1
|
Kitchigina VF. Colocalization of Neurotransmitters in Hippocampus and Afferent Systems: Possible Functional Role. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:61-78. [PMID: 40058974 DOI: 10.1134/s0006297924603915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 05/13/2025]
Abstract
In neurophysiology, the transmitter phenotype is considered as an indicator of neuronal identity. It has become known at the end of last century that a nerve cell can produce and use several different molecules to communicate with other neurons. These could be "classical" transmitters: glutamate or gamma-aminobutyric acid (or acetylcholine, serotonin, norepinephrine), as well as secondary messengers, mainly neuropeptides released from the same neurons. In the case, when classical neurotransmitters are released together from the same nerve cell, this event is called cotransmission or corelease (release from the same vesicles). In this review article, the term "cotransmission" is used in a broad sense, denoting neurons that can release more than one classical mediator. Since transmitters are often intermediate products of metabolism and are found in many cells, the neuron classification is currently based on the carrier proteins (transporters) that "pack" neurotransmitters synthesized in the cytoplasm into vesicles. Here, we limit the issue of colocalization of the main neurotransmitters in mammals to the neurons of hippocampus and those structures that send their pathways to it. The review considers problems concerning the mechanisms of multitransmitter signaling, as well as probable functional role of mediator colocalization in the work of hippocampus, which yet has been poorly understood. It has been suggested that co-expression of different mediator phenotypes is involved in maintaining the balance of excitation and inhibition in different regions of hippocampus, facilitates rapid selection of information processing mode, induction of long-term potentiation, maintenance of spatial coding by place cells, as well as ensuring flexibility of learning and formation of working memory. However, the functional role of mediator colocalization, as well as the mechanisms of release of "dual" transmitters, have not been fully elucidated. The solution of these problems will advance some areas of fundamental neuroscience and help in the treatment of those diseases, where disruption of the balance between excitation and inhibition is detected, such as, for example, in epilepsy, Alzheimer's disease, and many others.
Collapse
Affiliation(s)
- Valentina F Kitchigina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
2
|
Hirai H, Konno K, Yamasaki M, Watanabe M, Sakaba T, Hashimotodani Y. Distinct release properties of glutamate/GABA co-transmission serve as a frequency-dependent filtering of supramammillary inputs. eLife 2024; 13:RP99711. [PMID: 39680436 DOI: 10.7554/elife.99711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024] Open
Abstract
Glutamate and GABA co-transmitting neurons exist in several brain regions; however, the mechanism by which these two neurotransmitters are co-released from the same synaptic terminals remains unclear. Here, we show that the supramammillary nucleus (SuM) to dentate granule cell synapses, which co-release glutamate and GABA, exhibit differences between glutamate and GABA release properties in paired-pulse ratio, Ca2+-sensitivity, presynaptic receptor modulation, and Ca2+ channel-vesicle coupling configuration. Moreover, uniquantal synaptic responses show independent glutamatergic and GABAergic responses. Morphological analysis reveals that most SuM terminals form distinct glutamatergic and GABAergic synapses in proximity, each characterized by GluN1 and GABAAα1 labeling, respectively. Notably, glutamate/GABA co-transmission exhibits distinct short-term plasticities, with frequency-dependent depression of glutamate and frequency-independent stable depression of GABA. Our findings suggest that glutamate and GABA are co-released from different synaptic vesicles within the SuM terminals, and reveal that distinct transmission modes of glutamate/GABA co-release serve as frequency-dependent filters of SuM inputs.
Collapse
Affiliation(s)
- Himawari Hirai
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | | |
Collapse
|
3
|
Pedersen C, Chen VT, Herbst P, Zhang R, Elfert A, Krishan A, Azar DT, Chang JH, Hu WY, Kremsmayer TP, Jalilian E, Djalilian AR, Guaiquil VH, Rosenblatt MI. Target specification and therapeutic potential of extracellular vesicles for regulating corneal angiogenesis, lymphangiogenesis, and nerve repair. Ocul Surf 2024; 34:459-476. [PMID: 39426677 PMCID: PMC11921040 DOI: 10.1016/j.jtos.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Extracellular vesicles, including exosomes, are small extracellular vesicles that range in size from 30 nm to 10 μm in diameter and have specific membrane markers. They are naturally secreted and are present in various bodily fluids, including blood, urine, and saliva, and through the variety of their internal cargo, they contribute to both normal physiological and pathological processes. These processes include immune modulation, neuronal synapse formation, cell differentiation, cancer metastasis, angiogenesis, lymphangiogenesis, progression of infectious disease, and neurodegenerative disorders like Alzheimer's and Parkinson's disease. In recent years, interest has grown in the use of exosomes as a potential drug delivery system for various diseases and injuries. Importantly, exosomes originating from a patient's own cells exhibit minimal immunogenicity and possess remarkable stability along with inherent and adjustable targeting capabilities. This review explores the roles of exosomes in angiogenesis, lymphangiogenesis, and nerve repair with a specific emphasis on these processes within the cornea. Furthermore, it examines exosomes derived from specific cell types, discusses the advantages of exosome-based therapies in modulating these processes, and presents some of the most established methods for exosome isolation. Exosome-based treatments are emerging as potential minimally invasive and non-immunogenic therapies that modulate corneal angiogenesis and lymphangiogenesis, as well as enhance and accelerate endogenous corneal nerve repair.
Collapse
Affiliation(s)
- Cameron Pedersen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victoria T Chen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Runze Zhang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Amr Elfert
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Abhi Krishan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tobias P Kremsmayer
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Victor H Guaiquil
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Asmerian H, Alberts J, Sanetra AM, Diaz AJ, Silm K. Role of adaptor protein complexes in generating functionally distinct synaptic vesicle pools. J Physiol 2024. [PMID: 39034608 DOI: 10.1113/jp286179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
The synaptic vesicle (SV) cycle ensures the release of neurotransmitters and the replenishment of SVs to sustain neuronal activity. Multiple endocytosis and sorting pathways contribute to the recapture of the SV membrane and proteins after fusion. Adaptor protein (AP) complexes are among the critical components of the SV retrieval machinery. The canonical clathrin adaptor AP2 ensures the replenishment of most SVs across many neuronal populations. An alternative AP1/AP3-dependent process mediates the formation of a subset of SVs that differ from AP2 vesicles in molecular composition and respond preferentially during higher frequency firing. Furthermore, recent studies show that vesicular transporters for different neurotransmitters depend to a different extent on the AP3 pathway and this affects the release properties of the respective neurotransmitters. This review focuses on the current understanding of the AP-dependent molecular and functional diversity among SVs. We also discuss the contribution of these pathways to the regulation of neurotransmitter release across neuronal populations.
Collapse
Affiliation(s)
- Hrach Asmerian
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jacob Alberts
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anna M Sanetra
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexia J Diaz
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Katlin Silm
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
5
|
Wood OWG, Walby J, Yeung JH, Ke S, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A. Alzheimer's Disease-associated Region-specific Decrease of Vesicular Glutamate Transporter Immunoreactivity inthe Medial Temporal Lobe and Superior Temporal Gyrus. Neuroscience 2024; 546:75-87. [PMID: 38552733 DOI: 10.1016/j.neuroscience.2024.03.027] [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: 09/26/2023] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which there are very limited treatment options. Dysfunction of the excitatory neurotransmitter system is thought to play a major role in the pathogenesis of this condition. Vesicular glutamate transporters (VGLUTs) are key to controlling the quantal release of glutamate. Thus, expressional changes in disease can have implications for aberrant neuronal activity, raising the possibility of a therapeutic target. There is no information regarding the expression of VGLUTs in the human medial temporal lobe in AD, one of the earliest and most severely affected brain regions. This study aimed to quantify and compare the layer-specific expression of VGLUT1 and VGLUT2 between control and AD cases in the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus. Free-floating fluorescent immunohistochemistry was used to label VGLUT1 and VGLUT2 in the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus. Sections were imaged using laser-scanning confocal microscopy and transporter densitometric analysis was performed. VGLUT1 density was not significantly different in AD tissue, except lower staining density observed in the dentate gyrus stratum moleculare (p = 0.0051). VGLUT2 expression was not altered in the hippocampus and entorhinal cortex of AD cases but was significantly lower in the subiculum (p = 0.015) and superior temporal gyrus (p = 0.0023). This study indicates a regionally specific vulnerability of VGLUT1 and VGLUT2 expression in the medial temporal lobe and superior temporal gyrus in AD. However, the causes and functional consequences of these disturbances need to be further explored to assess VGLUT1 and VGLUT2 as viable therapeutic targets.
Collapse
Affiliation(s)
- Oliver W G Wood
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Josh Walby
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Jason H Yeung
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Stephen Ke
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, New Zealand; Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland.
| |
Collapse
|
6
|
Li D, Ai S, Huang C, Liu ZH, Wang HL. Icariin rescues developmental BPA exposure induced spatial memory deficits in rats. Toxicol Appl Pharmacol 2024; 482:116776. [PMID: 38043803 DOI: 10.1016/j.taap.2023.116776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Bisphenol A (BPA) has been implicated in cognitive impairment. Icariin is the main active ingredient extracted from Epimedium Herb with protective function of nervous system. However, the potential therapeutic effects of Icariin on spatial memory deficits induced by developmental BPA exposure in Sprague-Dawley rats have not been investigated. This study investigated the therapeutic effect of Icariin (10 mg/kg/day, from postnatal day (PND) 21 to PND 60 by gavage) on spatial memory deficits in rat induced by developmental BPA exposure (1 mg/kg/day, from embryonic to PND 60), demonstrating that Icariin can markedly improve spatial memory in BPA-exposed rat. Furthermore, intra-gastric administration of Icariin could attenuate abnormal hippocampal cell dispersion and loss, improved the dendritic spine density and Nissl bodies. Moreover, Icariin reversed BPA induced reduction of frequency of miniature excitatory postsynaptic currents(mEPSC) and decrease of Vesicular glutamate transporter 1(VGlut1). Collectively, Icariin could effectively rescue BPA-induced spatial memory impairment in male rats by preventing cell loss and reduction of dendritic spines in the hippocampus. In addition, we also found that VGlut1 is a critical target in the repair of BPA-induced spatial memory by Icariin. Thus, Icariin may be a promising therapeutic agent to attenuate BPA-induced spatial memory deficits.
Collapse
Affiliation(s)
- Danyang Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China
| | - Shu Ai
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China
| | - Chengqing Huang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China
| | - Zhi-Hua Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China.
| | - Hui-Li Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui Province 230009, China.
| |
Collapse
|
7
|
Kim SR, Eom Y, Lee SH. Comprehensive analysis of sex differences in the function and ultrastructure of hippocampal presynaptic terminals. Neurochem Int 2023; 169:105570. [PMID: 37451344 DOI: 10.1016/j.neuint.2023.105570] [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: 01/25/2023] [Revised: 05/08/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Sex differences in the brain, encompassing variations in specific brain structures, size, cognitive function, and synaptic connections, have been identified across numerous species. While previous research has explored sex differences in postsynaptic structures, synaptic plasticity, and hippocampus-dependent functions, the hippocampal presynaptic terminals remain largely uninvestigated. The hippocampus is a critical structure responsible for multiple brain functions. This study examined presynaptic differences in cultured hippocampal neurons derived from male and female mice using a combination of biochemical assays, functional analyses measuring exocytosis and endocytosis of synaptic vesicle proteins, ultrastructural analyses via electron microscopy, and presynaptic Ca2+-specific optical probes. Our findings revealed that female neurons exhibited a higher number of synaptic vesicles at presynaptic terminals compared to male neurons. However, no significant differences were observed in presynaptic protein expression, presynaptic terminal ultrastructure, synaptic vesicle exocytosis and endocytosis, or presynaptic Ca2+ alterations between male and female neurons.
