efficient diffusion of Glu into the active sites of GLOD via the porous channels, accelerating the cascade reaction with neighboring Fe-HOFs. Consequently, the constructed nanoreactor can offer superior activity and operational stability in the catalytic cascade for Glu biosensing. More importantly, rapid and selective detection can be achieved in the cerebrospinal fluid (CSF) collected from mice in a low sample consumption. Therefore, the successful fabrication of enzyme@HOFs may offer promise to develop high-performance biosensor for further biomedical applications.
Collapse
Affiliation(s)
- Chang Ye
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School, Nanjing University, Nanjing, 210032, PR China
| | - Tianci Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Shuai Wu
- Women & Children Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Tianyu Zeng
- Women & Children Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Jie Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School, Nanjing University, Nanjing, 210032, PR China.
| | - Yongmei Yin
- Women & Children Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
5
|
McGarry A, Hunter K, Gaughan J, Auinger P, Ferraro TN, Pradhan B, Ferrucci L, Egan JM, Moaddel R. An exploratory metabolomic comparison of participants with fast or absent functional progression from 2CARE, a randomized, double-blind clinical trial in Huntington's disease. Sci Rep 2024; 14:1101. [PMID: 38212353 PMCID: PMC10784537 DOI: 10.1038/s41598-023-50553-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024] Open
Abstract
Huntington's disease (HD) is increasingly recognized for diverse pathology outside of the nervous system. To describe the biology of HD in relation to functional progression, we previously analyzed the plasma and CSF metabolome in a cross-sectional study of participants who had various degrees of functional impairment. Here, we carried out an exploratory study in plasma from HD individuals over a 3-year time frame to assess whether differences exist between those with fast or absent clinical progression. There were more differences in circulating metabolite levels for fast progressors compared to absent progressors (111 vs 20, nominal p < 0.05). All metabolite changes in faster progressors were decreases, whereas some metabolite concentrations increased in absent progressors. Many of the metabolite levels that decreased in the fast progressors were higher at Screening compared to absent progressors but ended up lower by Year 3. Changes in faster progression suggest greater oxidative stress and inflammation (kynurenine, diacylglycerides, cysteine), disturbances in nitric oxide and urea metabolism (arginine, citrulline, ornithine, GABR), lower polyamines (putrescine and spermine), elevated glucose, and deficient AMPK signaling. Metabolomic differences between fast and absent progressors suggest the possibility of predicting functional decline in HD, and possibly delaying it with interventions to augment arginine, polyamines, and glucose regulation.
Collapse
Affiliation(s)
- Andrew McGarry
- Department of Neurology, Cooper University Hospital and Cooper Medical School at Rowan University, Camden, NJ, USA.
| | - Krystal Hunter
- Department of Medicine, Cooper Medical School at Rowan University, Camden, NJ, USA
| | - John Gaughan
- Department of Neurology, Cooper University Hospital and Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Peggy Auinger
- Department of Neurology, Center for Health and Technology, University of Rochester, Rochester, NY, USA
| | - Thomas N Ferraro
- Department of Biomedical Sciences, Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Basant Pradhan
- Department of Psychiatry, Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Luigi Ferrucci
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Josephine M Egan
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
6
|
Wang J, Wang L, Yang Y, Li H, Huang X, Liu Z, Yu S, Tang C, Chen J, Shi X, Li W, Chen P, Tong Q, Yu H, Sun X, Peng H. A Fiber Sensor for Long-Term Monitoring of Extracellular Potassium Ion Fluctuations in Chronic Neuropsychiatric Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2309862. [PMID: 38133487 DOI: 10.1002/adma.202309862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/22/2023] [Indexed: 12/23/2023]
Abstract
The extracellular potassium ion concentration in the brain exerts a significant influence on cellular excitability and intercellular communication. Perturbations in the extracellular potassium ion level are closely correlated with various chronic neuropsychiatric disorders including depression. However, a critical gap persists in performing real-time and long-term monitoring of extracellular potassium ions, which is necessary for comprehensive profiling of chronic neuropsychiatric diseases. Here, a fiber potassium ion sensor (FKS) that consists of a soft conductive fiber with a rough surface and a hydrophobic-treated transduction layer interfaced with a potassium ion-selective membrane is found to solve this problem. The FKS demonstrates stable interfaces between its distinct functional layers in an aqueous environment, conferring an exceptional stability of 6 months in vivo, in stark contrast to previous reports with working durations from hours to days. Upon implantation into the mouse brain, the FKS enables effective monitoring of extracellular potassium ion dynamics under diverse physiological states including anesthesia, forced swimming, and tail suspension. Using this FKS, tracking of extracellular potassium ion fluctuations that align with behaviors associated with the progression of depression over months is achieved, demonstrating its usability in studying chronic neuropsychiatric disorders from a new biochemical perspective.
Collapse
Affiliation(s)
- Jiajia Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Liyuan Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Yiqing Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - HongJian Li
- Vision Research Laboratory, School of Life Sciences, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200438, China
| | - Xinlin Huang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Ziwei Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Sihui Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Chengqiang Tang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Jiawei Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Xiang Shi
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Wenjun Li
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Peining Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Qi Tong
- Department of Aeronautics and Astronautics, Fudan University, Shanghai, 200433, China
| | - Hongbo Yu
- Vision Research Laboratory, School of Life Sciences, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200438, China
| | - Xuemei Sun
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| | - Huisheng Peng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Institute of Fiber Materials and Devices, and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438, China
| |
Collapse
|
7
|
Nithianandam P, Tzu-li L, Chen S, Yizhen J, Dong Y, Saul M, Tedeschi A, Wenjing S, Jinghua L. Flexible, Miniaturized Sensing Probes Inspired by Biofuel Cells for Monitoring Synaptically Released Glutamate in the Mouse Brain. Angew Chem Int Ed Engl 2023; 62:e202310245. [PMID: 37632702 PMCID: PMC10592105 DOI: 10.1002/anie.202310245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 08/28/2023]
Abstract
Chemical biomarkers in the central nervous system can provide valuable quantitative measures to gain insight into the etiology and pathogenesis of neurological diseases. Glutamate, one of the most important excitatory neurotransmitters in the brain, has been found to be upregulated in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, stroke, epilepsy, chronic pain, and migraines. However, quantitatively monitoring glutamate release in situ has been challenging. This work presents a novel class of flexible, miniaturized probes inspired by biofuel cells for monitoring synaptically released glutamate in the nervous system. The resulting sensors, with dimensions as low as 50 by 50 μm, can detect real-time changes in glutamate within the biologically relevant concentration range. Experiments exploiting the hippocampal circuit in mice models demonstrate the capability of the sensors in monitoring glutamate release via electrical stimulation using acute brain slices. These advances could aid in basic neuroscience studies and translational engineering, as the sensors provide a diagnostic tool for neurological disorders. Additionally, adapting the biofuel cell design to other neurotransmitters can potentially enable the detailed study of the effect of neurotransmitter dysregulation on neuronal cell signaling pathways and revolutionize neuroscience.
Collapse
Affiliation(s)
- Prasad Nithianandam
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Liu Tzu-li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Shulin Chen
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jia Yizhen
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Yan Dong
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Morgan Saul
- Department of Neuroscience, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Andrea Tedeschi
- Department of Neuroscience, The Ohio State University College of Medicine, Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| | - Sun Wenjing
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Li Jinghua
- Department of Materials Science and Engineering, Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
8
|
Bhatnagar A, Parmar V, Barbieri N, Bearoff F, Elefant F, Kortagere S. Novel EAAT2 activators improve motor and cognitive impairment in a transgenic model of Huntington's disease. Front Behav Neurosci 2023; 17:1176777. [PMID: 37351153 PMCID: PMC10282606 DOI: 10.3389/fnbeh.2023.1176777] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Glutamate excitotoxicity is causal in striatal neurodegeneration underlying motor dysfunction and cognitive deficits in Huntington's disease (HD). Excitatory amino acid transporter 2 (EAAT2), the predominant glutamate transporter accounting for >90% of glutamate transport, plays a key role in preventing excitotoxicity by clearing excess glutamate from the intrasynaptic cleft. Accordingly, EAAT2 has emerged as a promising therapeutic target for prevention of neuronal excitotoxicity underlying HD and other neurodegenerative diseases. Methods We have previously designed novel EAAT2 positive allosteric modulator GT951, GTS467, and GTS551, with low nanomolar efficacy in glutamate uptake and favorable pharmacokinetic properties. In this study, we test the neuroprotective abilities of these novel EAAT2 activators in vivo using the robust Drosophila HD transgenic model expressing human huntingtin gene with expanded repeats (Htt128Q). Results All three compounds significantly restored motor function impaired under HD pathology over a wide dose range. Additionally, treatment with all three compounds significantly improved HD-associated olfactory associative learning and short-term memory defects, while GT951 and GTS551 also improved middle-term memory in low-performing group. Similarly, treatment with GT951 and GTS551 partially protected against early mortality observed in our HD model. Further, treatment with all three EAAT2 activators induced epigenetic expression of EAAT2 Drosophila homolog and several cognition-associated genes. Conclusion Together, these results highlight the efficacy of GT951, GTS467 and GTS551 in treating motor and cognitive impairments under HD pathology and support their development for treatment of HD.
Collapse
Affiliation(s)
- Akanksha Bhatnagar
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
| | - Visha Parmar
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nicholas Barbieri
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Frank Bearoff
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Felice Elefant
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
9
|
Mackay JP, Smith-Dijak AI, Koch ET, Zhang P, Fung E, Nassrallah WB, Buren C, Schmidt M, Hayden MR, Raymond LA. Axonal ER Ca 2+ Release Selectively Enhances Activity-Independent Glutamate Release in a Huntington Disease Model. J Neurosci 2023; 43:3743-3763. [PMID: 36944490 PMCID: PMC10198457 DOI: 10.1523/jneurosci.1593-22.2023] [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: 08/19/2022] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
Action potential (AP)-independent (miniature) neurotransmission occurs at all chemical synapses but remains poorly understood, particularly in pathologic contexts. Axonal endoplasmic reticulum (ER) Ca2+ stores are thought to influence miniature neurotransmission, and aberrant ER Ca2+ handling is implicated in progression of Huntington disease (HD). Here, we report elevated mEPSC frequencies in recordings from YAC128 mouse (HD-model) neurons (from cortical cultures and striatum-containing brain slices, both from male and female animals). Pharmacological experiments suggest that this is mediated indirectly by enhanced tonic ER Ca2+ release. Calcium imaging, using an axon-localized sensor, revealed slow AP-independent ER Ca2+ release waves in both YAC128 and WT cultures. These Ca2+ waves occurred at similar frequencies in both genotypes but spread less extensively and were of lower amplitude in YAC128 axons, consistent with axonal ER Ca2+ store depletion. Surprisingly, basal cytosolic Ca2+ levels were lower in YAC128 boutons and YAC128 mEPSCs were less sensitive to intracellular Ca2+ chelation. Together, these data suggest that elevated miniature glutamate release in YAC128 cultures is associated with axonal ER Ca2+ depletion but not directly mediated by ER Ca2+ release into the cytoplasm. In contrast to increased mEPSC frequencies, cultured YAC128 cortical neurons showed less frequent AP-dependent (spontaneous) Ca2+ events in soma and axons, although evoked glutamate release detected by an intensity-based glutamate-sensing fluorescence reporter in brain slices was similar between genotypes. Our results indicate that axonal ER dysfunction selectively elevates miniature glutamate release from cortical terminals in HD. This, together with reduced spontaneous cortical neuron firing, may cause a shift from activity-dependent to -independent glutamate release in HD, with potential implications for fidelity and plasticity of cortical excitatory signaling.SIGNIFICANCE STATEMENT Miniature neurotransmitter release persists at all chemical neuronal synapses in the absence of action potential firing but remains poorly understood, particularly in disease states. We show enhanced miniature glutamate release from cortical neurons in the YAC128 mouse Huntington disease model. This effect is mediated by axonal ER Ca2+ store depletion, but is not obviously due to elevated ER-to-cytosol Ca2+ release. Conversely, YAC128 cortical pyramidal neurons fired fewer action potentials and evoked cortical glutamate release was similar between WT an YAC128 preparations, indicating axonal ER depletion selectively enhances miniature glutamate release in YAC128 mice. These results extend our understanding of action potential independent neurotransmission and highlight a potential involvement of elevated miniature glutamate release in Huntington disease pathology.
