1
|
Talbi R, Stincic TL, Ferrari K, Hae CJ, Walec K, Medve E, Gerutshang A, León S, McCarthy EA, Rønnekleiv OK, Kelly MJ, Navarro VM. POMC neurons control fertility through differential signaling of MC4R in Kisspeptin neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.18.580873. [PMID: 38915534 PMCID: PMC11195098 DOI: 10.1101/2024.02.18.580873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Inactivating mutations in the melanocortin 4 receptor (MC4R) gene cause monogenic obesity. Interestingly, female patients also display various degrees of reproductive disorders, in line with the subfertile phenotype of MC4RKO female mice. However, the cellular mechanisms by which MC4R regulates reproduction are unknown. Kiss1 neurons directly stimulate gonadotropin-releasing hormone (GnRH) release through two distinct populations; the Kiss1ARH neurons, controlling GnRH pulses, and the sexually dimorphic Kiss1AVPV/PeN neurons controlling the preovulatory LH surge. Here, we show that Mc4r expressed in Kiss1 neurons regulates fertility in females. In vivo, deletion of Mc4r from Kiss1 neurons in female mice replicates the reproductive impairments of MC4RKO mice without inducing obesity. Conversely, reinsertion of Mc4r in Kiss1 neurons of MC4R null mice restores estrous cyclicity and LH pulsatility without reducing their obese phenotype. In vitro, we dissect the specific action of MC4R on Kiss1ARH vs Kiss1AVPV/PeN neurons and show that MC4R activation excites Kiss1ARH neurons through direct synaptic actions. In contrast, Kiss1AVPV/PeN neurons are normally inhibited by MC4R activation except under elevated estradiol levels, thus facilitating the activation of Kiss1AVPV/PeN neurons to induce the LH surge driving ovulation in females. Our findings demonstrate that POMCARH neurons acting through MC4R, directly regulate reproductive function in females by stimulating the "pulse generator" activity of Kiss1ARH neurons and restricting the activation of Kiss1AVPV/PeN neurons to the time of the estradiol-dependent LH surge, and thus unveil a novel pathway of the metabolic regulation of fertility by the melanocortin system.
Collapse
Affiliation(s)
- Rajae Talbi
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Todd L. Stincic
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Kaitlin Ferrari
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Choi Ji Hae
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Karol Walec
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth Medve
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Achi Gerutshang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Silvia León
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Elizabeth A. McCarthy
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Oline K. Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Martin J. Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Víctor M. Navarro
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Program in Neuroscience, Boston, MA, USA
| |
Collapse
|
2
|
Aleksandrova EP, Ivlev AP, Kulikov AA, Naumova AA, Glazova MV, Chernigovskaya EV. Aging of Krushinsky-Molodkina audiogenic rats is accompanied with pronounced neurodegeneration and dysfunction of the glutamatergic system in the hippocampus. Brain Res 2024; 1846:149294. [PMID: 39461667 DOI: 10.1016/j.brainres.2024.149294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Advancing age strongly correlates with an increased risk of epilepsy development. On the other hand, epilepsy may exacerbate the negative effects of aging making it pathological. In turn, the possible link between aging and epileptogenesis is dysregulation of glutamatergic transmission. In the present study, we analyzed the functional state of the glutamatergic system in the hippocampus of aging (18-month-old) Krushinsky-Molodkina (KM) audiogenic rats to disclose alterations associated with aging on the background of inherited predisposition to audiogenic seizures (AGS). Naïve KM rats with no AGS experience were recruited in the experiments. Wistar rats of the corresponding age were used as a control. First of all, aging KM rats demonstrated a significant decrease in cell population and synaptopodin expression in the hippocampus indicating enhanced loss of cells and synapses. Meanwhile, elevated phosphorylation of ERK1/2 and CREB and increased glutamate in the neuronal perikarya were revealed indicating increased activity of the rest hippocampal cells and increased glutamate production. However, glutamate in the fibers and synapses was mainly unchanged, and the proteins regulating glutamate exocytosis showed variable changes which could compensate each other and maintain glutamate release at the unchanged level. In addition, we revealed downregulation of NMDA-receptor subunit GluN2B and upregulation of AMPA-receptor GluA2 subunit, which could also prevent overexcitation and support cell survival in the hippocampus of aging KM rats. Nevertheless, abnormally high glutamate production, observed in aging KM rats, may provide the basis for hyperexcitability of the hippocampus and increased seizure susceptibility in old age.
Collapse
Affiliation(s)
- Ekaterina P Aleksandrova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Andrey P Ivlev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Alexey A Kulikov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Alexandra A Naumova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Margarita V Glazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | - Elena V Chernigovskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, the Russian Academy of Sciences, St. Petersburg, Russian Federation.
| |
Collapse
|
3
|
Decet M, Scott P, Kuenen S, Meftah D, Swerts J, Calatayud C, Gallego SF, Kaempf N, Nachman E, Praschberger R, Schoovaerts N, Tang CC, Eidelberg D, Al Adawi S, Al Asmi A, Nandhagopal R, Verstreken P. A candidate loss-of-function variant in SGIP1 causes synaptic dysfunction and recessive parkinsonism. Cell Rep Med 2024; 5:101749. [PMID: 39332416 PMCID: PMC11513836 DOI: 10.1016/j.xcrm.2024.101749] [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: 03/14/2024] [Revised: 06/14/2024] [Accepted: 08/31/2024] [Indexed: 09/29/2024]
Abstract
Synaptic dysfunction is recognized as an early step in the pathophysiology of parkinsonism. Several genetic mutations affecting the integrity of synaptic proteins cause or increase the risk of developing disease. We have identified a candidate causative mutation in synaptic "SH3GL2 Interacting Protein 1" (SGIP1), linked to early-onset parkinsonism in a consanguineous Arab family. Additionally, affected siblings display intellectual, cognitive, and behavioral dysfunction. Metabolic network analysis of [18F]-fluorodeoxyglucose positron emission tomography scans shows patterns very similar to those of idiopathic Parkinson's disease. We show that the identified SGIP1 mutation causes a loss of protein function, and analyses in newly created Drosophila models reveal movement defects, synaptic transmission dysfunction, and neurodegeneration, including dopaminergic synapse loss. Histology and correlative light and electron microscopy reveal the absence of synaptic multivesicular bodies and the accumulation of degradative organelles. This research delineates a putative form of recessive parkinsonism, converging on defective synaptic proteostasis and opening avenues for diagnosis, genetic counseling, and treatment.
Collapse
Affiliation(s)
- Marianna Decet
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Patrick Scott
- Laboratory of Molecular Biology, Sainte-Justine University Hospital Center, Montréal QC H3T 1C5, Canada
| | - Sabine Kuenen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Douja Meftah
- Laboratory of Pulmonary Physiology, Department of Pediatrics, Sainte-Justine University Hospital Center, Montréal QC H3T 1C5, Canada
| | - Jef Swerts
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Carles Calatayud
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Sandra F Gallego
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Natalie Kaempf
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Eliana Nachman
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Roman Praschberger
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Nils Schoovaerts
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Samir Al Adawi
- Department of Behavioral Medicine, College of Medicine & Health Sciences, Sultan Qaboos University, Al Khod 123, Muscat, Oman
| | - Abdullah Al Asmi
- Neurology Unit, Department of Medicine, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod 123, Muscat, Oman
| | - Ramachandiran Nandhagopal
- Neurology Unit, Department of Medicine, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod 123, Muscat, Oman.