Collapse
Affiliation(s)
- Sung Rae Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; Brain Research Core Facilities of Korea Brain Research Institute (KBRI), Daegu 41068, Republic of Korea.
| | - Yunkyung Eom
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
8
|
Xu H, Oses-Prieto JA, Khvotchev M, Jain S, Liang J, Burlingame A, Edwards RH. Adaptor protein AP-3 produces synaptic vesicles that release at high frequency by recruiting phospholipid flippase ATP8A1. Nat Neurosci 2023; 26:1685-1700. [PMID: 37723322 DOI: 10.1038/s41593-023-01434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 08/14/2023] [Indexed: 09/20/2023]
Abstract
Neural systems encode information in the frequency of action potentials, which is then decoded by synaptic transmission. However, the rapid, synchronous release of neurotransmitters depletes synaptic vesicles (SVs), limiting release at high firing rates. How then do synapses convey information about frequency? Here, we show in mouse hippocampal neurons and slices that the adaptor protein AP-3 makes a subset of SVs that respond specifically to high-frequency stimulation. Neurotransmitter transporters slot onto these SVs in different proportions, contributing to the distinct properties of release observed at different excitatory synapses. Proteomics reveals that AP-3 targets the phospholipid flippase ATP8A1 to SVs; loss of ATP8A1 recapitulates the defect in SV mobilization at high frequency observed with loss of AP-3. The mechanism involves recruitment of synapsin by the cytoplasmically oriented phosphatidylserine translocated by ATP8A1. Thus, ATP8A1 enables the subset of SVs made by AP-3 to release at high frequency.
Collapse
Affiliation(s)
- Hongfei Xu
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Mikhail Khvotchev
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Shweta Jain
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Jocelyn Liang
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Alma Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Robert H Edwards
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA.
| |
Collapse
|
9
|
Xi C, Shi X, Wang Y, He J, Jiang S, Niu B, Chen Y, Zhao F, Cao Z. Influence of bifenthrin exposure at different gestational stages on the neural development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115365. [PMID: 37597292 DOI: 10.1016/j.ecoenv.2023.115365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023]
Abstract
Perinatal exposure to bifenthrin (BF) alters neurodevelopment. However, the most susceptible time period to BF exposure and the possible mechanisms are not clear. In the current study, pregnant female mice were treated with BF (0.5 mg/kg/d) at three different stages [gestational day (GD) 0-5, 6-15 and 16-birth (B)] and neurologic deficits were evaluated in offspring mice. BF exposure at GD 16-B significantly altered the locomotor activity and caused learning and memory impairments in 6-week-old offspring. Gestational BF exposure also caused neuronal loss in the region of cornu ammonis of hippocampi of 6-week-old offspring. Interestingly, neurobehavioral impairments and neuronal loss were not observed in offspring at 10-week-old. BF exposure at GD 16-B also decreased protein levels of VGluT1, NR1 and NR2A while increased the protein levels of NR2B and VGAT1, as well as the gene levels of Il-1β, Il-6 and Tnf-α in hippocampi of 6-week-old offspring. Collectively, these data demonstrate that gestational exposure to a low dose BF causes neurodevelopmental deficits that remit with the age and the late-stage of pregnancy is the most susceptible time window to BF exposure. Imbalance in excitatory/inhibitory neuronal transmission, altered expression levels of NMDA receptors and increased neural inflammation may be associated with BF prenatal exposure-triggered neurobehavioral impairments.
Collapse
Affiliation(s)
- Chuchu Xi
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Xiaoqian Shi
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Yujing Wang
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Jing He
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Shan Jiang
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Bo Niu
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Ying Chen
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - Fang Zhao
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| | - Zhengyu Cao
- Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| |
Collapse
|
10
|
Wang Y, Zhu Y, Li W, Yan S, Li C, Ma K, Hu M, Du C, Fu L, Sun J, Zhang CX. Synaptotagmin-11 Inhibits Synaptic Vesicle Endocytosis via Endophilin A1. J Neurosci 2023; 43:6230-6248. [PMID: 37474308 PMCID: PMC10490507 DOI: 10.1523/jneurosci.1348-21.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 05/12/2023] [Accepted: 06/02/2023] [Indexed: 07/22/2023] Open
Abstract
Synaptic vesicle (SV) endocytosis is a critical and well-regulated process for the maintenance of neurotransmission. We previously reported that synaptotagmin-11 (Syt11), an essential non-Ca2+-binding Syt associated with brain diseases, inhibits neuronal endocytosis (Wang et al., 2016). Here, we found that Syt11 deficiency caused accelerated SV endocytosis and vesicle recycling under sustained stimulation and led to the abnormal membrane partition of synaptic proteins in mouse hippocampal boutons of either sex. Furthermore, our study revealed that Syt11 has direct but Ca2+-independent binding with endophilin A1 (EndoA1), a membrane curvature sensor and endocytic protein recruiter, with high affinity. EndoA1-knockdown significantly reversed Syt11-KO phenotype, identifying EndoA1 as a main inhibitory target of Syt11 during SV endocytosis. The N-terminus of EndoA1 and the C2B domain of Syt11 were responsible for this interaction. A peptide (amino acids 314-336) derived from the Syt11 C2B efficiently blocked Syt11-EndoA1 binding both in vitro and in vivo Application of this peptide inhibited SV endocytosis in WT hippocampal neurons but not in EndoA1-knockdown neurons. Moreover, intracellular application of this peptide in mouse calyx of Held terminals of either sex effectively hampered both fast and slow SV endocytosis at physiological temperature. We thus propose that Syt11 ensures the precision of protein retrieval during SV endocytosis by inhibiting EndoA1 function at neuronal terminals.SIGNIFICANCE STATEMENT Endocytosis is a key stage of synaptic vesicle (SV) recycling. SV endocytosis retrieves vesicular membrane and protein components precisely to support sustained neurotransmission. However, the molecular mechanisms underlying the regulation of SV endocytosis remain elusive. Here, we reported that Syt11-KO accelerated SV endocytosis and impaired membrane partition of synaptic proteins. EndoA1 was identified as a main inhibitory target of Syt11 during SV endocytosis. Our study reveals a novel inhibitory mechanism of SV endocytosis in preventing hyperactivation of endocytosis, potentially safeguarding the recycling of synaptic proteins during sustained neurotransmission.
Collapse
Affiliation(s)
- Yalong Wang
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; CAS Key Laboratory of Brain Connectome and Manipulation; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Ying Zhu
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanru Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Shuxin Yan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Chao Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kunpeng Ma
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; CAS Key Laboratory of Brain Connectome and Manipulation; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
| | - Meiqin Hu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Cuilian Du
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Lei Fu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Jianyuan Sun
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; CAS Key Laboratory of Brain Connectome and Manipulation; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Claire Xi Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
11
|
Sansevrino R, Hoffmann C, Milovanovic D. Condensate biology of synaptic vesicle clusters. Trends Neurosci 2023; 46:293-306. [PMID: 36725404 DOI: 10.1016/j.tins.2023.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
Neuronal communication crucially relies on exocytosis of neurotransmitters from synaptic vesicles (SVs) which are clustered at synapses. To ensure reliable neurotransmitter release, synapses need to maintain an adequate pool of SVs at all times. Decades of research have established that SVs are clustered by synapsin 1, an abundant SV-associated phosphoprotein. The classical view postulates that SVs are crosslinked in a scaffold of protein-protein interactions between synapsins and their binding partners. Recent studies have shown that synapsins cluster SVs via liquid-liquid phase separation (LLPS), thus providing a new framework for the organization of the synapse. We discuss the evidence for phase separation of SVs, emphasizing emerging questions related to its regulation, specificity, and reversibility.
Collapse
Affiliation(s)
- Roberto Sansevrino
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Christian Hoffmann
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany.
| |
Collapse
|
12
|
Time-dependent and selective microglia-mediated removal of spinal synapses in neuropathic pain. Cell Rep 2023; 42:112010. [PMID: 36656715 DOI: 10.1016/j.celrep.2023.112010] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 09/20/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Neuropathic pain is a debilitating condition resulting from damage to the nervous system. Imbalance of spinal excitation and inhibition has been proposed to contribute to neuropathic pain. However, the structural basis of this imbalance remains unknown. Using a preclinical model of neuropathic pain, we show that microglia selectively engulf spinal synapses that are formed by central neurons and spare those of peripheral sensory neurons. Furthermore, we reveal that removal of inhibitory and excitatory synapses exhibits distinct temporal patterns, in which microglia-mediated inhibitory synapse removal precedes excitatory synapse removal. We also find selective and gradual increase in complement depositions on dorsal horn synapses that corresponds to the temporal pattern of microglial synapse pruning activity and type-specific synapse loss. Together, these results define a specific role for microglia in the progression of neuropathic pain pathogenesis and implicate these immune cells in structural remodeling of dorsal horn circuitry.
Collapse
|
13
|
Hendricks EL, Smith IR, Prates B, Barmaleki F, Liebl FLW. The CD63 homologs, Tsp42Ee and Tsp42Eg, restrict endocytosis and promote neurotransmission through differential regulation of synaptic vesicle pools. Front Cell Neurosci 2022; 16:957232. [PMID: 36072568 PMCID: PMC9441712 DOI: 10.3389/fncel.2022.957232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022] Open
Abstract
The Tetraspanin (Tsp), CD63, is a transmembrane component of late endosomes and facilitates vesicular trafficking through endosomal pathways. Despite being widely expressed in the human brain and localized to late endosomes, CD63's role in regulating endo- and exocytic cycling at the synapse has not been investigated. Synaptic vesicle pools are highly dynamic and disruptions in the mobilization and replenishment of these vesicle pools have adverse neuronal effects. We find that the CD63 homologs, Tsp42Ee and Tsp42Eg, are expressed at the Drosophila neuromuscular junction to regulate synaptic vesicle pools through both shared and unique mechanisms. Tsp42Ee and Tsp42Eg negatively regulate endocytosis and positively regulate neurotransmitter release. Both tsp mutants show impaired locomotion, reduced miniature endplate junctional current frequencies, and increased endocytosis. Expression of human CD63 in Drosophila neurons leads to impaired endocytosis suggesting the role of Tsps in endocytosis is conserved. We further show that Tsps influence the synaptic cytoskeleton and membrane composition by regulating Futsch loop formation and synaptic levels of SCAR and PI(4,5)P2. Finally, Tsp42Ee and Tsp42Eg influence the synaptic localization of several vesicle-associated proteins including Synapsin, Synaptotagmin, and Cysteine String Protein. Together, our results present a novel function for Tsps in the regulation of vesicle pools and provide insight into the molecular mechanisms of Tsp-related synaptic dysfunction.