Collapse
Affiliation(s)
- James P Mackay
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
| | - Amy I Smith-Dijak
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Graduate Program in Neuroscience
| | - Ellen T Koch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Graduate Program in Neuroscience
| | - Peng Zhang
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Evan Fung
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
| | - Wissam B Nassrallah
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- MD/PhD Program
| | - Caodu Buren
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Graduate Program in Neuroscience
| | - Mandi Schmidt
- Graduate Program in Neuroscience
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
| |
Collapse
|
10
|
Giangrasso DM, Veros KM, Timm MM, West PJ, Wilcox KS, Keefe KA. Glutamate dynamics in the dorsolateral striatum of rats with goal-directed and habitual cocaine-seeking behavior. Front Mol Neurosci 2023; 16:1160157. [PMID: 37251646 PMCID: PMC10213946 DOI: 10.3389/fnmol.2023.1160157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/17/2023] [Indexed: 05/31/2023] Open
Abstract
The shift from drug abuse to addiction is considered to arise from the transition between goal-directed and habitual control over drug behavior. Habitual responding for appetitive and skill-based behaviors is mediated by potentiated glutamate signaling in the dorsolateral striatum (DLS), but the state of the DLS glutamate system in the context of habitual drug-behavior remains undefined. Evidence from the nucleus accumbens of cocaine-experienced rats suggests that decreased transporter-mediated glutamate clearance and enhanced synaptic glutamate release contribute to the potentiated glutamate signaling that underlies the enduring vulnerability to relapse. Preliminary evidence from the dorsal striatum of cocaine-experienced rats suggests that this region exhibits similar alterations to glutamate clearance and release, but it is not known whether these glutamate dynamics are associated with goal-directed or habitual control over cocaine-seeking behavior. Therefore, we trained rats to self-administer cocaine in a chained cocaine-seeking and -taking paradigm, which yielded goal-directed, intermediate, and habitual cocaine-seeking rats. We then assessed glutamate clearance and release dynamics in the DLS of these rats using two different methods: synaptic transporter current (STC) recordings of patch-clamped astrocytes and the intensity-based glutamate sensing fluorescent reporter (iGluSnFr). While we observed a decreased rate of glutamate clearance in STCs evoked with single-pulse stimulation in cocaine-experienced rats, we did not observe any cocaine-induced differences in glutamate clearance rates from STCs evoked with high frequency stimulation (HFS) or iGluSnFr responses evoked with either double-pulse stimulation or HFS. Furthermore, GLT-1 protein expression in the DLS was unchanged in cocaine-experienced rats, regardless of their mode of control over cocaine-seeking behavior. Lastly, there were no differences in metrics of glutamate release between cocaine-experienced rats and yoked-saline controls in either assay. Together, these results suggest that glutamate clearance and release dynamics in the DLS are largely unaltered by a history of cocaine self-administration on this established cocaine seeking-taking paradigm, regardless of whether the control over the cocaine seeking behavior was habitual or goal directed.
Collapse
Affiliation(s)
- Danielle M. Giangrasso
- Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, United States
| | - Kaliana M. Veros
- Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, United States
| | - Maureen M. Timm
- Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Peter J. West
- Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, United States
- Anticonvulsant Drug Development Program, Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Karen S. Wilcox
- Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, United States
- Anticonvulsant Drug Development Program, Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Kristen A. Keefe
- Department of Pharmacology & Toxicology, University of Utah, Salt Lake City, UT, United States
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
11
|
Abstract
Huntington's disease (HD) is a fatal, monogenic, autosomal dominant neurodegenerative disease caused by a polyglutamine-encoding CAG expansion in the huntingtin (HTT) gene that results in mutant huntingtin proteins (mHTT) in cells throughout the body. Although large parts of the central nervous system (CNS) are affected, the striatum is especially vulnerable and undergoes marked atrophy. Astrocytes are abundant within the striatum and contain mHTT in HD, as well as in mouse models of the disease. We focus on striatal astrocytes and summarize how they participate in, and contribute to, molecular pathophysiology and disease-related phenotypes in HD model mice. Where possible, reference is made to pertinent astrocyte alterations in human HD. Astrocytic dysfunctions related to cellular morphology, extracellular ion and neurotransmitter homeostasis, and metabolic support all accompany the development and progression of HD, in both transgenic mouse and human cellular and chimeric models of HD. These findings reveal the potential for the therapeutic targeting of astrocytes so as to restore synaptic as well as tissue homeostasis in HD. Elucidation of the mechanisms by which astrocytes contribute to HD pathogenesis may inform a broader understanding of the role of glial pathology in neurodegenerative disorders and, by so doing, enable new strategies of glial-directed therapeutics.
Collapse
Affiliation(s)
- Baljit S. Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Steven A. Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
12
|
Alkahtani S, AL-Johani NS, Alarifi S. Mechanistic Insights, Treatment Paradigms, and Clinical Progress in Neurological Disorders: Current and Future Prospects. Int J Mol Sci 2023; 24:1340. [PMID: 36674852 PMCID: PMC9865061 DOI: 10.3390/ijms24021340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Neurodegenerative diseases (NDs) are a major cause of disability and are related to brain development. The neurological signs of brain lesions can vary from mild clinical shortfalls to more delicate and severe neurological/behavioral symptoms and learning disabilities, which are progressive. In this paper, we have tried to summarize a collective view of various NDs and their possible therapeutic outcomes. These diseases often occur as a consequence of the misfolding of proteins post-translation, as well as the dysfunctional trafficking of proteins. In the treatment of neurological disorders, a challenging hurdle to cross regarding drug delivery is the blood-brain barrier (BBB). The BBB plays a unique role in maintaining the homeostasis of the central nervous system (CNS) by exchanging components between the circulations and shielding the brain from neurotoxic pathogens and detrimental compounds. Here, we outline the current knowledge about BBB deterioration in the evolving brain, its origin, and therapeutic interventions. Additionally, we summarize the physiological scenarios of the BBB and its role in various cerebrovascular diseases. Overall, this information provides a detailed account of BBB functioning and the development of relevant treatments for neurological disorders. This paper will definitely help readers working in the field of neurological scientific communities.
Collapse
Affiliation(s)
- Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | | | | |
Collapse
|
13
|
Koch ET, Sepers MD, Cheng J, Raymond LA. Early Changes in Striatal Activity and Motor Kinematics in a Huntington's Disease Mouse Model. Mov Disord 2022; 37:2021-2032. [PMID: 35880748 PMCID: PMC9796416 DOI: 10.1002/mds.29168] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 06/30/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Huntington's disease is a progressive neurodegenerative disorder with no disease-modifying treatments. Patients experience motor, cognitive, and psychiatric disturbances, and the dorsal striatum is the main target of neurodegeneration. Mouse models of Huntington's disease show altered striatal synaptic signaling in vitro, but how these changes relate to behavioral deficits in vivo is unclear. OBJECTIVES We aimed to investigate how striatal activity correlates with behavior in vivo during motor learning and spontaneous behavior in a Huntington's disease mouse model at two disease stages. METHODS We used fiber photometry to record jGCaMP7f fluorescence, a read-out of neuronal activity, in the dorsal striatum of YAC128 (yeast artificial chromosome-128CAG) mice during accelerating rotarod and open-field behavior. RESULTS Mice showed increased striatal activity on the rotarod, which diminished by late stages of learning, leading to an inverse correlation between latency to fall and striatal activity. The 2- to 3-month-old YAC128 mice did not show a deficit in latency to fall, but displayed significant differences in paw kinematics, including increased paw slip frequency and variability in paw height. These mice exhibited a weaker correlation between latency to fall and striatal activity and aberrant striatal activity during paw slips. At 6 to 7 months, the YAC128 mice showed significantly reduced latency to fall, impaired paw kinematics, and increased striatal activity while on the rotarod. In the open field, the YAC128 mice showed elevated neuronal activity at rest. CONCLUSIONS We uncovered impaired motor coordination at a stage thought to be premotor manifest in YAC128 mice and aberrant striatal activity during the accelerating rotarod and open-field exploration. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ellen T. Koch
- Department of PsychiatryDjavad Mowafaghian Centre for Brain Health, University of British ColumbiaVancouverBCCanada,Graduate Program in NeuroscienceUniversity of British ColumbiaVancouverBCCanada
| | - Marja D. Sepers
- Department of PsychiatryDjavad Mowafaghian Centre for Brain Health, University of British ColumbiaVancouverBCCanada
| | - Judy Cheng
- Department of PsychiatryDjavad Mowafaghian Centre for Brain Health, University of British ColumbiaVancouverBCCanada,Graduate Program in NeuroscienceUniversity of British ColumbiaVancouverBCCanada
| | - Lynn A. Raymond
- Department of PsychiatryDjavad Mowafaghian Centre for Brain Health, University of British ColumbiaVancouverBCCanada
| |
Collapse
|
14
|
Abulseoud OA, Alasmari F, Hussein AM, Sari Y. Ceftriaxone as a Novel Therapeutic Agent for Hyperglutamatergic States: Bridging the Gap Between Preclinical Results and Clinical Translation. Front Neurosci 2022; 16:841036. [PMID: 35864981 PMCID: PMC9294323 DOI: 10.3389/fnins.2022.841036] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulation of glutamate homeostasis is a well-established core feature of neuropsychiatric disorders. Extracellular glutamate concentration is regulated by glutamate transporter 1 (GLT-1). The discovery of a beta-lactam antibiotic, ceftriaxone (CEF), as a safe compound with unique ability to upregulate GLT-1 sparked the interest in testing its efficacy as a novel therapeutic agent in animal models of neuropsychiatric disorders with hyperglutamatergic states. Indeed, more than 100 preclinical studies have shown the efficacy of CEF in attenuating the behavioral manifestations of various hyperglutamatergic brain disorders such as ischemic stroke, amyotrophic lateral sclerosis (ALS), seizure, Huntington’s disease, and various aspects of drug use disorders. However, despite rich and promising preclinical data, only one large-scale clinical trial testing the efficacy of CEF in patients with ALS is reported. Unfortunately, in that study, there was no significant difference in survival between placebo- and CEF-treated patients. In this review, we discussed the translational potential of preclinical efficacy of CEF based on four different parameters: (1) initiation of CEF treatment in relation to induction of the hyperglutamatergic state, (2) onset of response in preclinical models in relation to onset of GLT-1 upregulation, (3) mechanisms of action of CEF on GLT-1 expression and function, and (4) non-GLT-1-mediated mechanisms for CEF. Our detailed review of the literature brings new insights into underlying molecular mechanisms correlating the preclinical efficacy of CEF. We concluded here that CEF may be clinically effective in selected cases in acute and transient hyperglutamatergic states such as early drug withdrawal conditions.