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| |
Collapse
|
4
|
Boo KJ, Kim DH, Cho E, Kim DH, Jeon SJ, Shin CY. Neonatal dysregulation of 2-arachidonoylglycerol induces impaired brain function in adult mice. Neuropharmacology 2024; 257:110045. [PMID: 38885736 DOI: 10.1016/j.neuropharm.2024.110045] [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: 03/09/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
The endocannabinoid system (ECS) regulates neurotransmission linked to synaptic plasticity, cognition, and emotion. While it has been demonstrated that dysregulation of the ECS in adulthood is relevant not only to central nervous system (CNS) disorders such as autism spectrum disorder, cognitive dysfunction, and depression but also to brain function, there are few studies on how dysregulation of the ECS in the neonatal period affects the manifestation and pathophysiology of CNS disorders later in life. In this study, DO34, a diacylglycerol lipase alpha (DAGLα) inhibitor affecting endocannabinoid 2-AG production, was injected into C57BL/6N male mice from postnatal day (PND) 7 to PND 10, inducing dysregulation of the ECS in the neonatal period. Subsequently, we examined whether it affects neuronal function in adulthood through electrophysiological and behavioral evaluation. DO34-injected mice showed significantly decreased cognitive functions, attributed to impairment of hippocampal synaptic plasticity. The findings suggest that regulation of ECS activity in the neonatal period may induce enduring effects on adult brain function.
Collapse
Affiliation(s)
- Kyung-Jun Boo
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Dae Hyun Kim
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Eunbi Cho
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Dong Hyun Kim
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Republic of Korea.
| | - Se Jin Jeon
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon, Gangwon, 24252, Republic of Korea.
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea; Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
5
|
Rafael A, Tizzoni M, Justet C, Cairus A, Abudara V, Vitureira N. Glial Cx43 hemichannels and neuronal Panx1 hemichannels and P2X7 receptors orchestrate presynaptic homeostatic plasticity. Cell Signal 2024; 117:111113. [PMID: 38395185 DOI: 10.1016/j.cellsig.2024.111113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
The emerging role of glial cells in modulating neuronal excitability and synaptic strength is a growing field in neuroscience. In recent years, a pivotal role of gliotransmission in homeostatic presynaptic plasticity has been highlighted and glial-derived ATP arises as a key contributor. However, very little is known about the glial non-vesicular ATP-release pathway and how ATP participates in the modulation of synaptic strength. Here, we investigated the functional changes occurring in neurons upon chronic inactivity and the role of the purinergic signaling, connexin43 and pannexin1 hemichannels in this process. By using hippocampal dissociated cultures, we showed that blocking connexin43 and pannexin1 hemichannels decreases the amount of extracellular ATP. Moreover, Ca2+ imaging assays using Fluo-4/AM revealed that blocking connexin43, neuronal P2X7Rs and pannexin1 hemichannels decreases the amount of basal Ca2+ in neurons. A significant impairment in synaptic vesicle pool size was also evidenced under these conditions. Interestingly, rescue experiments where Panx1HCs are blocked showed that the compensatory adjustment of cytosolic Ca2+ was recovered after P2X7Rs activation, suggesting that Panx1 acts downstream P2X7Rs. These changes were accompanied by a modulation of neuronal permeability, as revealed by ethidium bromide uptake experiments. In particular, the permeability of neuronal P2X7Rs and pannexin1 hemichannels is increased upon 24 h of inactivity. Taken together, we have uncovered a role for connexin43-dependent ATP release and neuronal P2X7Rs and pannexin1 hemichannels in the adjustment of presynaptic strength by modulating neuronal permeability, the entrance of Ca2+ into neurons and the size of the recycling pool of synaptic vesicles.
Collapse
Affiliation(s)
- Alberto Rafael
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Avenida Gral. Flores 2125, CP 11800, Montevideo, Uruguay
| | - Marina Tizzoni
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Avenida Gral. Flores 2125, CP 11800, Montevideo, Uruguay
| | - Cristian Justet
- Departamento de Bioquímica, Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Avenida Gral. Flores 2125, CP 11800, Montevideo, Uruguay
| | - Andrea Cairus
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Avenida Gral. Flores 2125, CP 11800, Montevideo, Uruguay
| | - Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Avenida Gral. Flores 2125, CP 11800, Montevideo, Uruguay
| | - Nathalia Vitureira
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Avenida Gral. Flores 2125, CP 11800, Montevideo, Uruguay.
| |
Collapse
|
6
|
Pinky PD, Bloemer J, Smith WD, Du Y, Heslin RT, Setti SE, Pfitzer JC, Chowdhury K, Hong H, Bhattacharya S, Dhanasekaran M, Dityatev A, Reed MN, Suppiramaniam V. Prenatal Cannabinoid Exposure Elicits Memory Deficits Associated with Reduced PSA-NCAM Expression, Altered Glutamatergic Signaling, and Adaptations in Hippocampal Synaptic Plasticity. Cells 2023; 12:2525. [PMID: 37947603 PMCID: PMC10648717 DOI: 10.3390/cells12212525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 11/12/2023] Open
Abstract
Cannabis is now one of the most commonly used illicit substances among pregnant women. This is particularly concerning since developmental exposure to cannabinoids can elicit enduring neurofunctional and cognitive alterations. This study investigates the mechanisms of learning and memory deficits resulting from prenatal cannabinoid exposure (PCE) in adolescent offspring. The synthetic cannabinoid agonist WIN55,212-2 was administered to pregnant rats, and a series of behavioral, electrophysiological, and immunochemical studies were performed to identify potential mechanisms of memory deficits in the adolescent offspring. Hippocampal-dependent memory deficits in adolescent PCE animals were associated with decreased long-term potentiation (LTP) and enhanced long-term depression (LTD) at hippocampal Schaffer collateral-CA1 synapses, as well as an imbalance between GluN2A- and GluN2B-mediated signaling. Moreover, PCE reduced gene and protein expression of neural cell adhesion molecule (NCAM) and polysialylated-NCAM (PSA-NCAM), which are critical for GluN2A and GluN2B signaling balance. Administration of exogenous PSA abrogated the LTP deficits observed in PCE animals, suggesting PSA mediated alterations in GluN2A- and GluN2B- signaling pathways may be responsible for the impaired hippocampal synaptic plasticity resulting from PCE. These findings enhance our current understanding of how PCE affects memory and how this process can be manipulated for future therapeutic purposes.