Collapse
Affiliation(s)
| | | | | | | | - Faith L. W. Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, United States
| |
Collapse
|
14
|
Platzer K, Sticht H, Bupp C, Ganapathi M, Pereira EM, Le Guyader G, Bilan F, Henderson LB, Lemke JR, Taschenberger H, Brose N, Jamra RA, Wojcik SM. De novo missense variants in
SLC32A1
cause a developmental and epileptic encephalopathy due to impaired
GABAergic
neurotransmission. Ann Neurol 2022; 92:958-973. [DOI: 10.1002/ana.26485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Konrad Platzer
- Institute of Human Genetics University of Leipzig Medical Center Leipzig Germany
| | - Heinrich Sticht
- Institute of Biochemistry, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| | - Caleb Bupp
- Spectrum Health Medical Genetics Grand Rapids MI USA
| | - Mythily Ganapathi
- Department of Pathology and Cell Biology Columbia University Medical Center New York NY USA
| | - Elaine M. Pereira
- Department of Pediatrics Columbia University Irving Medical Center New York NY USA
| | - Gwenaël Le Guyader
- Department of Genetics Poitiers University Hospital Center Poitiers Cedex France
| | - Frederic Bilan
- Department of Genetics Poitiers University Hospital Center Poitiers Cedex France
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC) INSERM U1084 University of Poitiers Poitiers France
| | | | - Johannes R. Lemke
- Institute of Human Genetics University of Leipzig Medical Center Leipzig Germany
- Center for Rare Diseases University of Leipzig Medical Center Leipzig Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology Max Planck Institute for Multidisciplinary Sciences City Campus, Göttingen Germany
| | - Nils Brose
- Department of Molecular Neurobiology Max Planck Institute for Multidisciplinary Sciences City Campus, Göttingen Germany
| | - Rami Abou Jamra
- Institute of Human Genetics University of Leipzig Medical Center Leipzig Germany
| | - Sonja M. Wojcik
- Department of Molecular Neurobiology Max Planck Institute for Multidisciplinary Sciences City Campus, Göttingen Germany
| |
Collapse
|
15
|
Quintanilla J, Jia Y, Lauterborn JC, Pruess BS, Le AA, Cox CD, Gall CM, Lynch G, Gunn BG. Novel types of frequency filtering in the lateral perforant path projections to dentate gyrus. J Physiol 2022; 600:3865-3896. [PMID: 35852108 PMCID: PMC9513824 DOI: 10.1113/jp283012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/26/2022] [Indexed: 11/08/2022] Open
Abstract
Despite its evident importance to learning theory and models, the manner in which the lateral perforant path (LPP) transforms signals from entorhinal cortex to hippocampus is not well understood. The present studies measured synaptic responses in the dentate gyrus (DG) of adult mouse hippocampal slices during different patterns of LPP stimulation. Theta (5 Hz) stimulation produced a modest within-train facilitation that was markedly enhanced at the level of DG output. Gamma (50 Hz) activation resulted in a singular pattern with initial synaptic facilitation being followed by a progressively greater depression. DG output was absent after only two pulses. Reducing release probability with low extracellular calcium instated frequency facilitation to gamma stimulation while long-term potentiation, which increases release by LPP terminals, enhanced within-train depression. Relatedly, per terminal concentrations of VGLUT2, a vesicular glutamate transporter associated with high release probability, were much greater in the LPP than in CA3-CA1 connections. Attempts to circumvent the potent gamma filter using a series of short (three-pulse) 50 Hz trains spaced by 200 ms were only partially successful: composite responses were substantially reduced after the first burst, an effect opposite to that recorded in field CA1. The interaction between bursts was surprisingly persistent (>1.0 s). Low calcium improved throughput during theta/gamma activation but buffering of postsynaptic calcium did not. In all, presynaptic specializations relating to release probability produce an unusual but potent type of frequency filtering in the LPP. Patterned burst input engages a different type of filter with substrates that are also likely to be located presynaptically. KEY POINTS: The lateral perforant path (LPP)-dentate gyrus (DG) synapse operates as a low-pass filter, where responses to a train of 50 Hz, γ frequency activation are greatly suppressed. Activation with brief bursts of γ frequency information engages a secondary filter that persists for prolonged periods (lasting seconds). Both forms of LPP frequency filtering are influenced by presynaptic, as opposed to postsynaptic, processes; this contrasts with other hippocampal synapses. LPP frequency filtering is modified by the unique presynaptic long-term potentiation at this synapse. Computational simulations indicate that presynaptic factors associated with release probability and vesicle recycling may underlie the potent LPP-DG frequency filtering.
Collapse
Affiliation(s)
- Julian Quintanilla
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Yousheng Jia
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Julie C Lauterborn
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Benedict S Pruess
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Aliza A Le
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Conor D Cox
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Christine M Gall
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
- Departments of Neurobiology & Behavior, University of California, Irvine, CA, USA
| | - Gary Lynch
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
- Departments of Psychiatry & Human Behavior, University of California, Irvine, CA, USA
| | - Benjamin G Gunn
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| |
Collapse
|
16
|
Li H, Datunashvili M, Reyes RC, Voglmaier SM. Inositol hexakisphosphate kinases differentially regulate trafficking of vesicular glutamate transporters 1 and 2. Front Cell Neurosci 2022; 16:926794. [PMID: 35936490 PMCID: PMC9355605 DOI: 10.3389/fncel.2022.926794] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Inositol pyrophosphates have been implicated in cellular signaling and membrane trafficking, including synaptic vesicle (SV) recycling. Inositol hexakisphosphate kinases (IP6Ks) and their product, diphosphoinositol pentakisphosphate (PP-IP5 or IP7), directly and indirectly regulate proteins important in vesicle recycling by the activity-dependent bulk endocytosis pathway (ADBE). In the present study, we show that two isoforms, IP6K1 and IP6K3, are expressed in axons. The role of the kinases in SV recycling are investigated using pharmacologic inhibition, shRNA knockdown, and IP6K1 and IP6K3 knockout mice. Live-cell imaging experiments use optical reporters of SV recycling based on vesicular glutamate transporter isoforms, VGLUT1- and VGLUT2-pHluorins (pH), which recycle differently. VGLUT1-pH recycles by classical AP-2 dependent endocytosis under moderate stimulation conditions, while VGLUT2-pH recycles using AP-1 and AP-3 adaptor proteins as well. Using a short stimulus to release the readily releasable pool (RRP), we show that IP6K1 KO increases exocytosis of both VGLUT1-and VGLUT2-pH, while IP6K3 KO decreases the amount of both transporters in the RRP. In electrophysiological experiments we measure glutamate signaling with short stimuli and under the intense stimulation conditions that trigger bulk endocytosis. IP6K1 KO increases synaptic facilitation and IP6K3 KO decreases facilitation compared to wild type in CA1 hippocampal Schaffer collateral synapses. After intense stimulation, the rate of endocytosis of VGLUT2-pH, but not VGLUT1-pH, is increased by knockout, knockdown, and pharmacologic inhibition of IP6Ks. Thus IP6Ks differentially affect the endocytosis of two SV protein cargos that use different endocytic pathways. However, while IP6K1 KO and IP6K3 KO exert similar effects on endocytosis after stimulation, the isoforms exert different effects on exocytosis earlier in the stimulus and on the early phase of glutamate release. Taken together, the data indicate a role for IP6Ks both in exocytosis early in the stimulation period and in endocytosis, particularly under conditions that may utilize AP-1/3 adaptors.
Collapse
|
17
|
Rostamipour K, Talandashti R, Mehrnejad F. Atomistic insight into the luminal allosteric regulation of vesicular glutamate transporter 2 by chloride and protons: An
all‐atom
molecular dynamics simulation study. Proteins 2022; 90:2045-2057. [DOI: 10.1002/prot.26396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/27/2022] [Accepted: 06/28/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Kiana Rostamipour
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies University of Tehran Tehran Iran
| | - Reza Talandashti
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies University of Tehran Tehran Iran
| | - Faramarz Mehrnejad
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies University of Tehran Tehran Iran
| |
Collapse
|
18
|
Hori T, Takamori S. Physiological Perspectives on Molecular Mechanisms and Regulation of Vesicular Glutamate Transport: Lessons From Calyx of Held Synapses. Front Cell Neurosci 2022; 15:811892. [PMID: 35095427 PMCID: PMC8793065 DOI: 10.3389/fncel.2021.811892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/07/2021] [Indexed: 12/02/2022] Open
Abstract
Accumulation of glutamate, the primary excitatory neurotransmitter in the mammalian central nervous system, into presynaptic synaptic vesicles (SVs) depends upon three vesicular glutamate transporters (VGLUTs). Since VGLUTs are driven by a proton electrochemical gradient across the SV membrane generated by vacuolar-type H+-ATPases (V-ATPases), the rate of glutamate transport into SVs, as well as the amount of glutamate in SVs at equilibrium, are influenced by activities of both VGLUTs and V-ATPase. Despite emerging evidence that suggests various factors influencing glutamate transport by VGLUTs in vitro, little has been reported in physiological or pathological contexts to date. Historically, this was partially due to a lack of appropriate methods to monitor glutamate loading into SVs in living synapses. Furthermore, whether or not glutamate refilling of SVs can be rate-limiting for synaptic transmission is not well understood, primarily due to a lack of knowledge concerning the time required for vesicle reuse and refilling during repetitive stimulation. In this review, we first introduce a unique electrophysiological method to monitor glutamate refilling by VGLUTs in a giant model synapse from the calyx of Held in rodent brainstem slices, and we discuss the advantages and limitations of the method. We then introduce the current understanding of factors that potentially alter the amount and rate of glutamate refilling of SVs in this synapse, and discuss open questions from physiological viewpoints.
Collapse
Affiliation(s)
- Tetsuya Hori
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- *Correspondence: Tetsuya Hori Shigeo Takamori
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
- *Correspondence: Tetsuya Hori Shigeo Takamori
| |
Collapse
|
19
|
Fazekas CL, Szabó A, Török B, Bánrévi K, Correia P, Chaves T, Daumas S, Zelena D. A New Player in the Hippocampus: A Review on VGLUT3+ Neurons and Their Role in the Regulation of Hippocampal Activity and Behaviour. Int J Mol Sci 2022; 23:790. [PMID: 35054976 PMCID: PMC8775679 DOI: 10.3390/ijms23020790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 01/05/2023] Open
Abstract
Glutamate is the most abundant excitatory amino acid in the central nervous system. Neurons using glutamate as a neurotransmitter can be characterised by vesicular glutamate transporters (VGLUTs). Among the three subtypes, VGLUT3 is unique, co-localising with other "classical" neurotransmitters, such as the inhibitory GABA. Glutamate, manipulated by VGLUT3, can modulate the packaging as well as the release of other neurotransmitters and serve as a retrograde signal through its release from the somata and dendrites. Its contribution to sensory processes (including seeing, hearing, and mechanosensation) is well characterised. However, its involvement in learning and memory can only be assumed based on its prominent hippocampal presence. Although VGLUT3-expressing neurons are detectable in the hippocampus, most of the hippocampal VGLUT3 positivity can be found on nerve terminals, presumably coming from the median raphe. This hippocampal glutamatergic network plays a pivotal role in several important processes (e.g., learning and memory, emotions, epilepsy, cardiovascular regulation). Indirect information from anatomical studies and KO mice strains suggests the contribution of local VGLUT3-positive hippocampal neurons as well as afferentations in these events. However, further studies making use of more specific tools (e.g., Cre-mice, opto- and chemogenetics) are needed to confirm these assumptions.