Collapse
Affiliation(s)
- Osama A. Abulseoud
- Department of Psychiatry and Psychology, Alex School of Medicine at Mayo Clinic, Phoenix, AZ, United States
- *Correspondence: Osama A. Abulseoud,
| | - Fawaz Alasmari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Youssef Sari,
| |
Collapse
|
15
|
Brandner S, Aicher S, Schroeter S, Swierzy I, Kinfe TM, Buchfelder M, Maslarova A, Stadlbauer A. Real-time imaging of glutamate transients in the extracellular space of acute human brain slices using a single-wavelength glutamate fluorescence nanosensor. Sci Rep 2022; 12:3926. [PMID: 35273260 PMCID: PMC8913701 DOI: 10.1038/s41598-022-07940-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glutamate is the most important excitatory neurotransmitter in the brain. The ability to assess glutamate release and re-uptake with high spatial and temporal resolution is crucial to understand the involvement of this primary excitatory neurotransmitter in both normal brain function and different neurological disorders. Real-time imaging of glutamate transients by fluorescent nanosensors has been accomplished in rat brain slices. We performed for the first time single-wavelength glutamate nanosensor imaging in human cortical brain slices obtained from patients who underwent epilepsy surgery. The glutamate fluorescence nanosensor signals of the electrically stimulated human cortical brain slices showed steep intensity increase followed by an exponential decrease. The spatial distribution and the time course of the signal were in good agreement with the position of the stimulation electrode and the dynamics of the electrical stimulation, respectively. Pharmacological manipulation of glutamate release and reuptake was associated with corresponding changes in the glutamate fluorescence nanosensor signals. We demonstrated that the recently developed fluorescent nanosensors for glutamate allow to detect neuronal activity in acute human cortical brain slices with high spatiotemporal precision. Future application to tissue samples from different pathologies may provide new insights into pathophysiology without the limitations of an animal model.
Collapse
Affiliation(s)
- Sebastian Brandner
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Simon Aicher
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Sarah Schroeter
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany.,Center for Musculoskeletal Surgery Osnabrück (OZMC), Klinikum Osnabrück, Osnabrück, Germany
| | - Izabela Swierzy
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Thomas M Kinfe
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany.,Division of Functional Neurosurgery and Stereotaxy, University Hospital Erlangen, Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Anna Maslarova
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Andreas Stadlbauer
- Department of Neurosurgery, University Hospital Erlangen, Schwabachanlage 6, 91054, Erlangen, Germany.,Institute of Medical Radiology, University Clinic St. Pölten, Karl Landsteiner University of Health Sciences, St. Pölten, Austria
| |
Collapse
|
16
|
Hao Y, Plested AJ. Seeing glutamate at central synapses. J Neurosci Methods 2022; 375:109531. [DOI: 10.1016/j.jneumeth.2022.109531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
|
17
|
Hirschberg S, Dvorzhak A, Rasooli-Nejad SMA, Angelov S, Kirchner M, Mertins P, Lättig-Tünnemann G, Harms C, Schmitz D, Grantyn R. Uncoupling the Excitatory Amino Acid Transporter 2 From Its C-Terminal Interactome Restores Synaptic Glutamate Clearance at Corticostriatal Synapses and Alleviates Mutant Huntingtin-Induced Hypokinesia. Front Cell Neurosci 2022; 15:792652. [PMID: 35173582 PMCID: PMC8841566 DOI: 10.3389/fncel.2021.792652] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/21/2021] [Indexed: 02/05/2023] Open
Abstract
Rapid removal of glutamate from the sites of glutamate release is an essential step in excitatory synaptic transmission. However, despite many years of research, the molecular mechanisms underlying the intracellular regulation of glutamate transport at tripartite synapses have not been fully uncovered. This limits the options for pharmacological treatment of glutamate-related motor disorders, including Huntington’s disease (HD). We therefore investigated the possible binding partners of transgenic EAAT2 and their alterations under the influence of mutant huntingtin (mHTT). Mass spectrometry analysis after pull-down of striatal YFP-EAAT2 from wild-type (WT) mice and heterozygote (HET) Q175 mHTT-knock-in mice identified a total of 148 significant (FDR < 0.05) binders to full-length EAAT2. Of them 58 proteins exhibited mHTT-related differences. Most important, in 26 of the 58 mHTT-sensitive cases, protein abundance changed back toward WT levels when the mice expressed a C-terminal-truncated instead of full-length variant of EAAT2. These findings motivated new attempts to clarify the role of astrocytic EAAT2 regulation in cortico-basal movement control. Striatal astrocytes of Q175 HET mice were targeted by a PHP.B vector encoding EAAT2 with different degree of C-terminal modification, i.e., EAAT2-S506X (truncation at S506), EAAT2-4KR (4 lysine to arginine substitutions) or EAAT2 (full-length). The results were compared to HET and WT injected with a tag-only vector (CTRL). It was found that the presence of a C-terminal-modified EAAT2 transgene (i) increased the level of native EAAT2 protein in striatal lysates and perisynaptic astrocyte processes, (ii) enhanced the glutamate uptake of transduced astrocytes, (iii) stimulated glutamate clearance at individual corticostriatal synapses, (iv) increased the glutamate uptake of striatal astrocytes and (iv) alleviated the mHTT-related hypokinesia (open field indicators of movement initiation). In contrast, over-expression of full-length EAAT2 neither facilitated glutamate uptake nor locomotion. Together, our results support the new hypothesis that preventing abnormal protein-protein interactions at the C-terminal of EAAT2 could eliminate the mHTT-related deficits in corticostriatal synaptic glutamate clearance and movement initiation.
Collapse
Affiliation(s)
- Stefan Hirschberg
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anton Dvorzhak
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Seyed M. A. Rasooli-Nejad
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Svilen Angelov
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Marieluise Kirchner
- Proteomics Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Gilla Lättig-Tünnemann
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Cluster of Excellence NeuroCure, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Rosemarie Grantyn
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Cluster of Excellence NeuroCure, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- *Correspondence: Rosemarie Grantyn,
| |
Collapse
|
18
|
Mei Y, Zhang QW, Gu Q, Liu Z, He X, Tian Y. Pillar[5]arene-Based Fluorescent Sensor Array for Biosensing of Intracellular Multi-neurotransmitters through Host-Guest Recognitions. J Am Chem Soc 2022; 144:2351-2359. [PMID: 35099950 DOI: 10.1021/jacs.1c12959] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neurotransmitters are very important for neuron events and brain diseases. However, effective probes for analyzing specific neurotransmitters are currently lacking. Herein, we design and create a supramolecular fluorescent probe (CN-DFP5) by synthesizing a dual-functionalized fluorescent pillar[5]arene derivative with borate naphthalene and aldehyde coumarin recognition groups to identify large-scale neurotransmitters. The developed probe can detect seven model neurotransmitters by generating different fluorescence patterns through three types of host-guest interactions. The obtained signals are statistically processed by principal component analysis, thus the high-throughput analysis of neurotransmitters is realized under dual-channel fluorescence responses. The present probe combines the advantages of small-molecule-based probes to easily enter into living neurons and cross-reactive sensor arrays. Thus, the selective binding enables this probe to identify specific neurotransmitters in biofluids, living neurons, and tissues. High selectivity and sensitivity further demonstrate that the molecular device could extend to more applications to detect and image neurotransmitters.
Collapse
Affiliation(s)
- Yuxiao Mei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Qi-Wei Zhang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Qingyi Gu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Zhichao Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Xiao He
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P.R. China
| |
Collapse
|
19
|
Parrot S, Corscadden A, Lallemant L, Benyamine H, Comte JC, Huguet-Lachon A, Gourdon G, Gomes-Pereira M. Defects in Mouse Cortical Glutamate Uptake Can Be Unveiled In Vivo by a Two-in-One Quantitative Microdialysis. ACS Chem Neurosci 2022; 13:134-142. [PMID: 34923816 DOI: 10.1021/acschemneuro.1c00634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extracellular glutamate levels are maintained low by efficient transporters, whose dysfunction can cause neuronal hyperexcitability, excitotoxicity, and neurological disease. While many methods estimate glutamate uptake in vitro/ex vivo, a limited number of techniques address glutamate transport in vivo. Here, we used in vivo microdialysis in a two-in-one approach combining reverse dialysis of isotopic glutamate to measure uptake ability and zero-flow (ZF) methods to quantify extracellular glutamate levels. The complementarity of both techniques is discussed on methodological and anatomical basis. We used a transgenic mouse model of human disease, expressing low levels of the EAAT-2/GLT1 glutamate transporter, to validate our approach in a relevant animal model. As expected, isotopic analysis revealed an overall decrease in glutamate uptake, while the ZF method unveiled higher extracellular glutamate levels in these mice. We propose a sensitive and expedite two-in-one microdialysis approach that is sufficiently robust to reveal significant differences in neurotransmitter uptake and extracellular levels through the analysis of a relatively low number of animals.
Collapse
Affiliation(s)
- Sandrine Parrot
- Inserm U1028, CNRS UMR5292, Université de Lyon, Lyon Neuroscience Research Center, Bron F-69500, France
| | - Alex Corscadden
- Inserm U1028, CNRS UMR5292, Université de Lyon, Lyon Neuroscience Research Center, Bron F-69500, France
| | - Louison Lallemant
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Hélène Benyamine
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Jean-Christophe Comte
- Inserm U1028, CNRS UMR5292, Université de Lyon, Lyon Neuroscience Research Center, Bron F-69500, France
| | - Aline Huguet-Lachon
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Geneviève Gourdon
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Mário Gomes-Pereira
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| |
Collapse
|
20
|
Abstract
Glutamatergic neurotransmission is a widespread form of synaptic excitation in the mammalian brain. The development of genetically encoded fluorescent glutamate sensors allows monitoring synaptic signaling in living brain tissue in real time. Here, we describe single- and two-photon imaging of synaptically evoked glutamatergic population signals in acute hippocampal slices expressing the fluorescent glutamate sensor SF-iGluSnFR.A184S in CA1 or CA3 pyramidal neurons. The protocol can be readily used to study defective synaptic glutamate signaling in mouse models of neuropsychiatric disorders, such as Alzheimer disease. For complete details on the use and execution of this protocol, please refer to Zott et al. (2019).