Collapse
Affiliation(s)
- Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Department of Pharmaceutical and Biomedical Sciences, Touro College of Pharmacy, New York, NY 10036, USA
| | - Warren D. Smith
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Yifeng Du
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Ryan T. Heslin
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Sharay E. Setti
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Jeremiah C. Pfitzer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Kawsar Chowdhury
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Hao Hong
- Key Laboratory of Neuropsychiatric Diseases, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
- Keck Graduate Institute, School of Pharmacy and Health Sciences, Claremont Colleges, Claremont, CA 91711, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 37075 Magdeburg, Germany
| | - Alexander Dityatev
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 37075 Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, 39106 Magdeburg, Germany
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
7
|
Cheung G, Chever O, Rollenhagen A, Quenech'du N, Ezan P, Lübke JHR, Rouach N. Astroglial Connexin 43 Regulates Synaptic Vesicle Release at Hippocampal Synapses. Cells 2023; 12:cells12081133. [PMID: 37190042 DOI: 10.3390/cells12081133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 05/17/2023] Open
Abstract
Connexin 43, an astroglial gap junction protein, is enriched in perisynaptic astroglial processes and plays major roles in synaptic transmission. We have previously found that astroglial Cx43 controls synaptic glutamate levels and allows for activity-dependent glutamine release to sustain physiological synaptic transmissions and cognitiogns. However, whether Cx43 is important for the release of synaptic vesicles, which is a critical component of synaptic efficacy, remains unanswered. Here, using transgenic mice with a glial conditional knockout of Cx43 (Cx43-/-), we investigate whether and how astrocytes regulate the release of synaptic vesicles from hippocampal synapses. We report that CA1 pyramidal neurons and their synapses develop normally in the absence of astroglial Cx43. However, a significant impairment in synaptic vesicle distribution and release dynamics were observed. In particular, the FM1-43 assays performed using two-photon live imaging and combined with multi-electrode array stimulation in acute hippocampal slices, revealed a slower rate of synaptic vesicle release in Cx43-/- mice. Furthermore, paired-pulse recordings showed that synaptic vesicle release probability was also reduced and is dependent on glutamine supply via Cx43 hemichannel (HC). Taken together, we have uncovered a role for Cx43 in regulating presynaptic functions by controlling the rate and probability of synaptic vesicle release. Our findings further highlight the significance of astroglial Cx43 in synaptic transmission and efficacy.
Collapse
Affiliation(s)
- Giselle Cheung
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, 75231 Paris, France
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, 75231 Paris, France
| | - Astrid Rollenhagen
- Institute for Neuroscience and Medicine INM-10, Research Center Jülich, 52428 Jülich, Germany
- Jülich-Aachen Research Alliance Translational Brain Medicine, 52056 Aachen, Germany
| | - Nicole Quenech'du
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, 75231 Paris, France
| | - Pascal Ezan
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, 75231 Paris, France
| | - Joachim H R Lübke
- Institute for Neuroscience and Medicine INM-10, Research Center Jülich, 52428 Jülich, Germany
- Jülich-Aachen Research Alliance Translational Brain Medicine, 52056 Aachen, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Rheinisch-Westfaelische Technische Hochschule Aachen University, 52056 Aachen, Germany
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, 75231 Paris, France
| |
Collapse
|
8
|
Belloch FDB, Cortés-Erice M, Herzog E, Zhang XM, Díaz-Perdigon T, Puerta E, Tordera RM. Fast antidepressant action of ketamine in mouse models requires normal VGLUT1 levels from prefrontal cortex neurons. Prog Neuropsychopharmacol Biol Psychiatry 2023; 121:110640. [PMID: 36209771 DOI: 10.1016/j.pnpbp.2022.110640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 09/03/2022] [Accepted: 09/12/2022] [Indexed: 11/07/2022]
Abstract
The NMDA antagonist ketamine demonstrated a fast antidepressant activity in treatment-resistant depression. Pre-clinical studies suggest that de novo synthesis of the brain-derived neurotrophic factor (BDNF) in the PFC might be involved in the rapid antidepressant action of ketamine. Applying a genetic model of impaired glutamate release, this study aims to further identify the molecular mechanisms that could modulate antidepressant action and resistance to treatment. To that end, mice knocked-down for the vesicular glutamate transporter 1 (VGLUT1+/-) were used. We analyzed anhedonia and helpless behavior as well as the expression of the proteins linked to glutamate transmission in the PFC of mice treated with ketamine or the reference antidepressant reboxetine. Moreover, we analyzed the acute effects of ketamine in VGLUT1+/- mice pretreated with chronic reboxetine or those that received a PFC rescue expression of VGLUT1. Chronic reboxetine rescued the depressive-like phenotype of the VGLUT1+/- mice. In addition, it enhanced the expression of the proteins linked to the AMPA signaling pathway as well as the immature form of BDNF (pro-BDNF). Unlike WT mice, ketamine had no effect on anhedonia or pro-BDNF expression in VGLUT1+/- mice; it also failed to decrease phosphorylated eukaryote elongation factor 2 (p-eEF2). Nevertheless, we found that reboxetine administered as pretreatment or PFC overexpression of VGLUT1 did rescue the antidepressant-like activity of acute ketamine in the mice. Our results strongly suggest that not only do PFC VGLUT1 levels modulate the rapid-antidepressant action of ketamine, but also highlight a possible mechanism for antidepressant resistance in some patients.
Collapse
Affiliation(s)
| | - María Cortés-Erice
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Etienne Herzog
- Université de Bordeaux, Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Xiao Min Zhang
- Université de Bordeaux, Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Teresa Díaz-Perdigon
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Elena Puerta
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Rosa M Tordera
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain.
| |
Collapse
|
9
|
Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Mares-Barbosa TB, Patrón-Soberano A, Howe AG, Portales-Pérez DP, Miquelajáuregui Graf A, Estrada-Sánchez AM. Revealing the contribution of astrocytes to glutamatergic neuronal transmission. Front Cell Neurosci 2023; 16:1037641. [PMID: 36744061 PMCID: PMC9893894 DOI: 10.3389/fncel.2022.1037641] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Research on glutamatergic neurotransmission has focused mainly on the function of presynaptic and postsynaptic neurons, leaving astrocytes with a secondary role only to ensure successful neurotransmission. However, recent evidence indicates that astrocytes contribute actively and even regulate neuronal transmission at different levels. This review establishes a framework by comparing glutamatergic components between neurons and astrocytes to examine how astrocytes modulate or otherwise influence neuronal transmission. We have included the most recent findings about the role of astrocytes in neurotransmission, allowing us to understand the complex network of neuron-astrocyte interactions. However, despite the knowledge of synaptic modulation by astrocytes, their contribution to specific physiological and pathological conditions remains to be elucidated. A full understanding of the astrocyte's role in neuronal processing could open fruitful new frontiers in the development of therapeutic applications.
Collapse
Affiliation(s)
- Ares Orlando Cuellar-Santoyo
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Victor Manuel Ruiz-Rodríguez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Teresa Belem Mares-Barbosa
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | - Araceli Patrón-Soberano
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Andrew G. Howe
- Intelligent Systems Laboratory, HRL Laboratories, LLC, Malibu, CA, United States
| | - Diana Patricia Portales-Pérez
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | | | - Ana María Estrada-Sánchez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| |
Collapse
|
10
|
Taipala E, Pfitzer JC, Hellums M, Reed MN, Gramlich MW. rTg(TauP301L)4510 mice exhibit increased VGlut1 in hippocampal presynaptic glutamatergic vesicles and increased extracellular glutamate release. Front Synaptic Neurosci 2022; 14:925546. [PMID: 35989711 PMCID: PMC9383415 DOI: 10.3389/fnsyn.2022.925546] [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: 04/21/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
The molecular pathways that contribute to the onset of symptoms in tauopathy models, including Alzheimer’s disease (AD), are difficult to distinguish because multiple changes can happen simultaneously at different stages of disease progression. Understanding early synaptic alterations and their supporting molecular pathways is essential to develop better pharmacological targets to treat AD. Here, we focus on an early onset rTg(TauP301L)4510 tauopathy mouse model that exhibits hyperexcitability in hippocampal neurons of adult mice that is correlated with presynaptic changes and increased extracellular glutamate levels. However, it is not clear if increased extracellular glutamate is caused by presynaptic changes alone, or if presynaptic changes are a contributing factor among other factors. To determine whether pathogenic tau alters presynaptic function and glutamate release, we studied cultured hippocampal neurons at 14–18 days in vitro (DIV) from animals of both sexes to measure presynaptic changes in tauP301L positive mice. We observed that presynaptic vesicles exhibit increased vesicular glutamate transporter 1 (VGlut1) using immunohistochemistry of fixed cells and an established pH-sensitive green fluorescent protein approach. We show that tauP301L positive neurons exhibit a 40% increase in VGlut1 per vesicle compared to tauP301L negative littermates. Further, we use the extracellular glutamate reporter iGluSnFR to show that increased VGlut1 per vesicle directly translates into a 40% increase in extracellular glutamate. Together, these results show that increased extracellular glutamate levels observed in tauP301L mice are not caused by increased vesicle exocytosis probability but rather are directly related to increased VGlut1 transporters per synaptic vesicle.