Collapse
Affiliation(s)
- Csilla Lea Fazekas
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS) INSERM, Sorbonne Université, CNRS, 75005 Paris, France;
| | - Adrienn Szabó
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Bibiána Török
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Krisztina Bánrévi
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
| | - Pedro Correia
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Tiago Chaves
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Stéphanie Daumas
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS) INSERM, Sorbonne Université, CNRS, 75005 Paris, France;
| | - Dóra Zelena
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
20
|
Lindström SH, Sundberg SC, Larsson M, Andersson FK, Broman J, Granseth B. VGluT1 Deficiency Impairs Visual Attention and Reduces the Dynamic Range of Short-Term Plasticity at Corticothalamic Synapses. Cereb Cortex 2021; 30:1813-1829. [PMID: 31711131 PMCID: PMC7132919 DOI: 10.1093/cercor/bhz204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/08/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
The most common excitatory neurotransmitter in the central nervous system, glutamate, is loaded into synaptic vesicles by vesicular glutamate transporters (VGluTs). The primary isoforms, VGluT1 and 2, are expressed in complementary patterns throughout the brain and correlate with short-term synaptic plasticity. VGluT1 deficiency is observed in certain neurological disorders, and hemizygous (VGluT1+/−) mice display increased anxiety and depression, altered sensorimotor gating, and impairments in learning and memory. The synaptic mechanisms underlying these behavioral deficits are unknown. Here, we show that VGluT1+/− mice had decreased visual processing speeds during a sustained visual-spatial attention task. Furthermore, in vitro recordings of corticothalamic (CT) synapses revealed dramatic reductions in short-term facilitation, increased initial release probability, and earlier synaptic depression in VGluT1+/− mice. Our electron microscopy results show that VGluT1 concentration is reduced at CT synapses of hemizygous mice, but other features (such as vesicle number and active zone size) are unchanged. We conclude that VGluT1-haploinsuficiency decreases the dynamic range of gain modulation provided by CT feedback to the thalamus, and this deficiency contributes to the observed attentional processing deficit. We further hypothesize that VGluT1 concentration regulates release probability by applying a “brake” to an unidentified presynaptic protein that typically acts as a positive regulator of release.
Collapse
Affiliation(s)
- Sarah H Lindström
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Sofie C Sundberg
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Max Larsson
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Fredrik K Andersson
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Jonas Broman
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Björn Granseth
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| |
Collapse
|
21
|
Zhou JW, Zhao M, Rang WL, Zhang XY, Liu ZM, Zhang LR, Wang TX, Wu CT, Cheng XR, Zhou WX. Proteome Profiling Identified Amyloid-β Protein Precursor as a Novel Binding Partner and Modulator of VGLUT1. J Alzheimers Dis 2021; 81:981-1038. [PMID: 33896843 DOI: 10.3233/jad-210117] [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: 11/15/2022]
Abstract
BACKGROUND The toxicity of excessive glutamate release has been implicated in various acute and chronic neurodegenerative conditions. Vesicular glutamate transporters (VGLUTs) are the major mediators for the uptake of glutamate into synaptic vesicles. However, the dynamics and mechanism of this process in glutamatergic neurons are still largely unknown. OBJECTIVE This study aimed to investigate the candidate protein partners of VGLUT1 and their regulatory roles in the vesicles in rat brain. METHODS Pull down assay, co-immunoprecipitation assay, or split-ubiquitin membrane yeast two hybrid screening coupled with nanoRPLC-MS/MS were used to identify the candidate protein partners of VGLUT1 in the vesicles in rat brain. The in vitro and in vivo models were used to test effects of AβPP, Atp6ap2, Gja1, and Synataxin on VGLUT1 expression. RESULTS A total of 255 and 225 proteins and 172 known genes were identified in the pull down assay, co-immunoprecipitation assay, or split-ubiquitin yeast two-hybrid screening respectively. The physiological interactions of SV2A, Syntaxin 12, Gja1, AβPP, and Atp6ap2 to VGLUT1 were further confirmed. Knockdown of Atp6ap2, Gja1, and Synataxin increased VGLUT1 mRNA expression and only knockdown of AβPP increased both mRNA and protein levels of VGLUT1 in PC12 cells. The regulatory function of AβPP on VGLUT1 expression was further confirmed in the in vitro and in vivo models. CONCLUSION These results elucidate that the AβPP and VGLUT1 interacts at vesicular level and AβPP plays a role in the regulation of VGLUT1 expression which is essential for maintaining vesicular activities.
Collapse
Affiliation(s)
- Jin-Wu Zhou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Man Zhao
- Department of Blood Transfusion, Chinese PLA General Hospital, Beijing, China
| | - Wen-Liang Rang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao-Yan Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhen-Ming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Liang-Ren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Tong-Xing Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Chu-Tse Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao-Rui Cheng
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Wen-Xia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| |
Collapse
|
22
|
Nakakubo Y, Abe S, Yoshida T, Takami C, Isa M, Wojcik SM, Brose N, Takamori S, Hori T. Vesicular Glutamate Transporter Expression Ensures High-Fidelity Synaptic Transmission at the Calyx of Held Synapses. Cell Rep 2021; 32:108040. [PMID: 32814044 DOI: 10.1016/j.celrep.2020.108040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 07/01/2020] [Accepted: 07/24/2020] [Indexed: 12/26/2022] Open
Abstract
Recycling of synaptic vesicles (SVs) at presynaptic terminals is required for sustained neurotransmitter release. Although SV endocytosis is a rate-limiting step for synaptic transmission, it is unclear whether the rate of the subsequent SV refilling with neurotransmitter also influences synaptic transmission. By analyzing vesicular glutamate transporter 1 (VGLUT1)-deficient calyx of Held synapses, in which both VGLUT1 and VGLUT2 are co-expressed in wild-type situation, we found that VGLUT1 loss causes a drastic reduction in SV refilling rate down to ∼25% of wild-type values, with only subtle changes in basic synaptic parameters. Strikingly, VGLUT1-deficient synapses exhibited abnormal synaptic failures within a few seconds during high-frequency repetitive firing, which was recapitulated by manipulating presynaptic Cl- concentrations to retard SV refilling. Our data show that the speed of SV refilling can be rate limiting for synaptic transmission under certain conditions that entail reduced VGLUT levels during development as well as various neuropathological processes.
Collapse
Affiliation(s)
- Yutaro Nakakubo
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan
| | - Saeka Abe
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan
| | - Tomofumi Yoshida
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Chihiro Takami
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Masayuki Isa
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Sonja M Wojcik
- Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Göttingen 37075, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Göttingen 37075, Germany
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan.
| | - Tetsuya Hori
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan.
| |
Collapse
|
23
|
Noise Exposure Alters Glutamatergic and GABAergic Synaptic Connectivity in the Hippocampus and Its Relevance to Tinnitus. Neural Plast 2021; 2021:8833087. [PMID: 33510780 PMCID: PMC7822664 DOI: 10.1155/2021/8833087] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 12/16/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence implicates a role for brain structures outside the ascending auditory pathway in tinnitus, the phantom perception of sound. In addition to other factors such as age-dependent hearing loss, high-level sound exposure is a prominent cause of tinnitus. Here, we examined how noise exposure altered the distribution of excitatory and inhibitory synaptic inputs in the guinea pig hippocampus and determined whether these changes were associated with tinnitus. In experiment one, guinea pigs were overexposed to unilateral narrow-band noise (98 dB SPL, 2 h). Two weeks later, the density of excitatory (VGLUT-1/2) and inhibitory (VGAT) synaptic terminals in CA1, CA3, and dentate gyrus hippocampal subregions was assessed by immunohistochemistry. Overall, VGLUT-1 density primarily increased, while VGAT density decreased significantly in many regions. Then, to assess whether the noise-induced alterations were persistent and related to tinnitus, experiment two utilized a noise-exposure paradigm shown to induce tinnitus and assessed tinnitus development which was assessed using gap-prepulse inhibition of the acoustic startle (GPIAS). Twelve weeks after sound overexposure, changes in excitatory synaptic terminal density had largely recovered regardless of tinnitus status, but the recovery of GABAergic terminal density was dramatically different in animals expressing tinnitus relative to animals resistant to tinnitus. In resistant animals, inhibitory synapse density recovered to preexposure levels, but in animals expressing tinnitus, inhibitory synapse density remained chronically diminished. Taken together, our results suggest that noise exposure induces striking changes in the balance of excitatory and inhibitory synaptic inputs throughout the hippocampus and reveal a potential role for rebounding inhibition in the hippocampus as a protective factor leading to tinnitus resilience.
Collapse
|
24
|
Casamento A, Boucrot E. Molecular mechanism of Fast Endophilin-Mediated Endocytosis. Biochem J 2020; 477:2327-2345. [PMID: 32589750 PMCID: PMC7319585 DOI: 10.1042/bcj20190342] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/11/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
Endocytosis mediates the cellular uptake of micronutrients and cell surface proteins. Clathrin-mediated endocytosis (CME) is the housekeeping pathway in resting cells but additional Clathrin-independent endocytic (CIE) routes, including Fast Endophilin-Mediated Endocytosis (FEME), internalize specific cargoes and support diverse cellular functions. FEME is part of the Dynamin-dependent subgroup of CIE pathways. Here, we review our current understanding of the molecular mechanism of FEME. Key steps are: (i) priming, (ii) cargo selection, (iii) membrane curvature and carrier formation, (iv) membrane scission and (v) cytosolic transport. All steps are controlled by regulatory mechanisms mediated by phosphoinositides and by kinases such as Src, LRRK2, Cdk5 and GSK3β. A key feature of FEME is that it is not constitutively active but triggered upon the stimulation of selected cell surface receptors by their ligands. In resting cells, there is a priming cycle that concentrates Endophilin into clusters on discrete locations of the plasma membrane. In the absence of receptor activation, the patches quickly abort and new cycles are initiated nearby, constantly priming the plasma membrane for FEME. Upon activation, receptors are swiftly sorted into pre-existing Endophilin clusters, which then bud to form FEME carriers within 10 s. We summarize the hallmarks of FEME and the techniques and assays required to identify it. Next, we review similarities and differences with other CIE pathways and proposed cargoes that may use FEME to enter cells. Finally, we submit pending questions and future milestones and discuss the exciting perspectives that targeting FEME may boost treatments against cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessandra Casamento
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, U.K
- Institute of Structural and Molecular Biology, Birkbeck College, Malet Street, London WC1E 7HX, U.K
| |
Collapse
|
25
|
Pietrancosta N, Djibo M, Daumas S, El Mestikawy S, Erickson JD. Molecular, Structural, Functional, and Pharmacological Sites for Vesicular Glutamate Transporter Regulation. Mol Neurobiol 2020; 57:3118-3142. [PMID: 32474835 PMCID: PMC7261050 DOI: 10.1007/s12035-020-01912-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) control quantal size of glutamatergic transmission and have been the center of numerous studies over the past two decades. VGLUTs contain two independent transport modes that facilitate glutamate packaging into synaptic vesicles and phosphate (Pi) ion transport into the synaptic terminal. While a transmembrane proton electrical gradient established by a vacuolar-type ATPase powers vesicular glutamate transport, recent studies indicate that binding sites and flux properties for chloride, potassium, and protons within VGLUTs themselves regulate VGLUT activity as well. These intrinsic ionic binding and flux properties of VGLUTs can therefore be modulated by neurophysiological conditions to affect levels of glutamate available for release from synapses. Despite their extraordinary importance, specific and high-affinity pharmacological compounds that interact with these sites and regulate VGLUT function, distinguish between the various modes of transport, and the different isoforms themselves, are lacking. In this review, we provide an overview of the physiologic sites for VGLUT regulation that could modulate glutamate release in an over-active synapse or in a disease state.