Collapse
|
21
|
Jara CP, de Andrade Berti B, Mendes NF, Engel DF, Zanesco AM, Pereira de Souza GF, de Medeiros Bezerra R, de Toledo Bagatin J, Maria-Engler SS, Morari J, Velander WH, Velloso LA, Araújo EP. Glutamic acid promotes hair growth in mice. Sci Rep 2021; 11:15453. [PMID: 34326383 PMCID: PMC8322389 DOI: 10.1038/s41598-021-94816-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Glutamic acid is the main excitatory neurotransmitter acting both in the brain and in peripheral tissues. Abnormal distribution of glutamic acid receptors occurs in skin hyperproliferative conditions such as psoriasis and skin regeneration; however, the biological function of glutamic acid in the skin remains unclear. Using ex vivo, in vivo and in silico approaches, we showed that exogenous glutamic acid promotes hair growth and keratinocyte proliferation. Topical application of glutamic acid decreased the expression of genes related to apoptosis in the skin, whereas glutamic acid increased cell viability and proliferation in human keratinocyte cultures. In addition, we identified the keratinocyte glutamic acid excitotoxic concentration, providing evidence for the existence of a novel skin signalling pathway mediated by a neurotransmitter that controls keratinocyte and hair follicle proliferation. Thus, glutamic acid emerges as a component of the peripheral nervous system that acts to control cell growth in the skin. These results raise the perspective of the pharmacological and nutritional use of glutamic acid to treat skin diseases.
Collapse
Affiliation(s)
- Carlos Poblete Jara
- Faculty of Nursing, University of Campinas, UNICAMP, Tessalia Vieira de Camargo St., 126, Campinas, SP, 13083-887, Brazil.
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil.
- University of Campinas, Campinas, Brazil.
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE, USA.
| | - Beatriz de Andrade Berti
- Faculty of Nursing, University of Campinas, UNICAMP, Tessalia Vieira de Camargo St., 126, Campinas, SP, 13083-887, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - Natália Ferreira Mendes
- Faculty of Nursing, University of Campinas, UNICAMP, Tessalia Vieira de Camargo St., 126, Campinas, SP, 13083-887, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - Daiane Fátima Engel
- Faculty of Medical Sciences, University of Campinas, UNICAMP, Campinas, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - Ariane Maria Zanesco
- Faculty of Medical Sciences, University of Campinas, UNICAMP, Campinas, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - Gabriela Freitas Pereira de Souza
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - Renan de Medeiros Bezerra
- Faculty of Nursing, University of Campinas, UNICAMP, Tessalia Vieira de Camargo St., 126, Campinas, SP, 13083-887, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - Julia de Toledo Bagatin
- School of Pharmaceutical Sciences, Clinical Chemistry and Toxicology Department, University of São Paulo, São Paulo, Brazil
| | - Silvya Stuchi Maria-Engler
- School of Pharmaceutical Sciences, Clinical Chemistry and Toxicology Department, University of São Paulo, São Paulo, Brazil
| | - Joseane Morari
- Faculty of Medical Sciences, University of Campinas, UNICAMP, Campinas, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - William H Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE, USA
| | - Lício A Velloso
- Faculty of Medical Sciences, University of Campinas, UNICAMP, Campinas, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| | - Eliana Pereira Araújo
- Faculty of Nursing, University of Campinas, UNICAMP, Tessalia Vieira de Camargo St., 126, Campinas, SP, 13083-887, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- University of Campinas, Campinas, Brazil
| |
Collapse
|
22
|
Belo do Nascimento I, Damblon J, Ingelbrecht C, Goursaud S, Massart M, Dumont A, Desmet N, Hermans E. Pharmacological evidence for the concept of spare glutamate transporters. Neurochem Int 2021; 149:105142. [PMID: 34314789 DOI: 10.1016/j.neuint.2021.105142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/09/2021] [Accepted: 07/22/2021] [Indexed: 01/30/2023]
Abstract
Through the efficient clearance of extracellular glutamate, high affinity astrocytic glutamate transporters constantly shape excitatory neurotransmission in terms of duration and spreading. Even though the glutamate transporter GLT-1 (also known as EAAT2/SLC1A2) is amongst the most abundant proteins in the mammalian brain, its density and activity are tightly regulated. In order to study the influence of changes in the expression of GLT-1 on glutamate uptake capacity, we have developed a model in HEK cells where the density of the transporter can be manipulated thanks to a tetracycline-inducible promoter. Exposing the cells to doxycycline concentration-dependently increased GLT-1 expression and substrate uptake velocity. However, beyond a certain level of induction, increasing the density of transporters at the cell surface failed to increase the maximal uptake. This suggested the progressive generation of a pool of spare transporters, a hypothesis that was further validated using the selective GLT-1 blocker WAY-213613 of which potency was influenced by the density of the transporters. The curve showing inhibition of uptake by increasing concentrations of WAY-213613 was indeed progressively rightward shifted when tested in cells where the transporter density was robustly induced. As largely documented in the context of cell-surface receptors, the existence of 'spare' glutamate transporters in the nervous tissue and particularly in astrocytes could impact on the consequences of physiological or pathological regulation of these transporters.
Collapse
Affiliation(s)
- Inês Belo do Nascimento
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Jonathan Damblon
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Caroline Ingelbrecht
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Stéphanie Goursaud
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Marion Massart
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Amélie Dumont
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Nathalie Desmet
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium.
| |
Collapse
|
23
|
Andersen JV, Markussen KH, Jakobsen E, Schousboe A, Waagepetersen HS, Rosenberg PA, Aldana BI. Glutamate metabolism and recycling at the excitatory synapse in health and neurodegeneration. Neuropharmacology 2021; 196:108719. [PMID: 34273389 DOI: 10.1016/j.neuropharm.2021.108719] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/01/2021] [Accepted: 07/13/2021] [Indexed: 02/08/2023]
Abstract
Glutamate is the primary excitatory neurotransmitter of the brain. Cellular homeostasis of glutamate is of paramount importance for normal brain function and relies on an intricate metabolic collaboration between neurons and astrocytes. Glutamate is extensively recycled between neurons and astrocytes in a process known as the glutamate-glutamine cycle. The recycling of glutamate is closely linked to brain energy metabolism and is essential to sustain glutamatergic neurotransmission. However, a considerable amount of glutamate is also metabolized and serves as a metabolic hub connecting glucose and amino acid metabolism in both neurons and astrocytes. Disruptions in glutamate clearance, leading to neuronal overstimulation and excitotoxicity, have been implicated in several neurodegenerative diseases. Furthermore, the link between brain energy homeostasis and glutamate metabolism is gaining attention in several neurological conditions. In this review, we provide an overview of the dynamics of synaptic glutamate homeostasis and the underlying metabolic processes with a cellular focus on neurons and astrocytes. In particular, we review the recently discovered role of neuronal glutamate uptake in synaptic glutamate homeostasis and discuss current advances in cellular glutamate metabolism in the context of Alzheimer's disease and Huntington's disease. Understanding the intricate regulation of glutamate-dependent metabolic processes at the synapse will not only increase our insight into the metabolic mechanisms of glutamate homeostasis, but may reveal new metabolic targets to ameliorate neurodegeneration.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Kia H Markussen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Emil Jakobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Paul A Rosenberg
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
24
|
Boender AJ, Bontempi L, Nava L, Pelloux Y, Tonini R. Striatal Astrocytes Shape Behavioral Flexibility via Regulation of the Glutamate Transporter EAAT2. Biol Psychiatry 2021; 89:1045-1057. [PMID: 33516457 DOI: 10.1016/j.biopsych.2020.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 10/21/2020] [Accepted: 11/13/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Striatal circuits must be modulated for behavioral flexibility, the ability to adapt to environmental changes. Striatal astrocytes contribute to circuit neuromodulation by controlling the activity of ambient neurotransmitters. In particular, extracellular glutamate levels are tightly controlled by the astrocytic glutamate transporter EAAT2, influencing synaptic functioning and neural network activity. However, it remains unclear if EAAT2 responds to environmental cues to specifically shape action control. METHODS To investigate the relationship between behavioral flexibility and experience-dependent regulation of EAAT2 expression in the dorsal striatum, mice were trained on an instrumental task. We manipulated EAAT2 expression using chemogenetic activation of astrocytic Gq signaling or in vivo morpholinos and determined the ability to adapt to novel environmental contingencies. RESULTS The loss of behavioral flexibility with task overtraining is associated with the upregulation of EAAT2, which results in enhanced glutamate clearance and altered modulation of glutamatergic neurotransmission in the lateral part of the dorsal striatum. Interfering with EAAT2 upregulation in this striatal area preserves behavioral flexibility. CONCLUSIONS Astrocytes are emerging as critical regulators of striatal functions. This work demonstrates that plasticity of EAAT2 expression in the lateral part of the dorsal striatum shapes behavior, thus providing novel mechanistic insights into how flexibility in action control is regulated.
Collapse
Affiliation(s)
- Arjen J Boender
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Leonardo Bontempi
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Luca Nava
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Yann Pelloux
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy.
| |
Collapse
|
25
|
Buren C, Tu G, Raymond LA. Impaired Replenishment of Cortico-Striatal Synaptic Glutamate in Huntington's Disease Mouse Model. J Huntingtons Dis 2021; 9:149-161. [PMID: 32310183 DOI: 10.3233/jhd-200400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Huntington's disease (HD) is an inherited neurodegenerative disorder caused by expansion of CAG repeats in the Huntingtin gene (HTT). Studies suggest cortical to striatal (C-S) projections, which regulate movement and provide cell survival signals to SPNs, are altered in the pre-manifest and early symptomatic stages of HD. But whether and how presynaptic cortical terminals are affected in HD is not well explored. OBJECTIVE Test size and replenishment of readily releasable pool (RRP), and assess glutamate refill of C-S synapses in HD models. METHODS Immunocytochemistry was applied in C-S co-cultures generated from FVB/N (WT: wildtype) mice and YAC128, an HD mouse model expressing human HTT with 128 CAG repeats on the FVB/N background; Whole-cell patch clamp recordings from striatal neurons were performed both in cultures, with or without osmotic stimuli, and in acute brain slices from 6-month-old early symptomatic YAC128 mice and WT following prolonged trains of electrical stimuli in corpus callosum. RESULTS We found no change in the average size or vesicle replenishment rate of RRP in C-S synapses of YAC128, compared with WT, cultures at day in vitro 21, a time when immunocytochemistry showed comparable neuronal survival between the two genotypes. However, YAC128 C-S synapses showed a slowed rate of recovery of glutamate release in co-cultures as well as in acute brain slices. CONCLUSION Mutant HTT expression impairs glutamate refill but not RRP size or replenishment in C-S synapses. This work provides a foundation for examining the contribution of deficits in presynaptic cortical terminals on HD progression.