Collapse
Affiliation(s)
- Erika Taipala
- Department of Physics, Auburn University, Auburn, AL, United States
| | | | - Morgan Hellums
- Department of Physics, Auburn University, Auburn, AL, United States
| | - Miranda N. Reed
- Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Michael W. Gramlich
- Department of Physics, Auburn University, Auburn, AL, United States
- *Correspondence: Michael W. Gramlich,
| |
Collapse
|
11
|
Stincic TL, Kelly MJ. Estrogenic regulation of reproduction and energy homeostasis by a triumvirate of hypothalamic arcuate neurons. J Neuroendocrinol 2022; 34:e13145. [PMID: 35581942 DOI: 10.1111/jne.13145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17β-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
12
|
Vardar G, Salazar-Lázaro A, Zobel S, Trimbuch T, Rosenmund C. Syntaxin-1A modulates vesicle fusion in mammalian neurons via juxtamembrane domain dependent palmitoylation of its transmembrane domain. eLife 2022; 11:78182. [PMID: 35638903 PMCID: PMC9183232 DOI: 10.7554/elife.78182] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
SNAREs are undoubtedly one of the core elements of synaptic transmission. Contrary to the well characterized function of their SNARE domains bringing the plasma and vesicular membranes together, the level of contribution of their juxtamembrane domain (JMD) and the transmembrane domain (TMD) to the vesicle fusion is still under debate. To elucidate this issue, we analyzed three groups of STX1A mutations in cultured mouse hippocampal neurons: (1) elongation of STX1A’s JMD by three amino acid insertions in the junction of SNARE-JMD or JMD-TMD; (2) charge reversal mutations in STX1A’s JMD; and (3) palmitoylation deficiency mutations in STX1A’s TMD. We found that both JMD elongations and charge reversal mutations have position-dependent differential effects on Ca2+-evoked and spontaneous neurotransmitter release. Importantly, we show that STX1A’s JMD regulates the palmitoylation of STX1A’s TMD and loss of STX1A palmitoylation either through charge reversal mutation K260E or by loss of TMD cysteines inhibits spontaneous vesicle fusion. Interestingly, the retinal ribbon specific STX3B has a glutamate in the position corresponding to the K260E mutation in STX1A and mutating it with E259K acts as a molecular on-switch. Furthermore, palmitoylation of post-synaptic STX3A can be induced by the exchange of its JMD with STX1A’s JMD together with the incorporation of two cysteines into its TMD. Forced palmitoylation of STX3A dramatically enhances spontaneous vesicle fusion suggesting that STX1A regulates spontaneous release through two distinct mechanisms: one through the C-terminal half of its SNARE domain and the other through the palmitoylation of its TMD.
Collapse
Affiliation(s)
- Gülçin Vardar
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Salazar-Lázaro
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sina Zobel
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Thorsten Trimbuch
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Rosenmund
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
13
|
He L, Xu W, Zhang C, Ding Z, Guo Q, Zou W, Wang J. Dysregulation of Vesicular Glutamate Transporter VGluT2 via BDNF/TrkB Pathway Contributes to Morphine Tolerance in Mice. Front Pharmacol 2022; 13:861786. [PMID: 35559256 PMCID: PMC9086316 DOI: 10.3389/fphar.2022.861786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
Morphine is widely used in the treatment of moderate to severe pain. Long-term use of morphine leads to various adverse effects, such as tolerance and hyperalgesia. Vesicular glutamate transporter 2 (VGluT2) accumulates glutamate into synaptic vesicles and plays multiple roles in the central nervous system. However, the specific role of VGluT2 in morphine tolerance has not been fully elucidated. Here, we investigated the regulatory role of VGluT2 in morphine tolerance and assessed the potential role of the brain-derived neurotrophic factor (BDNF)/tyrosine kinase B (TrkB) pathway in VGluT2 mediated morphine antinociceptive tolerance in mice. In the present study, we found that VGluT2 is upregulated in the spinal cord after the development of morphine tolerance. Furthermore, inhibition of VGluT2 with its antagonist (Chicago sky blue 6 B, CSB6B) or knockdown of VGluT2 by lentivirus restored the analgesic effect of morphine, suppressed the activation of astrocytes and microglia, and decreased glial-derived pro-inflammatory cytokines. Overexpression of VGluT2 by lentivirus facilitated morphine tolerance and mechanical hyperalgesia. In addition, we found the expression of BDNF is correlated with VGluT2 expression in the spinal cord after chronic morphine administration. Intrathecal injection of the BDNF/TrkB pathway antagonist K252a attenuated the development of morphine tolerance and decreased the expression of VGluT2 in the spinal cord, which suggested the BDNF/TrkB pathway participates in the regulation of VGluT2 in morphine tolerance. This study elucidates the functional capability of VGluT2 in modulating morphine tolerance and identifies a novel mechanism and promising therapeutic target for morphine tolerance.
Collapse
Affiliation(s)
- Liqiong He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xu
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuofeng Ding
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Kosmowska B, Ossowska K, Wardas J. Blockade of adenosine A 2A receptors inhibits Tremulous Jaw Movements as well as expression of zif-268 and GAD65 mRNAs in brain motor structures. Behav Brain Res 2022; 417:113585. [PMID: 34536428 DOI: 10.1016/j.bbr.2021.113585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 11/26/2022]
Abstract
Tremor is one of the motor symptoms of Parkinson's disease (PD), present also in neuroleptic-induced parkinsonism. Tremulous Jaw Movements (TJMs) are suggested to be a well-validated rodent model of PD resting tremor. TJMs can be induced by typical antipsychotics and are known to be reduced by different drugs, including adenosine A2A receptor antagonists. The aim of the present study was to search for brain structures involved in the tremorolytic action of SCH58261, a selective A2A receptor antagonist, in TJMs induced by subchronic pimozide. Besides TJMs, we evaluated in the same animals the expression of zif-268 mRNA (neuronal responsiveness marker), and mRNA levels for glutamic acid decarboxylase 65-kDa isoform (GAD65) and vesicular glutamate transporters 1 and 2 (vGluT1/2) in selected brain structures, as markers of GABAergic and glutamatergic neurons, respectively. We found that SCH58261 reduced the pimozide-induced TJMs. Pimozide increased the zif-268 mRNA level in the striatum, nucleus accumbens (NAc) core, and substantia nigra pars reticulata (SNr). Additionally, it increased GAD65 mRNA in the striatum and SNr, and vGluT2 mRNA levels in the subthalamic nucleus (STN). A positive correlation between zif-268, GAD65 and vGluT2 mRNAs and TJMs was found. SCH58261 reversed the pimozide-increased zif-268 mRNA in the striatum and NAc core and GAD65 mRNA in the striatum and SNr. In contrast, SCH58261 did not influence vGluT2 mRNA in STN. The present study suggests an importance of the striato-subthalamo-nigro-thalamic circuit in neuroleptic-induced TJMs. The tremorolytic effect of A2A receptor blockade seems to involve this circuit bypassing, however, STN.