Collapse
Affiliation(s)
- Nicolas Pietrancosta
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France. .,Laboratoire des Biomolécules, Sorbonne Université, CNRS, ENS, LBM, 75005, Paris, France.
| | - Mahamadou Djibo
- Sorbonne Paris Cité, Université Paris Descartes, LCBPT, UMR 8601, 75006, Paris, France
| | - Stephanie Daumas
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Salah El Mestikawy
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France. .,Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 boulevard Lasalle, Verdun, Montreal, QC, Canada.
| | - Jeffrey D Erickson
- Neuroscience Center, Louisiana State University, New Orleans, LA, 70112, USA. .,Department of Pharmacology, Louisiana State University, New Orleans, LA, 70112, USA.
| |
Collapse
|
26
|
Luo S, Hou Y, Zhang Y, Ma T, Shao W, Xiao C. Adenosine A 2A Receptor Agonist PSB-0777 Modulates Synaptic Proteins and AMPA Receptor Expression in a Dose- and Time-Dependent Manner in Rat Primary Cortical Neurons. Biol Pharm Bull 2020; 43:1159-1171. [PMID: 32448843 DOI: 10.1248/bpb.b19-01007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regulating synaptic formation and transmission is critical for the physiology and pathology of psychiatric disorders. The adenosine A2A receptor subtype has attracted widespread attention as a key regulator of neuropsychiatric activity, neuroprotection and injury. In this study, we systematically investigated the regulatory effects of a novel A2A receptor agonist, PSB-0777, on the expression of synaptic proteins and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors (AMPA receptors) at the cellular level in a time- and dose-dependent manner. After 30 min of high-dose PSB-0777 stimulation, the expression of Synapsin-1 (Syn-1), postsynaptic density protein 95 (PSD95), and AMPA receptors and the number of synapses were rapidly and significantly increased in rat primary cortical neurons compared with the control. Sustained elevation was found in the low and medium-dose groups after 24 h and 3 d of treatment. In contrast, after stimulation with PSB-0777 for 3 consecutive days, the expression of Syn-1 was decreased, and PSD95, AMPA receptors and the number of synapses were no longer increased in the high-dose group. Our study focuses on the detailed and systematic regulation of synaptic proteins and AMPA receptors by an A2A receptor agonist, PSB-0777, which may result in both beneficial and detrimental effects on neurotransmission and neuroprotection and may contribute to the pathophysiology of psychiatric disorders related to A2A receptors. These experimental data may contribute to understanding of the mechanisms for neuroprotective and therapeutic effect of A2A receptor agonists.
Collapse
Affiliation(s)
| | | | | | - Tengfei Ma
- Yunnan University, School of Life Sciences, Center for Life Sciences
| | - Wenping Shao
- First Affiliated Hospital of Kunming Medical University, Department of Medical Examination
| | | |
Collapse
|
27
|
Nanostructural Diversity of Synapses in the Mammalian Spinal Cord. Sci Rep 2020; 10:8189. [PMID: 32424125 PMCID: PMC7235094 DOI: 10.1038/s41598-020-64874-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 04/21/2020] [Indexed: 11/25/2022] Open
Abstract
Functionally distinct synapses exhibit diverse and complex organisation at molecular and nanoscale levels. Synaptic diversity may be dependent on developmental stage, anatomical locus and the neural circuit within which synapses reside. Furthermore, astrocytes, which align with pre and post-synaptic structures to form ‘tripartite synapses’, can modulate neural circuits and impact on synaptic organisation. In this study, we aimed to determine which factors impact the diversity of excitatory synapses throughout the lumbar spinal cord. We used PSD95-eGFP mice, to visualise excitatory postsynaptic densities (PSDs) using high-resolution and super-resolution microscopy. We reveal a detailed and quantitative map of the features of excitatory synapses in the lumbar spinal cord, detailing synaptic diversity that is dependent on developmental stage, anatomical region and whether associated with VGLUT1 or VGLUT2 terminals. We report that PSDs are nanostructurally distinct between spinal laminae and across age groups. PSDs receiving VGLUT1 inputs also show enhanced nanostructural complexity compared with those receiving VGLUT2 inputs, suggesting pathway-specific diversity. Finally, we show that PSDs exhibit greater nanostructural complexity when part of tripartite synapses, and we provide evidence that astrocytic activation enhances PSD95 expression. Taken together, these results provide novel insights into the regulation and diversification of synapses across functionally distinct spinal regions and advance our general understanding of the ‘rules’ governing synaptic nanostructural organisation.
Collapse
|
28
|
Eriksen J, Li F, Edwards RH. The mechanism and regulation of vesicular glutamate transport: Coordination with the synaptic vesicle cycle. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183259. [PMID: 32147354 DOI: 10.1016/j.bbamem.2020.183259] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 01/30/2023]
Abstract
The transport of classical neurotransmitters into synaptic vesicles generally relies on a H+ electrochemical gradient (∆μH+). Synaptic vesicle uptake of glutamate depends primarily on the electrical component ∆ψ as the driving force, rather than the chemical component ∆pH. However, the vesicular glutamate transporters (VGLUTs) belong to the solute carrier 17 (SLC17) family, which includes closely related members that function as H+ cotransporters. Recent work has also shown that the VGLUTs undergo allosteric regulation by H+ and Cl-, and exhibit an associated Cl- conductance. These properties appear to coordinate VGLUT activity with the large ionic shifts that accompany the rapid recycling of synaptic vesicles driven by neural activity. Recent structural information also suggests common mechanisms that underlie the apparently divergent function of SLC17 family members, and that confer allosteric regulation.
Collapse
Affiliation(s)
- Jacob Eriksen
- Department of Physiology, UCSF School of Medicine, United States of America; Department of Neurology, UCSF School of Medicine, United States of America
| | - Fei Li
- Department of Physiology, UCSF School of Medicine, United States of America; Department of Neurology, UCSF School of Medicine, United States of America
| | - Robert H Edwards
- Department of Physiology, UCSF School of Medicine, United States of America; Department of Neurology, UCSF School of Medicine, United States of America.
| |
Collapse
|
29
|
Adeli A, Zendehdel M, Babapour V, Panahi N. Interaction between leptin and glutamatergic system on food intake regulation in neonatal chicken: role of NMDA and AMPA receptors. Int J Neurosci 2020; 130:713-721. [PMID: 31813315 DOI: 10.1080/00207454.2019.1702983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Objective: The aim of the current study was to determine the possible interaction of the central leptin and Glutamatergic systems on feeding behavior in neonatal 3-hours food deprived (FD3) broilers chickens.Methods: In experiment 1, FD3 chicken received intracerebroventricular (ICV) injection of control solution (group i) and 2.5, 5 and 10 µg of Leptin (groups ii-iv). In experiment 2, FD3 chicken were ICV injected with (group i) control solution and groups ii-iv with 2.5, 5 and 10 nmol of AG-490 (JAK2 antagonist). In experiment 3, injections were (i) control solution, (ii) Leptin (10 µg), (iii) AG-490 (2.5 nmol) and (iv) Leptin + AG-490. In experiment 4, broiler chickens were ICV injected with (i) control solution, (ii) Leptin (10 µg), (iii) MK-801 (NMDA glutamate receptors antagonist; 15 nmol) and (iv) Leptin + MK-801. Experiments 5-9 were similar to experiment 1, except chicken were ICV injected with CNQX (AMPA receptor antagonist, 390 nmol), UBP-302 (Kainate receptor antagonist, 390 nmol), AIDA (mGluR1 antagonist, 2 nmol), LY341495 (mGluR2 antagonist, 150 nmol) and UBP1112 (mGluR3 antagonist, 2 nmol) instead of MK-801. Then, food intake was measured until 120 min after injection.Results: ICV injection of leptin (2.5, 5 and 10 µg) significantly decreased food intake in a dose dependent manner (p < 0.05). Also, ICV injection of the JAK2 antagonist (2.5, 5 and 10 nmol) had hyperphagic effect in chicken (p < 0.05). Co-administration of leptin + AG-490, partially decreased leptin-induced hypophagia in broiler chicken (p < 0.05). In addition, co-injection of leptin + MK-801 significalty inhibited leptin-induced hypophagia in neonatal chicken (p < 0.05). Also, co-administration of leptin + CNQX partially attenuated hypophagic effect of leptin in chicken (p < 0.05).Conclusion: The results of present study suggest that leptin has hypophagic effect in neonatal chicken and this effect is probably mediated via NMDA and AMPA glutamatergic receptors.
Collapse
Affiliation(s)
- Amin Adeli
- Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Morteza Zendehdel
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahab Babapour
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Negar Panahi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
30
|
Wang J, Xu W, Kong Y, Huang J, Ding Z, Deng M, Guo Q, Zou W. SNAP-25 Contributes to Neuropathic Pain by Regulation of VGLuT2 Expression in Rats. Neuroscience 2019; 423:86-97. [PMID: 31705888 DOI: 10.1016/j.neuroscience.2019.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/23/2022]
Abstract
Synaptosomal-associated protein 25 (SNAP-25) plays an important role in neuropathic pain. However, the underlying mechanism is largely unknown. Vesicular glutamate transporter 2 (VGluT2) is an isoform of vesicular glutamate transporters that controls the storage and release of glutamate. In the present study, we found the expression levels of VGluT2 correlated with the upregulation of SNAP-25 in the spinal cord of rats following chronic constriction injury (CCI)-induced neuropathic pain. Cleavage of SNAP-25 by Botulinum toxin A (BoNT/A) attenuated mechanical allodynia, downregulated the expression of VGluT2 and reduced glutamate release. Overexpression of VGluT2 abolished the antinociceptive effect of BoNT/A. Upregulation of SNAP-25 in naive rats increased VGluT2 expression and induced pain-responsive behaviors. In pheochromocytoma (PC12) cells, the expression of VGluT2 was also depended on SNAP-25 dysregulation. Moreover, we found VGluT2 was involved in SNAP-25-mediated regulation of astrocyte expression and activation of the PKA/p-CREB pathway mediated the upregulation of SNAP-25 in neuropathic pain. The findings of our study indicate that VGluT2 contributes to the effect of SNAP-25 in maintaining the development of neuropathic pain and suggests a novel mechanism underlying SNAP-25 regulation of neuropathic pain.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wei Xu
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, China
| | - Yan Kong
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiangju Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhuofeng Ding
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Meiling Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
31
|
Zhang XM, François U, Silm K, Angelo MF, Fernandez-Busch MV, Maged M, Martin C, Bernard V, Cordelières FP, Deshors M, Pons S, Maskos U, Bemelmans AP, Wojcik SM, El Mestikawy S, Humeau Y, Herzog E. A proline-rich motif on VGLUT1 reduces synaptic vesicle super-pool and spontaneous release frequency. eLife 2019; 8:50401. [PMID: 31663854 PMCID: PMC6861006 DOI: 10.7554/elife.50401] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/27/2019] [Indexed: 12/29/2022] Open
Abstract
Glutamate secretion at excitatory synapses is tightly regulated to allow for the precise tuning of synaptic strength. Vesicular Glutamate Transporters (VGLUT) accumulate glutamate into synaptic vesicles (SV) and thereby regulate quantal size. Further, the number of release sites and the release probability of SVs maybe regulated by the organization of active-zone proteins and SV clusters. In the present work, we uncover a mechanism mediating an increased SV clustering through the interaction of VGLUT1 second proline-rich domain, endophilinA1 and intersectin1. This strengthening of SV clusters results in a combined reduction of axonal SV super-pool size and miniature excitatory events frequency. Our findings support a model in which clustered vesicles are held together through multiple weak interactions between Src homology three and proline-rich domains of synaptic proteins. In mammals, VGLUT1 gained a proline-rich sequence that recruits endophilinA1 and turns the transporter into a regulator of SV organization and spontaneous release.