Collapse
Affiliation(s)
- Caodu Buren
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada.,Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
| | - Gaqi Tu
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Lynn A Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
26
|
Wilkie CM, Barron JC, Brymer KJ, Barnes JR, Nafar F, Parsons MP. The Effect of GLT-1 Upregulation on Extracellular Glutamate Dynamics. Front Cell Neurosci 2021; 15:661412. [PMID: 33841104 PMCID: PMC8032948 DOI: 10.3389/fncel.2021.661412] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022] Open
Abstract
Pharmacological upregulation of glutamate transporter-1 (GLT-1), commonly achieved using the beta-lactam antibiotic ceftriaxone, represents a promising therapeutic strategy to accelerate glutamate uptake and prevent excitotoxic damage in neurological conditions. While excitotoxicity is indeed implicated in numerous brain diseases, it is typically restricted to select vulnerable brain regions, particularly in early disease stages. In healthy brain tissue, the speed of glutamate uptake is not constant and rather varies in both an activity- and region-dependent manner. Despite the widespread use of ceftriaxone in disease models, very little is known about how such treatments impact functional measures of glutamate uptake in healthy tissue, and whether GLT-1 upregulation can mask the naturally occurring activity-dependent and regional heterogeneities in uptake. Here, we used two different compounds, ceftriaxone and LDN/OSU-0212320 (LDN), to upregulate GLT-1 in healthy wild-type mice. We then used real-time imaging of the glutamate biosensor iGluSnFR to investigate functional consequences of GLT-1 upregulation on activity- and regional-dependent variations in glutamate uptake capacity. We found that while both ceftriaxone and LDN increased GLT-1 expression in multiple brain regions, they did not prevent activity-dependent slowing of glutamate clearance nor did they speed basal clearance rates, even in areas characterized by slow uptake (e.g., striatum). Unexpectedly, ceftriaxone but not LDN decreased glutamate release in the cortex, suggesting that ceftriaxone may alter release properties independent of its effects on GLT-1 expression. In sum, our data demonstrate the complexities of glutamate uptake by showing that GLT-1 expression does not necessarily translate to accelerated uptake. Furthermore, these data suggest that the mechanisms underlying activity- and regional-dependent differences in glutamate dynamics are independent of GLT-1 expression levels.
Collapse
Affiliation(s)
- Crystal M Wilkie
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Jessica C Barron
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Kyle J Brymer
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Jocelyn R Barnes
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Firoozeh Nafar
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| |
Collapse
|
27
|
Brymer KJ, Barnes JR, Parsons MP. Entering a new era of quantifying glutamate clearance in health and disease. J Neurosci Res 2021; 99:1598-1617. [PMID: 33618436 DOI: 10.1002/jnr.24810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/21/2022]
Abstract
Glutamate transporter proteins, expressed on both neurons and glia, serve as the main gatekeepers that dictate the spatial and temporal actions of extracellular glutamate. Glutamate is essential to the function of the healthy brain yet paradoxically contributes to the toxicity associated with many neurodegenerative diseases. Rapid transporter-mediated glutamate uptake, primarily occurring at astrocytic processes, tightens the efficiency of excitatory network activity and prevents toxic glutamate build-up in the extracellular space. Glutamate transporter dysfunction is thought to underlie myriad central nervous system (CNS) diseases including Alzheimer and Huntington disease. Over the past few decades, techniques such as biochemical uptake assays and electrophysiological recordings of transporter currents from individual astrocytes have revealed the remarkable ability of the CNS to efficiently clear extracellular glutamate. In more recent years, the rapidly evolving glutamate-sensing "sniffers" now allow researchers to visualize real-time glutamate transients on a millisecond time scale with single synapse spatial resolution in defined cell populations. As we transition to an increased reliance on optical-based methods of glutamate visualization and quantification, it is of utmost importance to understand not only the advantages that glutamate biosensors bring to the table but also the associated caveats and their implications for data interpretation. In this review, we summarize the strengths and limitations of the commonly used methods to quantify glutamate uptake. We then discuss what these techniques, when viewed as a complementary whole, have told us about the brain's ability to regulate glutamate levels, in both health and in the context of neurodegenerative disease.
Collapse
Affiliation(s)
- Kyle J Brymer
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jocelyn R Barnes
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Matthew P Parsons
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
28
|
Acioglu C, Li L, Elkabes S. Contribution of astrocytes to neuropathology of neurodegenerative diseases. Brain Res 2021; 1758:147291. [PMID: 33516810 DOI: 10.1016/j.brainres.2021.147291] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Classically, the loss of vulnerable neuronal populations in neurodegenerative diseases was considered to be the consequence of cell autonomous degeneration of neurons. However, progress in the understanding of glial function, the availability of improved animal models recapitulating the features of the human diseases, and the development of new approaches to derive glia and neurons from induced pluripotent stem cells obtained from patients, provided novel information that altered this view. Current evidence strongly supports the notion that non-cell autonomous mechanisms contribute to the demise of neurons in neurodegenerative disorders, and glia causally participate in the pathogenesis and progression of these diseases. In addition to microglia, astrocytes have emerged as key players in neurodegenerative diseases and will be the focus of the present review. Under the influence of pathological stimuli present in the microenvironment of the diseased CNS, astrocytes undergo morphological, transcriptional, and functional changes and become reactive. Reactive astrocytes are heterogeneous and exhibit neurotoxic (A1) or neuroprotective (A2) phenotypes. In recent years, single-cell or single-nucleus transcriptome analyses unraveled new, disease-specific phenotypes beyond A1/A2. These investigations highlighted the complexity of the astrocytic responses to CNS pathology. The present review will discuss the contribution of astrocytes to neurodegenerative diseases with particular emphasis on Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Some of the commonalties and differences in astrocyte-mediated mechanisms that possibly drive the pathogenesis or progression of the diseases will be summarized. The emerging view is that astrocytes are potential new targets for therapeutic interventions. A comprehensive understanding of astrocyte heterogeneity and disease-specific phenotypic complexity could facilitate the design of novel strategies to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Cigdem Acioglu
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Lun Li
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| |
Collapse
|
29
|
Morales-Martínez A, Zamorano-Carrillo A, Montes S, El-Hafidi M, Sánchez-Mendoza A, Soria-Castro E, Martínez-Lazcano JC, Martínez-Gopar PE, Ríos C, Pérez-Severiano F. Rich fatty acids diet of fish and olive oils modifies membrane properties in striatal rat synaptosomes. Nutr Neurosci 2021; 24:1-12. [PMID: 30822260 DOI: 10.1080/1028415x.2019.1584692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Background: Essential fatty acids (EFAs) and non-essential fatty acids (nEFAs) exert experimental and clinical neuroprotection in neurodegenerative diseases. The main EFAs, eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), nEFAs, and oleic acid (OA) contained in olive and fish oils are inserted into the cell membranes, but the exact mechanism through which they exert neuroprotection is still unknown. Objectives and Methods: In this study, we assessed the fatty acids content and membrane fluidity in striatal rat synaptosomes after fatty acid-rich diets (olive- or a fish-oil diet, 15% w/w). Then, we evaluated the effect of enriching striatum synaptosomes with fatty acids on the oxidative damage produced by the prooxidants ferrous sulfate (FeSO4) or quinolinic acid (QUIN). Results and Discussion: Lipid profile analysis in striatal synaptosomes showed that EPA content increased in the fish oil group in comparison with control and olive groups. Furthermore, we found that synaptosomes enriched with fatty acids and incubated with QUIN or FeSO4 showed a significant oxidative damage reduction. Results suggest that EFAs, particularly EPA, improve membrane fluidity and confer antioxidant effect.
Collapse
Affiliation(s)
- Adriana Morales-Martínez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
- Laboratorio de Investigación de Bioquímica y Biofísica Computacional, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Absalom Zamorano-Carrillo
- Laboratorio de Investigación de Bioquímica y Biofísica Computacional, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Sergio Montes
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| | - Mohammed El-Hafidi
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Alicia Sánchez-Mendoza
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Elizabeth Soria-Castro
- Departamento de Patología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | | | | | - Camilo Ríos
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México, México
| |
Collapse
|
30
|
Cepeda C, Levine MS. Synaptic Dysfunction in Huntington's Disease: Lessons from Genetic Animal Models. Neuroscientist 2020; 28:20-40. [PMID: 33198566 DOI: 10.1177/1073858420972662] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The understanding of the functional and structural changes occurring in the cerebral cortex and basal ganglia in Huntington's disease (HD) has benefited considerably from the generation of genetic animal models. Most studies of synaptic alterations in HD models have focused on the striatum, but a more complete picture of synaptic dysfunction in the cortico-basal ganglia-cortical loop is emerging. Here, we provide a review and analysis of current developments in the study of synaptic alterations in these areas using HD rodent models. Recent evidence indicates that cortical maldevelopment plays a role in synaptic dysfunction along the corticostriatal pathway that may have its roots in the way mutant huntingtin interacts with synaptic proteins. Furthermore, a progressive disconnection in the corticostriatal pathway leads to abnormal function engaging extrasynaptic N-methyl-D-aspartate glutamate receptors that contribute to eventual cell degeneration. In addition, biphasic increases followed by decreases in glutamate and dopamine release in the striatum could explain contrasting symptomatology in early and late stages of the disease. Changes in striatal output regions also are beginning to be examined. Finally, we highlight some therapeutic avenues aimed at rescuing synaptic dysfunction.
Collapse
Affiliation(s)
- Carlos Cepeda
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Michael S Levine
- IDDRC, Jane and Terry Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
31
|
Eles JR, Stieger KC, Kozai TDY. The temporal pattern of Intracortical Microstimulation pulses elicits distinct temporal and spatial recruitment of cortical neuropil and neurons. J Neural Eng 2020; 18. [PMID: 33075762 DOI: 10.1088/1741-2552/abc29c] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The spacing or distribution of stimulation pulses of therapeutic neurostimulation waveforms-referred to here as the Temporal Pattern (TP)-has emerged as an important parameter for tuning the response to deep-brain stimulation and intracortical microstimulation (ICMS). While it has long been assumed that modulating the TP of ICMS may be effective by altering the rate coding of the neural response, it is unclear how it alters the neural response at the neural network level. The present study is designed to elucidate the neural response to TP at the network level. APPROACH We use in vivo two-photon imaging of ICMS in mice expressing the calcium sensor Thy1-GCaMP or the glutamate sensor hSyn-iGluSnFr to examine the layer II/III neural response to stimulations with different TPs. We study the neuronal calcium and glutamate response to TPs with the same average frequency (10Hz) and same total charge injection, but varying degrees of bursting. We also investigate one control pattern with an average frequency of 100Hz and 10X the charge injection. MAIN RESULTS Stimulation trains with the same average frequency (10 Hz) and same total charge injection but distinct temporal patterns recruits distinct sets of neurons. More-than-half (60% of 309 cells) prefer one temporal pattern over the other. Despite their distinct spatial recruitment patterns, both cells exhibit similar ability to follow 30s trains of both TPs without failing, and they exhibit similar levels of glutamate release during stimulation. Both neuronal calcium and glutamate release train to the bursting TP pattern (~21-fold increase in relative power at the frequency of bursting. Bursting also results in a statistically significant elevation in the correlation between somatic calcium activity and neuropil activity, which we explore as a metric for inhibitory-excitatory tone. Interestingly, soma-neuropil correlation during the bursting pattern is a statistically significant predictor of cell preference for TP, which exposes a key link between inhibitory-excitatory tone. Finally, using mesoscale imaging, we show that both TPs result in distal inhibition during stimulation, which reveals complex spatial and temporal interactions between temporal pattern and inhibitory-excitatory tone in ICMS. SIGNIFICANCE Our results may ultimately suggest that TP is a valuable parameter space to modulate inhibitory-excitatory tone as well as distinct network activity in ICMS. This presents a broader mechanism of action than rate coding, as previously thought. By implicating these additional mechanisms, TP may have broader utility in the clinic and should be pursued to expand the efficacy of ICMS therapies.