Collapse
Affiliation(s)
- Barbara Kosmowska
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland.
| | - Krystyna Ossowska
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland.
| | - Jadwiga Wardas
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland.
| |
Collapse
|
15
|
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
|
16
|
Wichmann C, Kuner T. Heterogeneity of glutamatergic synapses: cellular mechanisms and network consequences. Physiol Rev 2022; 102:269-318. [PMID: 34727002 DOI: 10.1152/physrev.00039.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chemical synapses are commonly known as a structurally and functionally highly diverse class of cell-cell contacts specialized to mediate communication between neurons. They represent the smallest "computational" unit of the brain and are typically divided into excitatory and inhibitory as well as modulatory categories. These categories are subdivided into diverse types, each representing a different structure-function repertoire that in turn are thought to endow neuronal networks with distinct computational properties. The diversity of structure and function found among a given category of synapses is referred to as heterogeneity. The main building blocks for this heterogeneity are synaptic vesicles, the active zone, the synaptic cleft, the postsynaptic density, and glial processes associated with the synapse. Each of these five structural modules entails a distinct repertoire of functions, and their combination specifies the range of functional heterogeneity at mammalian excitatory synapses, which are the focus of this review. We describe synapse heterogeneity that is manifested on different levels of complexity ranging from the cellular morphology of the pre- and postsynaptic cells toward the expression of different protein isoforms at individual release sites. We attempt to define the range of structural building blocks that are used to vary the basic functional repertoire of excitatory synaptic contacts and discuss sources and general mechanisms of synapse heterogeneity. Finally, we explore the possible impact of synapse heterogeneity on neuronal network function.
Collapse
Affiliation(s)
- Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg, Germany
| |
Collapse
|
17
|
Vardar G, Salazar-Lázaro A, Brockmann M, Weber-Boyvat M, Zobel S, Kumbol VWA, Trimbuch T, Rosenmund C. Reexamination of N-terminal domains of syntaxin-1 in vesicle fusion from central murine synapses. eLife 2021; 10:69498. [PMID: 34427183 PMCID: PMC8416022 DOI: 10.7554/elife.69498] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/23/2021] [Indexed: 01/11/2023] Open
Abstract
Syntaxin-1 (STX1) and Munc18-1 are two requisite components of synaptic vesicular release machinery, so much so synaptic transmission cannot proceed in their absence. They form a tight complex through two major binding modes: through STX1’s N-peptide and through STX1’s closed conformation driven by its Habc- domain. However, physiological roles of these two reportedly different binding modes in synapses are still controversial. Here we characterized the roles of STX1’s N-peptide, Habc-domain, and open conformation with and without N-peptide deletion using our STX1-null mouse model system and exogenous reintroduction of STX1A mutants. We show, on the contrary to the general view, that the Habc-domain is absolutely required and N-peptide is dispensable for synaptic transmission. However, STX1A’s N-peptide plays a regulatory role, particularly in the Ca2+-sensitivity and the short-term plasticity of vesicular release, whereas STX1’s open conformation governs the vesicle fusogenicity. Strikingly, we also show neurotransmitter release still proceeds when the two interaction modes between STX1A and Munc18-1 are presumably intervened, necessitating a refinement of the conceptualization of STX1A–Munc18-1 interaction.
Collapse
Affiliation(s)
- Gülçin Vardar
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Andrea Salazar-Lázaro
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Marisa Brockmann
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Marion Weber-Boyvat
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Sina Zobel
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | | | - Thorsten Trimbuch
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Christian Rosenmund
- Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
Porter DT, Goodman RL, Hileman SM, Lehman MN. Evidence that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase is dependent on increasing levels of oestradiol. J Neuroendocrinol 2021; 33:e12945. [PMID: 33713519 PMCID: PMC7959185 DOI: 10.1111/jne.12945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 01/12/2021] [Accepted: 01/26/2021] [Indexed: 01/09/2023]
Abstract
Neurones in the arcuate nucleus co-expressing kisspeptin, neurokinin B (NKB) and dynorphin (KNDy) play a critical role in the control of gonadotrophin-releasing hormone (GnRH) and luteinising hormone (LH) secretion. In sheep, KNDy neurones mediate both steroid-negative- and -positive-feedback during pulsatile and preovulatory surge secretions of GnRH/LH, respectively. In addition, KNDy neurones receive glutamatergic inputs expressing vGlut2, a glutamate transporter that serves as a marker for those terminals, from both KNDy neurones and other populations of glutamatergic neurones. Previous work reported higher numbers of vGlut2-positive axonal inputs onto KNDy neurones during the LH surge than in luteal phase ewes. In the present study, we further examined the effects of the ovarian steroids progesterone (P) and oestradiol (E2 ) on glutamatergic inputs to KNDy neurones. Ovariectomised (OVX) ewes received either no further treatment (OVX) or steroid treatments that mimicked the luteal phase (low E2 + P), and early (low E2 ) or late follicular (high E2 ) phases of the oestrous cycle (n = 4 or 5 per group). Brain sections were processed for triple-label immunofluorescent detection of NKB/vGlut2/synaptophysin and analysed using confocal microscopy. We found higher numbers of vGlut2 inputs onto KNDy neurones in high E2 compared to the other three treatment groups. These results suggest that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase depend on increasing levels of E2 required for the preovulatory GnRH/surge. These synaptic changes likely contribute to the positive-feedback action of oestrogen on GnRH/LH secretion and thus the generation of the preovulatory surge in the sheep.