Collapse
Affiliation(s)
- Xiao Min Zhang
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France.,Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Urielle François
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| | - Kätlin Silm
- Neuroscience Paris Seine NPS, Université Pierre et Marie Curie INSERM U1130 CNRS UMR8246, Paris, France
| | - Maria Florencia Angelo
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| | - Maria Victoria Fernandez-Busch
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| | - Mona Maged
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| | - Christelle Martin
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| | - Véronique Bernard
- Neuroscience Paris Seine NPS, Université Pierre et Marie Curie INSERM U1130 CNRS UMR8246, Paris, France
| | - Fabrice P Cordelières
- Bordeaux Imaging Center, Université de Bordeaux, CNRS UMS 3420, INSERM US4, Bordeaux, France
| | - Melissa Deshors
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| | - Stéphanie Pons
- Institut Pasteur, CNRS UMR 3571, Unité NISC, Paris, France
| | - Uwe Maskos
- Institut Pasteur, CNRS UMR 3571, Unité NISC, Paris, France
| | - Alexis Pierre Bemelmans
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de laRecherche Fondamentale (DRF), Institut de Biologie François Jacob (IBFJ), MolecularImaging Research Center (MIRCen), Fontenay-aux-Roses, France
| | - Sonja M Wojcik
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Salah El Mestikawy
- Neuroscience Paris Seine NPS, Université Pierre et Marie Curie INSERM U1130 CNRS UMR8246, Paris, France.,Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Yann Humeau
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| | - Etienne Herzog
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience CNRS UMR 5297, Bordeaux, France
| |
Collapse
|
32
|
Brockmann MM, Maglione M, Willmes CG, Stumpf A, Bouazza BA, Velasquez LM, Grauel MK, Beed P, Lehmann M, Gimber N, Schmoranzer J, Sigrist SJ, Rosenmund C, Schmitz D. RIM-BP2 primes synaptic vesicles via recruitment of Munc13-1 at hippocampal mossy fiber synapses. eLife 2019; 8:43243. [PMID: 31535974 PMCID: PMC6752948 DOI: 10.7554/elife.43243] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 08/12/2019] [Indexed: 12/29/2022] Open
Abstract
All synapses require fusion-competent vesicles and coordinated Ca2+-secretion coupling for neurotransmission, yet functional and anatomical properties are diverse across different synapse types. We show that the presynaptic protein RIM-BP2 has diversified functions in neurotransmitter release at different central murine synapses and thus contributes to synaptic diversity. At hippocampal pyramidal CA3-CA1 synapses, RIM-BP2 loss has a mild effect on neurotransmitter release, by only regulating Ca2+-secretion coupling. However, at hippocampal mossy fiber synapses, RIM-BP2 has a substantial impact on neurotransmitter release by promoting vesicle docking/priming and vesicular release probability via stabilization of Munc13-1 at the active zone. We suggest that differences in the active zone organization may dictate the role a protein plays in synaptic transmission and that differences in active zone architecture is a major determinant factor in the functional diversity of synapses.
Collapse
Affiliation(s)
- Marisa M Brockmann
- Institut für Neurophysiologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Marta Maglione
- Freie Universität Berlin, Institut für Biologie, Berlin, Germany.,Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | | | - Alexander Stumpf
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Boris A Bouazza
- Institut für Neurophysiologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Laura M Velasquez
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - M Katharina Grauel
- Institut für Neurophysiologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Prateep Beed
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Niclas Gimber
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Stephan J Sigrist
- Freie Universität Berlin, Institut für Biologie, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany.,DZNE, German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Christian Rosenmund
- Institut für Neurophysiologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - Dietmar Schmitz
- NeuroCure Cluster of Excellence, Berlin, Germany.,DZNE, German Center for Neurodegenerative Diseases, Berlin, Germany.,Neuroscience Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
33
|
Watanabe S, Mamer LE, Raychaudhuri S, Luvsanjav D, Eisen J, Trimbuch T, Söhl-Kielczynski B, Fenske P, Milosevic I, Rosenmund C, Jorgensen EM. Synaptojanin and Endophilin Mediate Neck Formation during Ultrafast Endocytosis. Neuron 2019; 98:1184-1197.e6. [PMID: 29953872 DOI: 10.1016/j.neuron.2018.06.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/12/2018] [Accepted: 06/04/2018] [Indexed: 11/19/2022]
Abstract
Ultrafast endocytosis generates vesicles from the plasma membrane as quickly as 50 ms in hippocampal neurons following synaptic vesicle fusion. The molecular mechanism underlying the rapid maturation of these endocytic pits is not known. Here we demonstrate that synaptojanin-1, and its partner endophilin-A, function in ultrafast endocytosis. In the absence of synaptojanin or endophilin, the membrane is rapidly invaginated, but pits do not become constricted at the base. The 5-phosphatase activity of synaptojanin is involved in formation of the neck, but 4-phosphatase is not required. Nevertheless, these pits are eventually cleaved into vesicles; within a 30-s interval, synaptic endosomes form and are resolved by clathrin-mediated budding. Then synaptojanin and endophilin function at a second step to aid with the removal of clathrin coats from the regenerated vesicles. These data together suggest that synaptojanin and endophilin can mediate membrane remodeling on a millisecond timescale during ultrafast endocytosis.
Collapse
Affiliation(s)
- Shigeki Watanabe
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA.
| | - Lauren Elizabeth Mamer
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany; The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sumana Raychaudhuri
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Delgermaa Luvsanjav
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Julia Eisen
- Barnard College of Columbia University, New York, NY, USA
| | - Thorsten Trimbuch
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Berit Söhl-Kielczynski
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Pascal Fenske
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ira Milosevic
- Synaptic Vesicle Dynamics, European Neuroscience Institute, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Christian Rosenmund
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Erik M Jorgensen
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT 84112-0840, USA.
| |
Collapse
|
34
|
Fenske P, Grauel MK, Brockmann MM, Dorrn AL, Trimbuch T, Rosenmund C. Autaptic cultures of human induced neurons as a versatile platform for studying synaptic function and neuronal morphology. Sci Rep 2019; 9:4890. [PMID: 30894602 PMCID: PMC6427022 DOI: 10.1038/s41598-019-41259-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/05/2019] [Indexed: 02/08/2023] Open
Abstract
Recently developed technology to differentiate induced pluripotent stem cells (iPSCs) into human induced neurons (iNs) provides an exciting opportunity to study the function of human neurons. However, functional characterisations of iNs have been hampered by the reliance on mass culturing protocols which do not allow assessment of synaptic release characteristics and neuronal morphology at the individual cell level with quantitative precision. Here, we have developed for the first time a protocol to generate autaptic cultures of iPSC-derived iNs. We show that our method efficiently generates mature, autaptic iNs with robust spontaneous and action potential-driven synaptic transmission. The synaptic responses are sensitive to modulation by metabotropic receptor agonists as well as potentiation by acute phorbol ester application. Finally, we demonstrate loss of evoked and spontaneous release by Unc13A knockdown. This culture system provides a versatile platform allowing for quantitative and integrative assessment of morphophysiological and molecular parameters underlying human synaptic transmission.
Collapse
Affiliation(s)
- Pascal Fenske
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - M Katharina Grauel
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Marisa M Brockmann
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Anja L Dorrn
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Christian Rosenmund
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany. .,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany. .,Berlin Institute of Health, Anna-Louise-Karsch-Straße 2, 10178, Berlin, Germany.
| |
Collapse
|
35
|
Corponi F, Bonassi S, Vieta E, Albani D, Frustaci A, Ducci G, Landi S, Boccia S, Serretti A, Fabbri C. Genetic basis of psychopathological dimensions shared between schizophrenia and bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:23-29. [PMID: 30149091 DOI: 10.1016/j.pnpbp.2018.08.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/25/2022]
Abstract
Shared genetic vulnerability between schizophrenia (SCZ) and bipolar disorder (BP) was demonstrated, but the genetic underpinnings of specific symptom domains are unclear. This study investigated which genes and gene sets may modulate specific psychopathological domains and if genome-wide significant loci previously associated with SCZ or BP may play a role. Genome-wide data were available in patients with SCZ (n = 226) or BP (n = 228). Phenotypes under investigation were depressive and positive symptoms severity, suicidal ideation, onset age and substance use disorder comorbidity. Genome-wide analyses were performed at gene and gene set level, while 148 genome-wide significant loci previously associated with SCZ and/or BP were investigated. Each sample was analyzed separately then a meta-analysis was performed. SH3GL2 and CLVS1 genes were associated with suicidal ideation in SCZ (p = 5.62e-08 and 0.01, respectively), the former also in the meta-analysis (p = .01). SHC4 gene was associated with depressive symptoms severity in BP (p = .003). A gene set involved in cellular differentiation (GO:0048661) was associated with substance disorder comorbidity in the meta-analysis (p = .03). Individual loci previously associated with SCZ or BP did not modulate the phenotypes of interest. This study provided confirmatory and new findings. SH3GL2 (endophilin A1) showed a role in suicidal ideation that may be due to its relevance to the glutamate system. SHC4 regulates BDNF-induced MAPK activation and was previously associated with depression. CLVS1 is involved in lysosome maturation and was for the first time associated with a psychiatric trait. GO:0048661 may mediate the risk of substance disorder through an effect on neurodevelopment/neuroplasticity.
Collapse
Affiliation(s)
- Filippo Corponi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy
| | - Stefano Bonassi
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Pisana, Rome, Italy; Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | - Eduard Vieta
- Bipolar Disorders Unit, Institute of Neuroscience, Hospital Clínic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Diego Albani
- Laboratory of Biology of Neurodegenerative Disorders, Neuroscience Department, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Alessandra Frustaci
- Barnet, Enfield and Haringey Mental Health NHS Trust, St.Ann's Hospital, St.Ann's Road, N15 3 TH London, UK
| | | | - Stefano Landi
- Dipartimento di Biologia, Universita' di Pisa, Pisa, Italy
| | - Stefania Boccia
- Section of Hygiene, Institute of Public Health, Universita' Cattolica del Sacro Cuore, Fondazione Policlinico "Agostino Gemelli" IRCCS, Rome, Italy
| | - Alessandro Serretti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy.
| | - Chiara Fabbri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Italy
| |
Collapse
|
36
|
Synaptic Organization of VGLUT3 Expressing Low-Threshold Mechanosensitive C Fiber Terminals in the Rodent Spinal Cord. eNeuro 2019; 6:eN-NWR-0007-19. [PMID: 30783617 PMCID: PMC6378328 DOI: 10.1523/eneuro.0007-19.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 11/21/2022] Open
Abstract
Low-threshold mechanosensitive C fibers (C-LTMRs) that express the vesicular glutamate transporter VGLUT3 are thought to signal affective touch, and may also play a role in mechanical allodynia. However, the nature of the central termination of C-LTMRs in the dorsal horn remains largely unexplored. Here, we used light and electron microscopy in combination with VGLUT3 immunolabeling as a marker of C-LTMR terminations to investigate this issue. VGLUT3+ C-LTMRs formed central terminals of Type II glomeruli in the inner part of lamina II of the dorsal horn, often establishing multiple asymmetric synapses with postsynaptic dendrites but also participating in synaptic configurations with presynaptic axons and dendrites. Unexpectedly, essentially all VGLUT3+ C-LTMR terminals showed substantial VGLUT1 expression in the rat, whereas such terminals in mice lacked VGLUT1. Most VGLUT3+ C-LTMR terminals exhibited weak-to-moderate VGLUT2 expression. Further, C-LTMR terminals formed numerous synapses with excitatory protein kinase Cγ (PKCγ) interneurons and inhibitory parvalbumin neurons, whereas synapses with calretinin neurons were scarce. C-LTMR terminals rarely if ever established synapses with neurokinin 1 receptor (NK1R)-possessing dendrites traversing lamina II. Thus, VGLUT3+ C-LTMR terminals appear to largely correspond to neurofilament-lacking central terminals of Type II glomeruli in inner lamina II and can thus be identified at the ultrastructural level by morphological criteria. The participation of C-LTMR terminals in Type II glomeruli involving diverse populations of interneuron indicates highly complex modes of integration of C-LTMR mediated signaling in the dorsal horn. Furthermore, differences in VGLUT1 expression indicate distinct species differences in synaptic physiology of C-LTMR terminals.