Collapse
Affiliation(s)
- James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Kevin C Stieger
- Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Takashi D Yoshida Kozai
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Ave, 5059-BST3, Pittsburgh, PA 15213, USA, Pittsburgh, Pennsylvania, UNITED STATES
| |
Collapse
|
32
|
Romanos J, Benke D, Pietrobon D, Zeilhofer HU, Santello M. Astrocyte dysfunction increases cortical dendritic excitability and promotes cranial pain in familial migraine. SCIENCE ADVANCES 2020; 6:eaaz1584. [PMID: 32548257 PMCID: PMC7274778 DOI: 10.1126/sciadv.aaz1584] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 03/25/2020] [Indexed: 05/02/2023]
Abstract
Astrocytes are essential contributors to neuronal function. As a consequence, disturbed astrocyte-neuron interactions are involved in the pathophysiology of several neurological disorders, with a strong impact on brain circuits and behavior. Here, we describe altered cortical physiology in a genetic mouse model of familial hemiplegic migraine type 2 (FHM2), with reduced expression of astrocytic Na+,K+-ATPases. We used whole-cell electrophysiology, two-photon microscopy, and astrocyte gene rescue to demonstrate that an impairment in astrocytic glutamate uptake promotes NMDA spike generation in dendrites of cingulate cortex pyramidal neurons and enhances output firing of these neurons. Astrocyte compensation of the defective ATPase in the cingulate cortex rescued glutamate uptake, prevented abnormal NMDA spikes, and reduced sensitivity to cranial pain triggers. Together, our results demonstrate that impaired astrocyte function alters neuronal activity in the cingulate cortex and facilitates migraine-like cranial pain states in a mouse model of migraine.
Collapse
Affiliation(s)
- Jennifer Romanos
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
- CNR Institute of Neuroscience, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Mirko Santello
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
- Corresponding author.
| |
Collapse
|
33
|
Hippocampal Synaptic Dysfunction in a Mouse Model of Huntington Disease Is Not Alleviated by Ceftriaxone Treatment. eNeuro 2020; 7:ENEURO.0440-19.2020. [PMID: 32354757 PMCID: PMC7242817 DOI: 10.1523/eneuro.0440-19.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022] Open
Abstract
Glutamate transporters, particularly glutamate transporter 1 (GLT-1), help to prevent the adverse effects associated with glutamate toxicity by rapidly clearing glutamate from the extracellular space. Since GLT-1 expression and/or function are reduced in many neurodegenerative diseases, upregulation of GLT-1 is a favorable approach to treat the symptoms of these diseases. Ceftriaxone, a β-lactam antibiotic reported to increase GLT-1 expression, can exert neuroprotective effects in a variety of neurodegenerative diseases; however, many of these diseases do not exhibit uniform brain pathology. In contrast, as a drug that readily crosses the blood–brain barrier, ceftriaxone administration is likely to increase GLT-1 levels globally throughout the neuroaxis. In Huntington disease (HD), low GLT-1 expression is observed in the striatum in postmortem tissue and animal models. While ceftriaxone was reported to increase striatal GLT-1 and ameliorate the motor symptoms in a mouse model of HD, the extrastriatal effects of ceftriaxone in HD are unknown. Using electrophysiology and high-speed imaging of the glutamate biosensor iGluSnFR, we quantified real-time glutamate dynamics and synaptic plasticity in the hippocampus of the Q175FDN mouse model of HD, following intraperitoneal injections of either saline or ceftriaxone. We observed an activity-dependent increase in extracellular glutamate accumulation within the HD hippocampus, which was not the result of reduced GLT-1 expression. Surprisingly, ceftriaxone had little effect on glutamate clearance rates and negatively impacted synaptic plasticity. These data provide evidence for glutamate dysregulation in the HD hippocampus but also caution the use of ceftriaxone as a treatment for HD.
Collapse
|
34
|
Armbruster M, Dulla CG, Diamond JS. Effects of fluorescent glutamate indicators on neurotransmitter diffusion and uptake. eLife 2020; 9:54441. [PMID: 32352378 PMCID: PMC7255799 DOI: 10.7554/elife.54441] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 04/29/2020] [Indexed: 01/10/2023] Open
Abstract
Genetically encoded fluorescent glutamate indicators (iGluSnFRs) enable neurotransmitter release and diffusion to be visualized in intact tissue. Synaptic iGluSnFR signal time courses vary widely depending on experimental conditions, often lasting 10–100 times longer than the extracellular lifetime of synaptically released glutamate estimated with uptake measurements. iGluSnFR signals typically also decay much more slowly than the unbinding kinetics of the indicator. To resolve these discrepancies, here we have modeled synaptic glutamate diffusion, uptake and iGluSnFR activation to identify factors influencing iGluSnFR signal waveforms. Simulations suggested that iGluSnFR competes with transporters to bind synaptically released glutamate, delaying glutamate uptake. Accordingly, synaptic transporter currents recorded from iGluSnFR-expressing astrocytes in mouse cortex were slower than those in control astrocytes. Simulations also suggested that iGluSnFR reduces free glutamate levels in extrasynaptic spaces, likely limiting extrasynaptic receptor activation. iGluSnFR and lower affinity variants, nonetheless, provide linear indications of vesicle release, underscoring their value for optical quantal analysis.
Collapse
Affiliation(s)
- Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, United States
| | - Jeffrey S Diamond
- Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, United States
| |
Collapse
|
35
|
The Relationship Between Glutamate Dynamics and Activity-Dependent Synaptic Plasticity. J Neurosci 2020; 40:2793-2807. [PMID: 32102922 DOI: 10.1523/jneurosci.1655-19.2020] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
The spatiotemporal dynamics of excitatory neurotransmission must be tightly regulated to achieve efficient synaptic communication. By limiting spillover, glutamate transporters are believed to prevent excessive activation of extrasynaptically located receptors that can impair synaptic plasticity. While glutamate transporter expression is reduced in numerous neurodegenerative diseases, the contributions of transporter dysfunction to disease pathophysiology remain ambiguous as the fundamental relationship between glutamate dynamics and plasticity, and the mechanisms linking these two phenomena, remain poorly understood. Here, we combined electrophysiology and real-time high-speed imaging of extracellular glutamate transients during LTP induction and characterized the sensitivity of the relationship between glutamate dynamics during theta burst stimulation (TBS) and the resulting magnitude of LTP consolidation, both in control conditions and following selective and nonselective glutamate transporter blockade. Glutamate clearance times were negatively correlated with LTP magnitude following nonselective glutamate transporter inhibition but not following selective blockade of a majority of GLT-1, the brain's most abundant glutamate transporter. Although glutamate transporter inhibition reduced the postsynaptic population response to TBS, calcium responses to TBS were greatly exaggerated. The source of excess calcium was dependent on NMDARs, L-type VGCCs, GluA2-lacking AMPARs, and internal calcium stores. Surprisingly, inhibition of L-type VGCCs, but not GluA2-lacking AMPARs or ryanodine receptors, was required to restore robust LTP. In all, these data provide a detailed understanding of the relationship between glutamate dynamics and plasticity and uncover important mechanisms by which poor glutamate uptake can negatively impact LTP consolidation.SIGNIFICANCE STATEMENT Specific patterns of neural activity can promote long-term changes in the strength of synaptic connections through a phenomenon known as synaptic plasticity. Synaptic plasticity is well accepted to represent the cellular mechanisms underlying learning and memory, and many forms of plasticity are initiated by the excitatory neurotransmitter glutamate. While essential for rapid cellular communication in the brain, excessive levels of extracellular glutamate can negatively impact brain function. In this study, we demonstrate that pharmacological manipulations that increase the availability of extracellular glutamate during neural activity can have profoundly negative consequences on synaptic plasticity. We identify mechanisms through which excess glutamate can negatively influence synaptic plasticity, and we discuss the relevance of these findings to neurodegenerative diseases and in the aging brain.
Collapse
|
36
|
Blumenstock S, Dudanova I. Cortical and Striatal Circuits in Huntington's Disease. Front Neurosci 2020; 14:82. [PMID: 32116525 PMCID: PMC7025546 DOI: 10.3389/fnins.2020.00082] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 12/28/2022] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder that typically manifests in midlife with motor, cognitive, and/or psychiatric symptoms. The disease is caused by a CAG triplet expansion in exon 1 of the huntingtin gene and leads to a severe neurodegeneration in the striatum and cortex. Classical electrophysiological studies in genetic HD mouse models provided important insights into the disbalance of excitatory, inhibitory and neuromodulatory inputs, as well as progressive disconnection between the cortex and striatum. However, the involvement of local cortical and striatal microcircuits still remains largely unexplored. Here we review the progress in understanding HD-related impairments in the cortical and basal ganglia circuits, and outline new opportunities that have opened with the development of modern circuit analysis methods. In particular, in vivo imaging studies in mouse HD models have demonstrated early structural and functional disturbances within the cortical network, and optogenetic manipulations of striatal cell types have started uncovering the causal roles of certain neuronal populations in disease pathogenesis. In addition, the important contribution of astrocytes to HD-related circuit defects has recently been recognized. In parallel, unbiased systems biology studies are providing insights into the possible molecular underpinnings of these functional defects at the level of synaptic signaling and neurotransmitter metabolism. With these approaches, we can now reach a deeper understanding of circuit-based HD mechanisms, which will be crucial for the development of effective and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sonja Blumenstock
- Department of Molecules – Signaling – Development, Max Planck Institute of Neurobiology, Martinsried, Germany
- Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Irina Dudanova
- Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
37
|
Eles JR, Kozai TDY. In vivo imaging of calcium and glutamate responses to intracortical microstimulation reveals distinct temporal responses of the neuropil and somatic compartments in layer II/III neurons. Biomaterials 2020; 234:119767. [PMID: 31954232 DOI: 10.1016/j.biomaterials.2020.119767] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/22/2019] [Accepted: 01/05/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Intracortical microelectrode implants can generate a tissue response hallmarked by glial scarring and neuron cell death within 100-150 μm of the biomaterial device. Many have proposed that any performance decline in intracortical microstimulation (ICMS) due to this foreign body tissue response could be offset by increasing the stimulation amplitude. The mechanisms of this approach are unclear, however, as there has not been consensus on how increasing amplitude affects the spatial and temporal recruitment patterns of ICMS. APPROACH We clarify these unknowns using in vivo two-photon imaging of mice transgenically expressing the calcium sensor GCaMP6s in Thy1 neurons or virally expressing the glutamate sensor iGluSnFr in neurons. Calcium and neurotransmitter activity are tracked in the neuronal somas and neuropil during long-train stimulation in Layer II/III of somatosensory cortex. MAIN RESULTS Neural calcium activity and glutamate release are dense and strongest within 20-40 μm around the electrode, falling off with distance from the electrode. Neuronal calcium increases with higher amplitude stimulations. During prolonged stimulation trains, a sub-population of somas fail to maintain calcium activity. Interestingly, neuropil calcium activity is 3-fold less correlated to somatic calcium activity for cells that drop-out during the long stimulation train compared to cells that sustain activity throughout the train. Glutamate release is apparent only within 20 μm of the electrode and is sustained for at least 10s after cessation of the 15 and 20 μA stimulation train, but not lower amplitudes. SIGNIFICANCE These results demonstrate that increasing amplitude can increase the radius and intensity of neural recruitment, but it also alters the temporal response of some neurons. Further, dense glutamate release is highest within the first 20 μm of the electrode site even at high amplitudes, suggesting that there may be spatial limitations to the amplitude parameter space. The glutamate elevation outlasts stimulation, suggesting that high-amplitude stimulation may affect neurotransmitter re-uptake. This ultimately suggests that increasing the amplitude of ICMS device stimulation may fundamentally alter the temporal neural response, which could have implications for using amplitude to improve the ICMS effect or "offset" the effects of glial scarring.