Collapse
Affiliation(s)
- Danielle T. Porter
- Neuroscience Graduate Program, Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center
| | | | | | - Michael N. Lehman
- Brain Health Research Institute and Department of Biological Sciences, Kent State University
- Corresponding author and reprint requests to: Michael N. Lehman, Brain Health Research Institute, Kent State University, 251K Integrated Sciences Building, Kent, Ohio, 44242-0001 USA, Phone: 330-672-2732;
| |
Collapse
|
22
|
Takahashi T. Presynaptic Black Box Opened by Pioneers at Biophysics Department in University College London. Neuroscience 2020; 439:10-21. [DOI: 10.1016/j.neuroscience.2019.04.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 11/15/2022]
|
23
|
Syeda T, Foguth RM, Llewellyn E, Cannon JR. PhIP exposure in rodents produces neuropathology potentially relevant to Alzheimer's disease. Toxicology 2020; 437:152436. [PMID: 32169473 PMCID: PMC7218929 DOI: 10.1016/j.tox.2020.152436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/01/2020] [Accepted: 03/09/2020] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a public health crisis due to debilitating cognitive symptoms and lack of curative treatments, in the context of increasing prevalence. Thus, it is critical to identify modifiable risk factors. High levels of meat consumption may increase AD risk. Many toxins are formed during meat cooking such as heterocyclic aromatic amines (HAAs). Our prior studies have shown that HAAs produce dopaminergic neurotoxicity. Given the mechanistic and pathological overlap between AD and dopaminergic disorders we investigated whether exposure to 2-amino-1-methyl-6-phenylimidazo [4,5-b] pyridine (PhIP), a prevalent dietary HAA formed during high-temperature meat cooking, may produce AD-relevant neurotoxicity. Here, C57BL/6 mice were treated with 100 or 200 mg/kg PhIP for 8 h or 75 mg/kg for 4 weeks and 16 weeks. PhIP exposure for 8 h produced oxidative damage, and AD-relevant alterations in hippocampal synaptic proteins, Amyloid-beta precursor protein (APP), and β-Site amyloid precursor protein cleaving enzyme 1 (BACE1). PhIP exposure for 4 weeks resulted in an increase in BACE1. PhIP exposure for 16 weeks resulted in increased hippocampal oxidative damage, APP, BACE1, Aβ aggregation, and tau phosphorylation. Quantification of intracellular nitrotyrosine revealed oxidative damage in cholinergic neurons after 8 h, 4 weeks and 16 weeks of PhIP exposure. Our study demonstrates that increase in oxidative damage, APP and BACE1 might be a possible mechanism by which PhIP promotes Aβ aggregation. Given many patients with AD or PD exhibit neuropathological overlap, our study suggests that HAA exposure should be further studied for roles in mediating pathogenic overlap.
Collapse
Affiliation(s)
- Tauqeerunnisa Syeda
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, United States; Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN, 47907, United States
| | - Rachel M Foguth
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, United States; Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN, 47907, United States
| | - Emily Llewellyn
- Summer Research Opportunities Program, Purdue, University, West Lafayette, IN, 47907, United States; Department of Biology, Utah Valley University, Orem, Utah, 84058, United States
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, United States; Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN, 47907, United States.
| |
Collapse
|
24
|
Glial ATP and Large Pore Channels Modulate Synaptic Strength in Response to Chronic Inactivity. Mol Neurobiol 2020; 57:2856-2869. [PMID: 32388797 DOI: 10.1007/s12035-020-01919-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/14/2020] [Indexed: 01/08/2023]
Abstract
A key feature of neurotransmission is its ability to adapt to changes in neuronal environment, which is essential for many brain functions. Homeostatic synaptic plasticity (HSP) emerges as a compensatory mechanism used by neurons to adjust their excitability in response to changes in synaptic activity. Recently, glial cells emerged as modulators for neurotransmission by releasing gliotransmitters into the synaptic cleft through pathways that include P2X7 receptors (P2X7R), connexons, and pannexons. However, the role of gliotransmission in the activity-dependent adjustment of presynaptic strength is still an open question. Here, we investigated whether glial cells participate in HSP upon chronic inactivity and the role of adenosine triphosphate (ATP), connexin43 hemichannels (Cx43HCs), and pannexin1 (Panx1) channels in this process. We used immunocytochemistry against vesicular glutamate transporter 1 (vGlut1) to estimate changes in synaptic strength in hippocampal dissociated cultures. Pharmacological manipulations indicate that glial-derived ATP and P2X7R are required for HSP. In addition, inhibition of Cx43 and Panx1 channels reveals a pivotal role for these channels in the compensatory adjustment of synaptic strength, emerging as new pathways for ATP release upon inactivity. The involvement of Panx1 channels was confirmed by using Panx1-deficient animals. Lacking Panx1 in neurons is sufficient to prevent the P2X7R-dependent upregulation of presynaptic strength; however, the P2X7R-dependent compensatory adjustment of synapse density requires both neuronal and glial Panx1. Together, our data supports an essential role for glial ATP signaling and Cx43HCs and Panx1 channels in the homeostatic adjustment of synaptic strength in hippocampal cultures upon chronic inactivity.
Collapse
|
25
|
Transient Conductive Hearing Loss Regulates Cross-Modal VGLUT Expression in the Cochlear Nucleus of C57BL/6 Mice. Brain Sci 2020; 10:brainsci10050260. [PMID: 32365514 PMCID: PMC7287693 DOI: 10.3390/brainsci10050260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/19/2022] Open
Abstract
Auditory nerve fibers synapse onto the cochlear nucleus (CN) and are labeled using the vesicular glutamate transporter-1 (VGLUT-1), whereas non-auditory inputs are labeled using the VGLUT-2. However, the underlying regulatory mechanism of VGLUT expression in the CN remains unknown. We examined whether a sound level decrease, without primary neural damage, induces cellular and VGLUT expression change in the CN, and examined the potential for neural plasticity of the CN using unilateral conductive hearing loss models. We inserted earplugs in 8-week-old mice unilaterally for 4 weeks and subsequently removed them for another 4 weeks. Although the threshold of an auditory brainstem response significantly increased across all tested frequencies following earplug insertion, it completely recovered after earplug removal. Auditory deprivation had no significant impact on spiral ganglion and ventral CN (VCN) neurons’ survival. Conversely, although the cell size and VGLUT-1 expression in the VCN significantly decreased after earplug insertion, VGLUT-2 expression in the granule cell lamina significantly increased. These cell sizes decreased and the alterations in VGLUT-1 and -2 expression almost completely recovered at 1 month after earplug removal. Our results suggested that the cell size and VGLUT expression in the CN have a neuroplasticity capacity, which is regulated by increases and decreases in sound levels. Restoration of the sound levels might partly prevent cell size decrease and maintain VGLUT expression in the CN.
Collapse
|
26
|
Du X, Li J, Li M, Yang X, Qi Z, Xu B, Liu W, Xu Z, Deng Y. Research progress on the role of type I vesicular glutamate transporter (VGLUT1) in nervous system diseases. Cell Biosci 2020; 10:26. [PMID: 32158532 PMCID: PMC7057577 DOI: 10.1186/s13578-020-00393-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Glutamate (Glu) is the predominant excitatory neurotransmitter in the central nervous system (CNS). Glutamatergic transmission is critical for controlling neuronal activity. In presynaptic neurons, Glu is stored in synaptic vesicles and released by stimulation. The homeostasis of glutamatergic system is maintained by a set of transporters in the membrane of synaptic vesicles. The family of vesicular Glu transporters in mammals is comprised of three highly homologous proteins: VGLUT1-3. Among them, VGLUT1 accounts for the largest proportion. However, most of the Glu is transported into the synaptic vesicles via the type 1 vesicle Glu transporter (VGLUT1). So, the expression of particular VGLUT1 is largely complementary with limited overlap and so far it is most specific markers for neurons that use Glu as neurotransmitter. Controlling the activity of VGLUT1 could potentially modulate the efficiency of excitatory neuro-transmission and change the filling level of synaptic vesicles. This review summarizes the recent knowledge concerning molecular and functional characteristic of VGLUT1, their development, contribution to a series of central nervous system and peripheral nervous system diseases such as learning and memory disorders, Alzheimer's disease, Parkinson's disease and sensitized nociception or pain pathology et al.