Collapse
|
37
|
Morsy A, Trippier PC. Current and Emerging Pharmacological Targets for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2019; 72:S145-S176. [PMID: 31594236 DOI: 10.3233/jad-190744] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
No cure or disease-modifying therapy for Alzheimer's disease (AD) has yet been realized. However, a multitude of pharmacological targets have been identified for possible engagement to enable drug discovery efforts for AD. Herein, we review these targets comprised around three main therapeutic strategies. First is an approach that targets the main pathological hallmarks of AD: amyloid-β (Aβ) oligomers and hyperphosphorylated tau tangles which primarily focuses on reducing formation and aggregation, and/or inducing their clearance. Second is a strategy that modulates neurotransmitter signaling. Comprising this strategy are the cholinesterase inhibitors and N-methyl-D-aspartate receptor blockade treatments that are clinically approved for the symptomatic treatment of AD. Additional targets that aim to stabilize neuron signaling through modulation of neurotransmitters and their receptors are also discussed. Finally, the third approach comprises a collection of 'sensitive targets' that indirectly influence Aβ or tau accumulation. These targets are proteins that upon Aβ accumulation in the brain or direct Aβ-target interaction, a modification in the target's function is induced. The process occurs early in disease progression, ultimately causing neuronal dysfunction. This strategy aims to restore normal target function to alleviate Aβ-induced toxicity in neurons. Overall, we generally limit our analysis to targets that have emerged in the last decade and targets that have been validated using small molecules in in vitro and/or in vivo models. This review is not an exhaustive list of all possible targets for AD but serves to highlight the most promising and critical targets suitable for small molecule drug intervention.
Collapse
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
38
|
McGregor G, Harvey J. Regulation of Hippocampal Synaptic Function by the Metabolic Hormone, Leptin: Implications for Health and Neurodegenerative Disease. Front Cell Neurosci 2018; 12:340. [PMID: 30386207 PMCID: PMC6198461 DOI: 10.3389/fncel.2018.00340] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/13/2018] [Indexed: 11/13/2022] Open
Abstract
The role of the endocrine hormone leptin in controlling energy homeostasis in the hypothalamus are well documented. However the CNS targets for leptin are not restricted to the hypothalamus as a high density of leptin receptors are also expressed in several parts of the brain involved in higher cognitive functions including the hippocampus. Numerous studies have identified that in the hippocampus, leptin has cognitive enhancing actions as exogenous application of this hormone facilitates hippocampal-dependent learning and memory, whereas lack or insensitivity to leptin results in significant memory deficits. Leptin also markedly influences some of the main cellular changes that are involved in learning and memory including NMDA-receptor dependent synaptic plasticity and glutamate receptor trafficking. Like other metabolic hormones, there is a significant decline in neuronal sensitivity to leptin during the ageing process. Indeed, the capacity of leptin to modulate the functioning of hippocampal synapses is substantially reduced in aged compared to adult tissue. Clinical studies have also identified an association between circulating leptin levels and the risk of certain neurodegenerative disorders such as Alzheimer’s disease (AD). In view of this, targeting leptin and/or its receptor/signaling mechanisms may be an innovative approach for developing therapies to treat AD. In support of this, accumulating evidence indicates that leptin has cognitive enhancing and neuroprotective actions in various models of AD. Here we assess recent evidence that supports an important regulatory role for leptin at hippocampal CA1 synapses, and we discuss how age-related alterations in this hormonal system influences neurodegenerative disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Jenni Harvey
- Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
39
|
Overexpression of endophilin A1 exacerbates synaptic alterations in a mouse model of Alzheimer's disease. Nat Commun 2018; 9:2968. [PMID: 30061577 PMCID: PMC6065365 DOI: 10.1038/s41467-018-04389-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 04/27/2018] [Indexed: 02/05/2023] Open
Abstract
Endophilin A1 (EP) is a protein enriched in synaptic terminals that has been linked to Alzheimer's disease (AD). Previous in vitro studies have shown that EP can bind to a variety of proteins, which elicit changes in synaptic transmission of neurotransmitters and spine formation. Additionally, we previously showed that EP protein levels are elevated in AD patients and AD transgenic animal models. Here, we establish the in vivo consequences of upregulation of EP expression in amyloid-β peptide (Aβ)-rich environments, leading to changes in both long-term potentiation and learning and memory of transgenic animals. Specifically, increasing EP augmented cerebral Aβ accumulation. EP-mediated signal transduction via reactive oxygen species (ROS)/p38 mitogen-activated protein (MAP) kinase contributes to Aβ-induced mitochondrial dysfunction, synaptic injury, and cognitive decline, which could be rescued by blocking either ROS or p38 MAP kinase activity.
Collapse
|
40
|
Yang Y, Chen J, Guo Z, Deng S, Du X, Zhu S, Ye C, Shi YS, Liu JJ. Endophilin A1 Promotes Actin Polymerization in Dendritic Spines Required for Synaptic Potentiation. Front Mol Neurosci 2018; 11:177. [PMID: 29892212 PMCID: PMC5985315 DOI: 10.3389/fnmol.2018.00177] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/09/2018] [Indexed: 12/04/2022] Open
Abstract
Endophilin A1 is a member of the N-BAR domain-containing endophilin A protein family that is involved in membrane dynamics and trafficking. At the presynaptic terminal, endophilin As participate in synaptic vesicle recycling and autophagosome formation. By gene knockout studies, here we report that postsynaptic endophilin A1 functions in synaptic plasticity. Ablation of endophilin A1 in the hippocampal CA1 region of mature mouse brain impairs long-term spatial and contextual fear memory. Its loss in CA1 neurons postsynaptic of the Schaffer collateral pathway causes impairment in their AMPA-type glutamate receptor-mediated synaptic transmission and long-term potentiation. In KO neurons, defects in the structural and functional plasticity of dendritic spines can be rescued by overexpression of endophilin A1 but not A2 or A3. Further, endophilin A1 promotes actin polymerization in dendritic spines during synaptic potentiation. These findings reveal a physiological role of endophilin A1 distinct from that of other endophilin As at the postsynaptic site.
Collapse
Affiliation(s)
- Yanrui Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Zhenzhen Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Shikun Deng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Xiangyang Du
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Shaoxia Zhu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Chang Ye
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yun S Shi
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Jia-Jia Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
41
|
Richter K, Schmutz I, Darna M, Zander JF, Chavan R, Albrecht U, Ahnert-Hilger G. VGLUT1 Binding to Endophilin or Intersectin1 and Dynamin Phosphorylation in a Diurnal Context. Neuroscience 2018; 371:29-37. [DOI: 10.1016/j.neuroscience.2017.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 10/18/2022]
|
42
|
Steinkellner T, Zell V, Farino ZJ, Sonders MS, Villeneuve M, Freyberg RJ, Przedborski S, Lu W, Freyberg Z, Hnasko TS. Role for VGLUT2 in selective vulnerability of midbrain dopamine neurons. J Clin Invest 2018; 128:774-788. [PMID: 29337309 DOI: 10.1172/jci95795] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
Parkinson's disease is characterized by the loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). DA neurons in the ventral tegmental area are more resistant to this degeneration than those in the SNc, though the mechanisms for selective resistance or vulnerability remain poorly understood. A key to elucidating these processes may lie within the subset of DA neurons that corelease glutamate and express the vesicular glutamate transporter VGLUT2. Here, we addressed the potential relationship between VGLUT expression and DA neuronal vulnerability by overexpressing VGLUT in DA neurons of flies and mice. In Drosophila, VGLUT overexpression led to loss of select DA neuron populations. Similarly, expression of VGLUT2 specifically in murine SNc DA neurons led to neuronal loss and Parkinsonian behaviors. Other neuronal cell types showed no such sensitivity, suggesting that DA neurons are distinctively vulnerable to VGLUT2 expression. Additionally, most DA neurons expressed VGLUT2 during development, and coexpression of VGLUT2 with DA markers increased following injury in the adult. Finally, conditional deletion of VGLUT2 made DA neurons more susceptible to Parkinsonian neurotoxins. These data suggest that the balance of VGLUT2 expression is a crucial determinant of DA neuron survival. Ultimately, manipulation of this VGLUT2-dependent process may represent an avenue for therapeutic development.
Collapse
Affiliation(s)
| | - Vivien Zell
- Department of Neurosciences, UCSD, La Jolla, California, USA
| | - Zachary J Farino
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Michael Villeneuve
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robin J Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Serge Przedborski
- Department of Neurology, and.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Thomas S Hnasko
- Department of Neurosciences, UCSD, La Jolla, California, USA
| |
Collapse
|
43
|
McIntosh BB, Pyrpassopoulos S, Holzbaur ELF, Ostap EM. Opposing Kinesin and Myosin-I Motors Drive Membrane Deformation and Tubulation along Engineered Cytoskeletal Networks. Curr Biol 2018; 28:236-248.e5. [PMID: 29337076 DOI: 10.1016/j.cub.2017.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/29/2017] [Accepted: 12/07/2017] [Indexed: 01/22/2023]
Abstract
Microtubule and actin filament molecular motors such as kinesin-1 and myosin-Ic (Myo1c) transport and remodel membrane-bound vesicles; however, it is unclear how they coordinate to accomplish these tasks. We introduced kinesin-1- and Myo1c-bound giant unilamellar vesicles (GUVs) into a micropatterned in vitro cytoskeletal matrix modeled after the subcellular architecture where vesicular sorting and membrane remodeling are observed. This array was composed of sparse microtubules intersecting regions dense with actin filaments, and revealed that Myo1c-dependent tethering of GUVs enabled kinesin-1-driven membrane deformation and tubulation. Membrane remodeling at actin/microtubule intersections was modulated by lipid composition and the addition of the Bin-Amphiphysin-Rvs-domain (BAR-domain) proteins endophilin or FCH-domain-only (FCHo). Myo1c not only tethered microtubule-transported cargo, but also transported, deformed, and tubulated GUVs along actin filaments in a lipid-composition- and BAR-protein-responsive manner. These results suggest a mechanism for actin-based involvement in vesicular transport and remodeling of intracellular membranes, and implicate lipid composition as a key factor in determining whether vesicles will undergo transport, deformation, or tubulation driven by opposing actin and microtubule motors and BAR-domain proteins.
Collapse
Affiliation(s)
- Betsy B McIntosh
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - Serapion Pyrpassopoulos
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - Erika L F Holzbaur
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA.
| | - E Michael Ostap
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA; Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104-6085, USA.
| |
Collapse
|
44
|
Mori Y, Takamori S. Molecular Signatures Underlying Synaptic Vesicle Cargo Retrieval. Front Cell Neurosci 2018; 11:422. [PMID: 29379416 PMCID: PMC5770824 DOI: 10.3389/fncel.2017.00422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/15/2017] [Indexed: 12/31/2022] Open
Abstract
Efficient retrieval of the synaptic vesicle (SV) membrane from the presynaptic plasma membrane, a process called endocytosis, is crucial for the fidelity of neurotransmission, particularly during sustained neural activity. Although multiple modes of endocytosis have been identified, it is clear that the efficient retrieval of the major SV cargos into newly formed SVs during any of these modes is fundamental for synaptic transmission. It is currently believed that SVs are eventually reformed via a clathrin-dependent pathway. Various adaptor proteins recognize SV cargos and link them to clathrin, ensuring the efficient retrieval of the cargos into newly formed SVs. Here, we summarize our current knowledge of the molecular signatures within individual SV cargos that underlie efficient retrieval into SV membranes, as well as discuss possible contributions of the mechanisms under physiological conditions.