Collapse
Affiliation(s)
- James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Carnegie Mellon University, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
38
|
Skotte NH, Andersen JV, Santos A, Aldana BI, Willert CW, Nørremølle A, Waagepetersen HS, Nielsen ML. Integrative Characterization of the R6/2 Mouse Model of Huntington's Disease Reveals Dysfunctional Astrocyte Metabolism. Cell Rep 2019; 23:2211-2224. [PMID: 29768217 DOI: 10.1016/j.celrep.2018.04.052] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/23/2018] [Accepted: 04/12/2018] [Indexed: 01/05/2023] Open
Abstract
Huntington's disease is a fatal neurodegenerative disease, where dysfunction and loss of striatal and cortical neurons are central to the pathogenesis of the disease. Here, we integrated quantitative studies to investigate the underlying mechanisms behind HD pathology in a systems-wide manner. To this end, we used state-of-the-art mass spectrometry to establish a spatial brain proteome from late-stage R6/2 mice and compared this with wild-type littermates. We observed altered expression of proteins in pathways related to energy metabolism, synapse function, and neurotransmitter homeostasis. To support these findings, metabolic 13C labeling studies confirmed a compromised astrocytic metabolism and regulation of glutamate-GABA-glutamine cycling, resulting in impaired release of glutamine and GABA synthesis. In recent years, increasing attention has been focused on the role of astrocytes in HD, and our data support that therapeutic strategies to improve astrocytic glutamine homeostasis may help ameliorate symptoms in HD.
Collapse
Affiliation(s)
- Niels H Skotte
- Proteomics Program, The Novo Nordisk Foundation Centre for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alberto Santos
- Proteomics Program, The Novo Nordisk Foundation Centre for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie W Willert
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nørremølle
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael L Nielsen
- Proteomics Program, The Novo Nordisk Foundation Centre for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
39
|
Creus-Muncunill J, Ehrlich ME. Cell-Autonomous and Non-cell-Autonomous Pathogenic Mechanisms in Huntington's Disease: Insights from In Vitro and In Vivo Models. Neurotherapeutics 2019; 16:957-978. [PMID: 31529216 PMCID: PMC6985401 DOI: 10.1007/s13311-019-00782-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Huntington's disease (HD) is an autosomal dominant disorder caused by an expansion in the trinucleotide CAG repeat in exon-1 in the huntingtin gene, located on chromosome 4. When the number of trinucleotide CAG exceeds 40 repeats, disease invariably is manifested, characterized by motor, cognitive, and psychiatric symptoms. The huntingtin (Htt) protein and its mutant form (mutant huntingtin, mHtt) are ubiquitously expressed but although multiple brain regions are affected, the most vulnerable brain region is the striatum. Striatal medium-sized spiny neurons (MSNs) preferentially degenerate, followed by the cortical pyramidal neurons located in layers V and VI. Proposed HD pathogenic mechanisms include, but are not restricted to, excitotoxicity, neurotrophic support deficits, collapse of the protein degradation mechanisms, mitochondrial dysfunction, transcriptional alterations, and disorders of myelin. Studies performed in cell type-specific and regionally selective HD mouse models implicate both MSN cell-autonomous properties and cell-cell interactions, particularly corticostriatal but also with non-neuronal cell types. Here, we review the intrinsic properties of MSNs that contribute to their selective vulnerability and in addition, we discuss how astrocytes, microglia, and oligodendrocytes, together with aberrant corticostriatal connectivity, contribute to HD pathophysiology. In addition, mHtt causes cell-autonomous dysfunction in cell types other than MSNs. These findings have implications in terms of therapeutic strategies aimed at preventing neuronal dysfunction and degeneration.
Collapse
Affiliation(s)
- Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
40
|
Wang Y, DelRosso NV, Vaidyanathan TV, Cahill MK, Reitman ME, Pittolo S, Mi X, Yu G, Poskanzer KE. Accurate quantification of astrocyte and neurotransmitter fluorescence dynamics for single-cell and population-level physiology. Nat Neurosci 2019; 22:1936-1944. [PMID: 31570865 PMCID: PMC6858541 DOI: 10.1038/s41593-019-0492-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023]
Abstract
Recent work examining astrocytic physiology centers on fluorescence imaging, due to development of sensitive fluorescent indicators and observation of spatiotemporally complex calcium activity. However, the field remains hindered in characterizing these dynamics, both within single cells and at the population level, because of the insufficiency of current region-of-interest (ROI)-based approaches to describe activity that is often spatially unfixed, size-varying, and propagative. Here, we present an analytical framework that releases astrocyte biologists from ROI-based tools. The Astrocyte Quantitative Analysis (AQuA) software takes an event-based perspective to model and accurately quantify complex calcium and neurotransmitter activity in fluorescence imaging datasets. We apply AQuA to a range of ex vivo and in vivo imaging data, and uncover novel physiological phenomena. Since AQuA is data-driven and based on machine learning principles, it can be applied across model organisms, fluorescent indicators, experimental modes, and imaging resolutions and speeds, enabling researchers to elucidate fundamental neural physiology.
Collapse
Affiliation(s)
- Yizhi Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA
| | - Nicole V DelRosso
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Trisha V Vaidyanathan
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle K Cahill
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Michael E Reitman
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Silvia Pittolo
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Xuelong Mi
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA
| | - Guoqiang Yu
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA.
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA. .,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA. .,Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA.
| |
Collapse
|
41
|
Siracusa R, Fusco R, Cuzzocrea S. Astrocytes: Role and Functions in Brain Pathologies. Front Pharmacol 2019; 10:1114. [PMID: 31611796 PMCID: PMC6777416 DOI: 10.3389/fphar.2019.01114] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/30/2019] [Indexed: 12/16/2022] Open
Abstract
Astrocytes are a population of cells with distinctive morphological and functional characteristics that differ within specific areas of the brain. Postnatally, astrocyte progenitors migrate to reach their brain area and related properties. They have a regulatory role of brain functions that are implicated in neurogenesis and synaptogenesis, controlling blood-brain barrier permeability and maintaining extracellular homeostasis. Mature astrocytes also express some genes enriched in cell progenitors, suggesting they can retain proliferative potential. Considering heterogeneity of cell population, it is not surprising that their disorders are related to a wide range of different neuro-pathologies. Brain diseases are characterized by the active inflammatory state of the astrocytes, which is usually described as up-regulation of glial fibrillary acidic protein (GFAP). In particular, the loss of astrocytes function as a result of cellular senescence could have implications for the neurodegenerative disorders, such as Alzheimer disease and Huntington disease, and for the aging brain. Astrocytes can also drive the induction and the progression of the inflammatory state due to their Ca2+ signals and that it is strongly related to the disease severity/state. Moreover, they contribute to the altered neuronal activity in several frontal cortex pathologies such as ischemic stroke and epilepsy. There, we describe the current knowledge pertaining to astrocytes' role in brain pathologies and discuss the possibilities to target them as approach toward pharmacological therapies for neuro-pathologies.
Collapse
Affiliation(s)
- Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, Italy.,Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
42
|
Soares C, Trotter D, Longtin A, Béïque JC, Naud R. Parsing Out the Variability of Transmission at Central Synapses Using Optical Quantal Analysis. Front Synaptic Neurosci 2019; 11:22. [PMID: 31474847 PMCID: PMC6702664 DOI: 10.3389/fnsyn.2019.00022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
Properties of synaptic release dictates the core of information transfer in neural circuits. Despite decades of technical and theoretical advances, distinguishing bona fide information content from the multiple sources of synaptic variability remains a challenging problem. Here, we employed a combination of computational approaches with cellular electrophysiology, two-photon uncaging of MNI-Glutamate and imaging at single synapses. We describe and calibrate the use of the fluorescent glutamate sensor iGluSnFR and found that its kinetic profile is close to that of AMPA receptors, therefore providing several distinct advantages over slower methods relying on NMDA receptor activation (i.e., chemical or genetically encoded calcium indicators). Using an array of statistical methods, we further developed, and validated on surrogate data, an expectation-maximization algorithm that, by biophysically constraining release variability, extracts the quantal parameters n (maximum number of released vesicles) and p (unitary probability of release) from single-synapse iGluSnFR-mediated transients. Together, we present a generalizable mathematical formalism which, when applied to optical recordings, paves the way to an increasingly precise investigation of information transfer at central synapses.
Collapse
Affiliation(s)
- Cary Soares
- Department of Cellular and Molecular Medicine, uOttawa Brain and Mind Research Institute, Center for Neural Dynamics, University of Ottawa, Ottawa, ON, Canada
| | - Daniel Trotter
- Department of Physics, University of Ottawa, Ottawa, ON, Canada
| | - André Longtin
- Department of Cellular and Molecular Medicine, uOttawa Brain and Mind Research Institute, Center for Neural Dynamics, University of Ottawa, Ottawa, ON, Canada
- Department of Physics, University of Ottawa, Ottawa, ON, Canada
| | - Jean-Claude Béïque
- Department of Cellular and Molecular Medicine, uOttawa Brain and Mind Research Institute, Center for Neural Dynamics, University of Ottawa, Ottawa, ON, Canada
| | - Richard Naud
- Department of Cellular and Molecular Medicine, uOttawa Brain and Mind Research Institute, Center for Neural Dynamics, University of Ottawa, Ottawa, ON, Canada
- Department of Physics, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
43
|
Koch ET, Raymond LA. Dysfunctional striatal dopamine signaling in Huntington's disease. J Neurosci Res 2019; 97:1636-1654. [PMID: 31304622 DOI: 10.1002/jnr.24495] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/06/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022]
Abstract
Dopamine signaling in the striatum is critical for a variety of behaviors including movement, behavioral flexibility, response to reward and many forms of learning. Alterations to dopamine transmission contribute to pathological features of many neurological diseases, including Huntington's disease (HD). HD is an autosomal dominant genetic disorder caused by a CAG repeat expansion in the Huntingtin gene. The striatum is preferentially degenerated in HD, and this region receives dopaminergic input from the substantia nigra. Studies of HD patients and genetic rodent models have shown changes to levels of dopamine and its receptors in the striatum, and alterations in dopamine receptor signaling and modulation of other neurotransmitters, notably glutamate. Throughout his career, Dr. Michael Levine's research has furthered our understanding of dopamine signaling in the striatum of healthy rodents and HD mouse models. This review will focus on the work of his group and others in elucidating alterations to striatal dopamine signaling that contribute to pathophysiology in HD mouse models, and how these findings relate to human HD studies. We will also discuss current and potential therapeutic interventions for HD that target the dopamine system, and future research directions for this field.