Collapse
Affiliation(s)
- Xianchao Du
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Minghui Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Xinxin Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Zhipeng Qi
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| |
Collapse
|
27
|
Kosmowska B, Ossowska K, Konieczny J, Lenda T, Berghauzen-Maciejewska K, Wardas J. Inhibition of Excessive Glutamatergic Transmission in the Ventral Thalamic Nuclei by a Selective Adenosine A1 Receptor Agonist, 5′-Chloro-5′-Deoxy-(±)-ENBA Underlies its Tremorolytic Effect in the Harmaline-Induced Model of Essential Tremor. Neuroscience 2020; 429:106-118. [DOI: 10.1016/j.neuroscience.2019.12.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/16/2019] [Accepted: 12/28/2019] [Indexed: 12/20/2022]
|
28
|
Ma YN, Sun YX, Wang T, Wang H, Zhang Y, Su YA, Li JT, Si TM. Subchronic MK-801 treatment during adolescence induces long-term, not permanent, excitatory-inhibitory imbalance in the rat hippocampus. Eur J Pharmacol 2020; 867:172807. [DOI: 10.1016/j.ejphar.2019.172807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/28/2023]
|
29
|
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
|
30
|
Wang W, Zeng F, Hu Y, Li X. A Mini-Review of the Role of Glutamate Transporter in Drug Addiction. Front Neurol 2019; 10:1123. [PMID: 31695674 PMCID: PMC6817614 DOI: 10.3389/fneur.2019.01123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 10/08/2019] [Indexed: 12/29/2022] Open
Abstract
Goals: The development of new treatment for drug abuse requires identification of targetable molecular mechanisms. The pathology of glutamate neurotransmission system in the brain reward circuit is related to the relapse of multiple drugs. Glutamate transporter regulates glutamate signaling by removing excess glutamate from the synapse. And the mechanisms between glutamate transporter and drug addiction are still unclear. Methods: A systematic review of the literature searched in Pubmed and reporting drug addiction in relation to glutamate transporter. Studies were screened by title, abstract, and full text. Results: This review is to highlight the effects of drug addiction on glutamate transporter and glutamate uptake, and targeting glutamate transporter as an addictive drug addiction treatment. We focus on the roles of glutamate transporter in different brain regions in drug addiction. More importantly, we suggest the functional roles of glutamate transporter may prove beneficial in the treatment of drug addiction. Conclusion: Overall, understanding how glutamate transporter impacts central nervous system may provide a new insight for treatment of drug addiction.
Collapse
Affiliation(s)
- Wenjun Wang
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Yingying Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| |
Collapse
|
31
|
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: 65] [Impact Index Per Article: 10.8] [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
|
32
|
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
|
33
|
Bloemer J, Pinky PD, Smith WD, Bhattacharya D, Chauhan A, Govindarajulu M, Hong H, Dhanasekaran M, Judd R, Amin RH, Reed MN, Suppiramaniam V. Adiponectin Knockout Mice Display Cognitive and Synaptic Deficits. Front Endocrinol (Lausanne) 2019; 10:819. [PMID: 31824431 PMCID: PMC6886372 DOI: 10.3389/fendo.2019.00819] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022] Open
Abstract
Adiponectin is an adipokine that has recently been under investigation for potential neuroprotective effects in various brain disorders including Alzheimer's disease, stroke, and depression. Adiponectin receptors (AdipoR1 and AdipoR2) are found throughout various brain regions, including the hippocampus. However, the role of these receptors in synaptic and cognitive function is not clear. Therefore, the goal of the current study was to evaluate synaptic and cognitive function in the absence of adiponectin. The current study utilized 12-month-old adiponectin knockout (APN-KO) mice and age-matched controls to study cognitive and hippocampal synaptic alterations. We determined that AdipoR1 and AdipoR2 are present in the synaptosome, with AdipoR2 displaying increased presynaptic vs. postsynaptic localization, whereas AdipoR1 was enriched in both the presynaptic and postsynaptic fractions. APN-KO mice displayed cognitive deficits in the novel object recognition (NOR) and Y-maze tests. This was mirrored by deficits in long-term potentiation (LTP) of the hippocampal Schaefer collateral pathway in APN-KO mice. APN-KO mice also displayed a reduction in basal synaptic transmission and an increase in presynaptic release probability. Deficits in LTP were rescued through hippocampal slice incubation with the adiponectin receptor agonist, AdipoRon, indicating that acute alterations in adiponectin receptor signaling influence synaptic function. Along with the deficits in LTP, altered levels of key presynaptic and postsynaptic proteins involved in glutamatergic neurotransmission were observed in APN-KO mice. Taken together, these results indicate that adiponectin is an important regulator of cognition and synaptic function in the hippocampus. Future studies should examine the role of specific adiponectin receptors in synaptic processes.
Collapse
Affiliation(s)
- Jenna Bloemer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Warren D. Smith
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Dwipayan Bhattacharya
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Alisa Chauhan
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Hao Hong
- Center for Neuroscience, Auburn University, Auburn, AL, United States
- Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Robert Judd
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Rajesh H. Amin
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
- *Correspondence: Miranda N. Reed
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
- Vishnu Suppiramaniam
| |
Collapse
|
34
|
Trudeau LE, El Mestikawy S. Glutamate Cotransmission in Cholinergic, GABAergic and Monoamine Systems: Contrasts and Commonalities. Front Neural Circuits 2018; 12:113. [PMID: 30618649 PMCID: PMC6305298 DOI: 10.3389/fncir.2018.00113] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/03/2018] [Indexed: 11/13/2022] Open
Abstract
Multiple discoveries made since the identification of vesicular glutamate transporters (VGLUTs) two decades ago revealed that many neuronal populations in the brain use glutamate in addition to their "primary" neurotransmitter. Such a mode of cotransmission has been detected in dopamine (DA), acetylcholine (ACh), serotonin (5-HT), norepinephrine (NE) and surprisingly even in GABA neurons. Interestingly, work performed by multiple groups during the past decade suggests that the use of glutamate as a cotransmitter takes different forms in these different populations of neurons. In the present review, we will provide an overview of glutamate cotransmission in these different classes of neurons, highlighting puzzling differences in: (1) the proportion of such neurons expressing a VGLUT in different brain regions and at different stages of development; (2) the sub-cellular localization of the VGLUT; (3) the localization of the VGLUT in relation to the neurons' other vesicular transporter; and (4) the functional role of glutamate cotransmission.
Collapse
Affiliation(s)
- Louis-Eric Trudeau
- CNS Research Group, Department of Pharmacology and Physiology, Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Salah El Mestikawy
- Department of Psychiatry, Faculty of Medicine, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Sorbonne Universités, Université Pierre et Marie Curie UM 119-CNRS UMR 8246-INSERM U1130, Neurosciences Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS), Paris, France
| |
Collapse
|
35
|
Moyer CE, Hiolski EM, Marcinek DJ, Lefebvre KA, Smith DR, Zuo Y. Repeated low level domoic acid exposure increases CA1 VGluT1 levels, but not bouton density, VGluT2 or VGAT levels in the hippocampus of adult mice. HARMFUL ALGAE 2018; 79:74-86. [PMID: 30420019 PMCID: PMC6237202 DOI: 10.1016/j.hal.2018.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Domoic acid (DA) is a neurotoxin produced during harmful algal blooms that accumulates in marine organisms that serve as food resources for humans. While acute DA neurotoxicity can cause seizures and hippocampal lesions, less is known regarding how chronic, subacute DA exposure in adulthood impacts the hippocampus. With more frequent occurrences of harmful algal blooms, it is important to understand the potential impact of repeated, low-level DA exposure on human health. To model repeated, low-dose DA exposure, adult mice received a single low-dose (0.75 ± 0.05 μg/g) of DA or vehicle weekly for 22 consecutive weeks. Quantitative immunohistochemistry was performed to assess the effects of repeated, low-level DA exposure on hippocampal cells and synapses. Vesicular glutamate transporter 1 (VGluT1) immunoreactivity within excitatory boutons in CA1 of DA-exposed mice was increased. Levels of other vesicular transporter proteins (i.e., VGluT2 and the vesicular GABA transporter (VGAT)) within boutons, and corresponding bouton densities, were not significantly altered in CA1, CA3, or dentate gyrus. There were no significant changes in neuron density or glial fibrillary acidic protein (GFAP) immunoreactivity following chronic, low-dose exposure. This suggests that repeated low doses of DA, unlike high doses of DA, do not cause neuronal loss or astrocyte activation in hippocampus in adult mice. Instead, these findings demonstrate that repeated exposure to low levels of DA leads to subtle changes in VGluT1 expression within CA1 excitatory boutons, which may alter glutamatergic transmission in CA1 and disrupt behaviors dependent on spatial memory.