Collapse
Affiliation(s)
- Yasunori Mori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| |
Collapse
|
45
|
Optogenetic Tools for Subcellular Applications in Neuroscience. Neuron 2017; 96:572-603. [PMID: 29096074 DOI: 10.1016/j.neuron.2017.09.047] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/30/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022]
Abstract
The ability to study cellular physiology using photosensitive, genetically encoded molecules has profoundly transformed neuroscience. The modern optogenetic toolbox includes fluorescent sensors to visualize signaling events in living cells and optogenetic actuators enabling manipulation of numerous cellular activities. Most optogenetic tools are not targeted to specific subcellular compartments but are localized with limited discrimination throughout the cell. Therefore, optogenetic activation often does not reflect context-dependent effects of highly localized intracellular signaling events. Subcellular targeting is required to achieve more specific optogenetic readouts and photomanipulation. Here we first provide a detailed overview of the available optogenetic tools with a focus on optogenetic actuators. Second, we review established strategies for targeting these tools to specific subcellular compartments. Finally, we discuss useful tools and targeting strategies that are currently missing from the optogenetics repertoire and provide suggestions for novel subcellular optogenetic applications.
Collapse
|
46
|
Li H, Santos MS, Park CK, Dobry Y, Voglmaier SM. VGLUT2 Trafficking Is Differentially Regulated by Adaptor Proteins AP-1 and AP-3. Front Cell Neurosci 2017; 11:324. [PMID: 29123471 PMCID: PMC5662623 DOI: 10.3389/fncel.2017.00324] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/28/2017] [Indexed: 01/25/2023] Open
Abstract
Release of the major excitatory neurotransmitter glutamate by synaptic vesicle exocytosis depends on glutamate loading into synaptic vesicles by vesicular glutamate transporters (VGLUTs). The two principal isoforms, VGLUT1 and 2, exhibit a complementary pattern of expression in adult brain that broadly distinguishes cortical (VGLUT1) and subcortical (VGLUT2) systems, and correlates with distinct physiological properties in synapses expressing these isoforms. Differential trafficking of VGLUT1 and 2 has been suggested to underlie their functional diversity. Increasing evidence suggests individual synaptic vesicle proteins use specific sorting signals to engage specialized biochemical mechanisms to regulate their recycling. We observed that VGLUT2 recycles differently in response to high frequency stimulation than VGLUT1. Here we further explore the trafficking of VGLUT2 using a pHluorin-based reporter, VGLUT2-pH. VGLUT2-pH exhibits slower rates of both exocytosis and endocytosis than VGLUT1-pH. VGLUT2-pH recycling is slower than VGLUT1-pH in both hippocampal neurons, which endogenously express mostly VGLUT1, and thalamic neurons, which endogenously express mostly VGLUT2, indicating that protein identity, not synaptic vesicle membrane or neuronal cell type, controls sorting. We characterize sorting signals in the C-terminal dileucine-like motif, which plays a crucial role in VGLUT2 trafficking. Disruption of this motif abolishes synaptic targeting of VGLUT2 and essentially eliminates endocytosis of the transporter. Mutational and biochemical analysis demonstrates that clathrin adaptor proteins (APs) interact with VGLUT2 at the dileucine-like motif. VGLUT2 interacts with AP-2, a well-studied adaptor protein for clathrin mediated endocytosis. In addition, VGLUT2 also interacts with the alternate adaptors, AP-1 and AP-3. VGLUT2 relies on distinct recycling mechanisms from VGLUT1. Abrogation of these differences by pharmacological and molecular inhibition reveals that these mechanisms are dependent on the adaptor proteins AP-1 and AP-3. Further, shRNA-mediated knockdown reveals differential roles for AP-1 and AP-3 in VGLUT2 recycling.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Magda S Santos
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Chihyung K Park
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Yuriy Dobry
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Susan M Voglmaier
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
47
|
Lencer R, Mills LJ, Alliey-Rodriguez N, Shafee R, Lee AM, Reilly JL, Sprenger A, McDowell JE, McCarroll SA, Keshavan MS, Pearlson GD, Tamminga CA, Clementz BA, Gershon ES, Sweeney JA, Bishop JR. Genome-wide association studies of smooth pursuit and antisaccade eye movements in psychotic disorders: findings from the B-SNIP study. Transl Psychiatry 2017; 7:e1249. [PMID: 29064472 PMCID: PMC5682604 DOI: 10.1038/tp.2017.210] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/14/2017] [Indexed: 02/07/2023] Open
Abstract
Eye movement deviations, particularly deficits of initial sensorimotor processing and sustained pursuit maintenance, and antisaccade inhibition errors, are established intermediate phenotypes for psychotic disorders. We here studied eye movement measures of 849 participants from the Bipolar-Schizophrenia Network on Intermediate Phenotypes (B-SNIP) study (schizophrenia N=230, schizoaffective disorder N=155, psychotic bipolar disorder N=206 and healthy controls N=258) as quantitative phenotypes in relation to genetic data, while controlling for genetically derived ancestry measures, age and sex. A mixed-modeling genome-wide association studies approach was used including ~4.4 million genotypes (PsychChip and 1000 Genomes imputation). Across participants, sensorimotor processing at pursuit initiation was significantly associated with a single nucleotide polymorphism in IPO8 (12p11.21, P=8 × 10-11), whereas suggestive associations with sustained pursuit maintenance were identified with SNPs in SH3GL2 (9p22.2, P=3 × 10-8). In participants of predominantly African ancestry, sensorimotor processing was also significantly associated with SNPs in PCDH12 (5q31.3, P=1.6 × 10-10), and suggestive associations were observed with NRSN1 (6p22.3, P=5.4 × 10-8) and LMO7 (13q22.2, P=7.3x10-8), whereas antisaccade error rate was significantly associated with a non-coding region at chromosome 7 (P=6.5 × 10-9). Exploratory pathway analyses revealed associations with nervous system development and function for 40 top genes with sensorimotor processing and pursuit maintenance (P=4.9 × 10-2-9.8 × 10-4). Our findings suggest novel patterns of genetic variation relevant for brain systems subserving eye movement control known to be impaired in psychotic disorders. They include genes involved in nuclear trafficking and gene silencing (IPO8), fast axonal guidance and synaptic specificity (PCDH12), transduction of nerve signals (NRSN1), retinal degeneration (LMO7), synaptic glutamate release (SH3GL2), and broader nervous system development and function.
Collapse
Affiliation(s)
- R Lencer
- Department of Psychiatry and Psychotherapy, Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Muenster, Muenster, Germany
| | - L J Mills
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - N Alliey-Rodriguez
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - R Shafee
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - A M Lee
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - J L Reilly
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - A Sprenger
- Department of Neurology, University of Luebeck, Luebeck, Germany
| | - J E McDowell
- Department of Psychology and Neuroscience, BioImaging Research Center, University of Georgia, Athens, GA, USA
| | - S A McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - M S Keshavan
- Department of Psychiatry, Harvard Medical School, Beth Israel Deacones Medical Center, Boston, MA, USA
| | - G D Pearlson
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, CT, USA
- Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - C A Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - B A Clementz
- Department of Psychology and Neuroscience, BioImaging Research Center, University of Georgia, Athens, GA, USA
| | - E S Gershon
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - J A Sweeney
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - J R Bishop
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
- Department of Psychiatry, University of Minnesota College of Medicine, Minneapolis, MN, USA
| |
Collapse
|
48
|
Guillaud L, Dimitrov D, Takahashi T. Presynaptic morphology and vesicular composition determine vesicle dynamics in mouse central synapses. eLife 2017; 6. [PMID: 28432787 PMCID: PMC5423771 DOI: 10.7554/elife.24845] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 04/18/2017] [Indexed: 11/13/2022] Open
Abstract
Transport of synaptic vesicles (SVs) in nerve terminals is thought to play essential roles in maintenance of neurotransmission. To identify factors modulating SV movements, we performed real-time imaging analysis of fluorescently labeled SVs in giant calyceal and conventional hippocampal terminals. Compared with small hippocampal terminals, SV movements in giant calyceal terminals were faster, longer and kinetically more heterogeneous. Morphological maturation of giant calyceal terminals was associated with an overall reduction in SV mobility and displacement heterogeneity. At the molecular level, SVs over-expressing vesicular glutamate transporter 1 (VGLUT1) showed higher mobility than VGLUT2-expressing SVs. Pharmacological disruption of the presynaptic microtubule network preferentially reduced long directional movements of SVs between release sites. Functionally, synaptic stimulation appeared to recruit SVs to active zones without significantly altering their mobility. Hence, the morphological features of nerve terminals and the molecular signature of vesicles are key elements determining vesicular dynamics and movements in central synapses.
Collapse
Affiliation(s)
- Laurent Guillaud
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | - Dimitar Dimitrov
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | - Tomoyuki Takahashi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| |
Collapse
|
49
|
Characterization of a Human Point Mutation of VGLUT3 (p.A211V) in the Rodent Brain Suggests a Nonuniform Distribution of the Transporter in Synaptic Vesicles. J Neurosci 2017; 37:4181-4199. [PMID: 28314816 DOI: 10.1523/jneurosci.0282-16.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 11/21/2022] Open
Abstract
The atypical vesicular glutamate transporter type 3 (VGLUT3) is expressed by subpopulations of neurons using acetylcholine, GABA, or serotonin as neurotransmitters. In addition, VGLUT3 is expressed in the inner hair cells of the auditory system. A mutation (p.A211V) in the gene that encodes VGLUT3 is responsible for progressive deafness in two unrelated families. In this study, we investigated the consequences of the p.A211V mutation in cell cultures and in the CNS of a mutant mouse. The mutation substantially decreased VGLUT3 expression (-70%). We measured VGLUT3-p.A211V activity by vesicular uptake in BON cells, electrophysiological recording of isolated neurons, and its ability to stimulate serotonergic accumulation in cortical synaptic vesicles. Despite a marked loss of expression, the activity of the mutated isoform was only minimally altered. Furthermore, mutant mice displayed none of the behavioral alterations that have previously been reported in VGLUT3 knock-out mice. Finally, we used stimulated emission depletion microscopy to analyze how the mutation altered VGLUT3 distribution within the terminals of mice expressing the mutated isoform. The mutation appeared to reduce the expression of the VGLUT3 transporter by simultaneously decreasing the number of VGLUT3-positive synaptic vesicles and the amount of VGLUT3 per synapses. These observations suggested that VGLUT3 global activity is not linearly correlated with VGLUT3 expression. Furthermore, our data unraveled a nonuniform distribution of VGLUT3 in synaptic vesicles. Identifying the mechanisms responsible for this complex vesicular sorting will be critical to understand VGLUT's involvement in normal and pathological conditions.SIGNIFICANCE STATEMENT VGLUT3 is an atypical member of the vesicular glutamate transporter family. A point mutation of VGLUT3 (VGLUT3-p.A211V) responsible for a progressive loss of hearing has been identified in humans. We observed that this mutation dramatically reduces VGLUT3 expression in terminals (∼70%) without altering its function. Furthermore, using stimulated emission depletion microscopy, we found that reducing the expression levels of VGLUT3 diminished the number of VGLUT3-positive vesicles at synapses. These unexpected findings challenge the vision of a uniform distribution of synaptic vesicles at synapses. Therefore, the overall activity of VGLUT3 is not proportional to the level of VGLUT3 expression. These data will be key in interpreting the role of VGLUTs in human pathologies.
Collapse
|
50
|
Effective Mechanism for Synthesis of Neurotransmitter Glutamate and its Loading into Synaptic Vesicles. Neurochem Res 2016; 42:64-76. [DOI: 10.1007/s11064-016-2037-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 08/14/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023]
|