Collapse
Affiliation(s)
- Ellen T Koch
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
44
|
Ciriachi C, Svane‐Petersen D, Rickhag M. Genetic tools to study complexity of striatal function. J Neurosci Res 2019; 97:1181-1193. [DOI: 10.1002/jnr.24479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 01/11/2023]
Affiliation(s)
- Chiara Ciriachi
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - David Svane‐Petersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Mattias Rickhag
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
45
|
Smith‐Dijak AI, Sepers MD, Raymond LA. Alterations in synaptic function and plasticity in Huntington disease. J Neurochem 2019; 150:346-365. [DOI: 10.1111/jnc.14723] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/28/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Amy I. Smith‐Dijak
- Graduate Program in Neuroscience the University of British Columbia Vancouver British Columbia Canada
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| | - Marja D. Sepers
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| | - Lynn A. Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
46
|
Valtcheva S, Venance L. Control of Long-Term Plasticity by Glutamate Transporters. Front Synaptic Neurosci 2019; 11:10. [PMID: 31024287 PMCID: PMC6465798 DOI: 10.3389/fnsyn.2019.00010] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Activity-dependent long-term changes in synaptic strength constitute key elements for learning and memory formation. Long-term plasticity can be induced in vivo and ex vivo by various physiologically relevant activity patterns. Depending on their temporal statistics, such patterns can induce long-lasting changes in the synaptic weight by potentiating or depressing synaptic transmission. At excitatory synapses, glutamate uptake operated by excitatory amino acid transporters (EAATs) has a critical role in regulating the strength and the extent of receptor activation by afferent activity. EAATs tightly control synaptic transmission and glutamate spillover. EAATs activity can, therefore, determine the polarity and magnitude of long-term plasticity by regulating the spatiotemporal profile of the glutamate transients and thus, the glutamate access to pre- and postsynaptic receptors. Here, we summarize compelling evidence that EAATs regulate various forms of long-term synaptic plasticity and the consequences of such regulation for behavioral output. We speculate that experience-dependent plasticity of EAATs levels can determine the sensitivity of synapses to frequency- or time-dependent plasticity paradigms. We propose that EAATs contribute to the gating of relevant inputs eligible to induce long-term plasticity and thereby select the operating learning rules that match the physiological function of the synapse adapted to the behavioral context.
Collapse
Affiliation(s)
- Silvana Valtcheva
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241/INSERM U1050, Paris, France
| | - Laurent Venance
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241/INSERM U1050, Paris, France
| |
Collapse
|
47
|
Romanos J, Benke D, Saab AS, Zeilhofer HU, Santello M. Differences in glutamate uptake between cortical regions impact neuronal NMDA receptor activation. Commun Biol 2019; 2:127. [PMID: 30963115 PMCID: PMC6451009 DOI: 10.1038/s42003-019-0367-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/05/2019] [Indexed: 01/06/2023] Open
Abstract
Removal of synaptically-released glutamate by astrocytes is necessary to spatially and temporally limit neuronal activation. Recent evidence suggests that astrocytes may have specialized functions in specific circuits, but the extent and significance of such specialization are unclear. By performing direct patch-clamp recordings and two-photon glutamate imaging, we report that in the somatosensory cortex, glutamate uptake by astrocytes is slower during sustained synaptic stimulation when compared to lower stimulation frequencies. Conversely, glutamate uptake capacity is increased in the frontal cortex during higher frequency synaptic stimulation, thereby limiting extracellular buildup of glutamate and NMDA receptor activation in layer 5 pyramidal neurons. This efficient glutamate clearance relies on Na+/K+-ATPase function and both GLT-1 and non-GLT-1 transporters. Thus, by enhancing their glutamate uptake capacity, astrocytes in the frontal cortex may prevent excessive neuronal excitation during intense synaptic activity. These results may explain why diseases associated with network hyperexcitability differentially affect individual brain areas.
Collapse
Affiliation(s)
- Jennifer Romanos
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Aiman S. Saab
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Mirko Santello
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| |
Collapse
|
48
|
Michelson NJ, Vanni MP, Murphy TH. Comparison between transgenic and AAV-PHP.eB-mediated expression of GCaMP6s using in vivo wide-field functional imaging of brain activity. NEUROPHOTONICS 2019; 6:025014. [PMID: 31763351 PMCID: PMC6864505 DOI: 10.1117/1.nph.6.2.025014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/22/2019] [Indexed: 05/13/2023]
Abstract
We employ transcranial wide-field single-photon imaging to compare genetically encoded calcium sensors under transgenic or viral vector expression strategies. Awake, head-fixed animals and brief visual flash stimuli are used to assess function. The use of awake transcranial imaging may reduce confounds attributed to cranial window implantation or anesthesia states. We report differences in wide-field epifluorescence brightness and peak Δ F / F 0 response to visual stimulation between expression strategies. Other metrics for indicator performance include fluctuation analysis (standard deviation) and regional correlation maps made from spontaneous activity. We suggest that multiple measures, such as stimulus-evoked signal-to-noise ratio, brightness, and averaged visual Δ F / F 0 response, may be necessary to characterize indicator sensitivity and methods of expression. Furthermore, we show that strategies using blood brain barrier-permeable viruses, such as PHP.eB, yield comparable expression and function as those derived from transgenic mice. We suggest that testing of new genetically engineered activity sensors could employ a single-photon, wide-field imaging pipeline involving visual stimulation in awake mice that have been intravenously injected with PHP.eB.
Collapse
Affiliation(s)
- Nicholas J. Michelson
- Kinsmen Laboratory of Neurological Research, Department of Psychiatry, Vancouver, British Columbia, Canada
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, British Columbia, Canada
| | - Matthieu P. Vanni
- Kinsmen Laboratory of Neurological Research, Department of Psychiatry, Vancouver, British Columbia, Canada
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, British Columbia, Canada
- Université de Montréal, School of Optometry, Montréal, Québec, Canada
| | - Timothy H. Murphy
- Kinsmen Laboratory of Neurological Research, Department of Psychiatry, Vancouver, British Columbia, Canada
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, British Columbia, Canada
- Address all correspondence to Timothy H. Murphy E-mail:
| |
Collapse
|
49
|
Single Synapse Indicators of Impaired Glutamate Clearance Derived from Fast iGlu u Imaging of Cortical Afferents in the Striatum of Normal and Huntington (Q175) Mice. J Neurosci 2019; 39:3970-3982. [PMID: 30819797 DOI: 10.1523/jneurosci.2865-18.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/05/2019] [Accepted: 02/19/2019] [Indexed: 01/23/2023] Open
Abstract
Changes in the balance between glutamate (Glu) release and uptake may stimulate synaptic reorganization and even synapse loss. In the case of neurodegeneration, a mismatch between astroglial Glu uptake and presynaptic Glu release could be detected if both parameters were assessed independently and at a single-synapse level. This has now become possible due to a new imaging assay with the genetically encoded ultrafast Glu sensor iGlu u We report findings from individual corticostriatal synapses in acute slices prepared from mice of either sex that were >1 year of age. Contrasting patterns of short-term plasticity and a size criterion identified two classes of terminals, presumably corresponding to the previously defined IT (intratelencephalic) and PT (pyramidal tract) synapses. The latter exhibited a higher degree of frequency potentiation/residual Glu accumulation and were selected for our first iGlu u single-synapse study in Q175 mice, a model of Huntington's disease (HD). In HD mice, the decay time constant of the perisynaptic Glu concentration (TauD), as an indicator of uptake, and the peak iGlu u amplitude, as an indicator of release, were prolonged and reduced, respectively. Treatment of WT preparations with the astrocytic Glu uptake blocker TFB-TBOA (100 nm) mimicked the TauD changes in homozygotes. Considering the largest TauD values encountered in WT, ∼40% of PT synapses tested in Q175 heterozygotes can be classified as dysfunctional. Moreover, HD but not WT synapses exhibited a positive correlation between TauD and the peak amplitude of iGlu u Finally, EAAT2 (excitatory amino acid transport protein 2) immunoreactivity was reduced next to corticostriatal terminals. Thus, astrocytic Glu transport remains a promising target for therapeutic intervention.SIGNIFICANCE STATEMENT Alterations in astrocytic Glu uptake can play a role in synaptic plasticity and neurodegeneration. Until now, the sensitivity of synaptic responses to pharmacological transport block and the resulting activation of NMDA receptors were regarded as reliable evidence for a mismatch between synaptic uptake and release. But the latter parameters are interdependent. Using a new genetically encoded sensor to monitor extracellular glutamate concentration ([Glu]) at individual corticostriatal synapses, we can now quantify the time constant of perisynaptic [Glu] decay (as an indicator of uptake) and the maximal [Glu] elevation next to the active zone (as an indicator of Glu release). The results provide a positive answer to the hitherto unresolved question of whether neurodegeneration (e.g., Huntington's disease) associates with a glutamate uptake deficit at tripartite excitatory synapses.
Collapse
|
50
|
Peteri UK, Niukkanen M, Castrén ML. Astrocytes in Neuropathologies Affecting the Frontal Cortex. Front Cell Neurosci 2019; 13:44. [PMID: 30809131 PMCID: PMC6379461 DOI: 10.3389/fncel.2019.00044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/28/2019] [Indexed: 01/15/2023] Open
Abstract
To an increasing extent, astrocytes are connected with various neuropathologies. Astrocytes comprise of a heterogeneous population of cells with region- and species-specific properties. The frontal cortex exhibits high levels of plasticity that is required for high cognitive functions and memory making this region especially susceptible to damage. Aberrations in the frontal cortex are involved with several cognitive disorders, including Alzheimer’s disease, Huntington’s disease and frontotemporal dementia. Human induced pluripotent stem cells (iPSCs) provide an alternative for disease modeling and offer possibilities for studies to investigate pathological mechanisms in a cell type-specific manner. Patient-specific iPSC-derived astrocytes have been shown to recapitulate several disease phenotypes. Addressing astrocyte heterogeneity may provide an improved understanding of the mechanisms underlying neurodegenerative diseases.
Collapse
Affiliation(s)
- Ulla-Kaisa Peteri
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Niukkanen
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maija L Castrén
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|