Collapse
Affiliation(s)
- Caitlin E Moyer
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, United States
| | - Emma M Hiolski
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, United States
| | - David J Marcinek
- Departments of Radiology, Pathology, and Bioengineering, University of Washington, South Lake Union Campus, 850 Republican St., Brotman 142, Box 358050, Seattle, WA, 98109, United States
| | - Kathi A Lefebvre
- Environmental and Fisheries Science Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. East, Seattle, WA 98112, United States
| | - Donald R Smith
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, United States
| | - Yi Zuo
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, United States.
| |
Collapse
|
36
|
Estradiol Drives the Anorexigenic Activity of Proopiomelanocortin Neurons in Female Mice. eNeuro 2018; 5:eN-NWR-0103-18. [PMID: 30310864 PMCID: PMC6179576 DOI: 10.1523/eneuro.0103-18.2018] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/11/2018] [Accepted: 07/02/2018] [Indexed: 12/14/2022] Open
Abstract
Energy balance is regulated by anorexigenic proopiomelanocortin (POMC) and orexigenic neuropeptide Y/agouti-related peptide (NPY/AgRP) neurons of the hypothalamic arcuate nucleus. POMC neurons make extensive projections and are thought to release both amino acid and peptide neurotransmitters. However, whether they communicate directly with NPY/AgRP neurons is debated. Initially, using single-cell RT-PCR, we determined that mouse POMCeGFP neurons express Slc17a6 (Vglut2) and Slc18a2 (Vmat2), but not Slc31a1 (Vgat) mRNA, suggesting glutamate and non-canonical GABA release. Quantitative (q)RT-PCR of POMCeGFP cells revealed that Vglut2 and Vmat2 expression was significantly increased in E2- versus oil-treated, ovariectomized (OVX) female mice. Since 17β-estradiol (E2) is anorexigenic, we hypothesized that an underlying mechanism is enhancement of POMC signaling. Therefore, we optogenetically stimulated POMC neurons in hypothalamic slices to examine evoked release of neurotransmitters onto NPY/AgRP neurons. Using brief light pulses, we primarily observed glutamatergic currents and, based on the paired pulse ratio (PPR), determined that release probability was higher in E2- versus oil-treated, OVX female, congruent with increased Vlgut2 expression. Moreover, bath perfusion of the Gq-coupled membrane estrogen receptor (ER) agonist STX recapitulated the effects of E2 treatment. In addition, high-frequency (20 Hz) stimulation generated a slow outward current that reversed near Ek+ and was antagonized by naloxone, indicative of β-endorphin release. Furthermore, individual NPY/AgRP neurons were found to express Oprm1, the transcript for μ-opioid receptor, and DAMGO, a selective agonist, elicited an outward current. Therefore, POMC excitability and neurotransmission are enhanced by E2, which would facilitate decreased food consumption through marked inhibition of NPY/AgRP neurons.
Collapse
|
37
|
Shaib AH, Staudt A, Harb A, Klose M, Shaaban A, Schirra C, Mohrmann R, Rettig J, Becherer U. Paralogs of the Calcium-Dependent Activator Protein for Secretion Differentially Regulate Synaptic Transmission and Peptide Secretion in Sensory Neurons. Front Cell Neurosci 2018; 12:304. [PMID: 30254567 PMCID: PMC6141663 DOI: 10.3389/fncel.2018.00304] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/21/2018] [Indexed: 12/29/2022] Open
Abstract
The two paralogs of the calcium-dependent activator protein for secretion (CAPS) are priming factors for synaptic vesicles (SVs) and neuropeptide containing large dense-core vesicles (LDCVs). Yet, it is unclear whether CAPS1 and CAPS2 regulate exocytosis of these two vesicle types differentially in dorsal root ganglion (DRG) neurons, wherein synaptic transmission and neuropeptide release are of equal importance. These sensory neurons transfer information from the periphery to the spinal cord (SC), releasing glutamate as the primary neurotransmitter, with co-transmission via neuropeptides in a subset of so called peptidergic neurons. Neuropeptides are key components of the information-processing machinery of pain perception and neuropathic pain generation. Here, we compared the ability of CAPS1 and CAPS2 to support priming of both vesicle types in single and double knock-out mouse (DRG) neurons using a variety of high-resolution live cell imaging methods. While CAPS1 was localized to synapses of all DRG neurons and promoted synaptic transmission, CAPS2 was found exclusively in peptidergic neurons and mediated LDCV exocytosis. Intriguingly, ectopic expression of CAPS2 empowered non-peptidergic neurons to drive LDCV fusion, thereby identifying CAPS2 as an essential molecular determinant for peptidergic signaling. Our results reveal that these distinct functions of both CAPS paralogs are based on their differential subcellular localization in DRG neurons. Our data suggest a major role for CAPS2 in neuropathic pain via control of neuropeptide release.
Collapse
Affiliation(s)
- Ali H. Shaib
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Angelina Staudt
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ali Harb
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Margarete Klose
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ahmed Shaaban
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Claudia Schirra
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ralf Mohrmann
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Jens Rettig
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ute Becherer
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| |
Collapse
|
38
|
Prion acute synaptotoxicity is largely driven by protease-resistant PrPSc species. PLoS Pathog 2018; 14:e1007214. [PMID: 30089152 PMCID: PMC6101418 DOI: 10.1371/journal.ppat.1007214] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/20/2018] [Accepted: 07/12/2018] [Indexed: 01/09/2023] Open
Abstract
Although misfolding of normal prion protein (PrPC) into abnormal conformers (PrPSc) is critical for prion disease pathogenesis our current understanding of the underlying molecular pathophysiology is rudimentary. Exploiting an electrophysiology paradigm, herein we report that at least modestly proteinase K (PK)-resistant PrPSc (PrPres) species are acutely synaptotoxic. Brief exposure to ex vivo PrPSc from two mouse-adapted prion strains (M1000 and MU02) prepared as crude brain homogenates (cM1000 and cMU02) and cell lysates from chronically M1000-infected RK13 cells (MoRK13-Inf) caused significant impairment of hippocampal CA1 region long-term potentiation (LTP), with the LTP disruption approximating that reported during the evolution of murine prion disease. Proof of PrPSc (especially PrPres) species as the synaptotoxic agent was demonstrated by: significant rescue of LTP following selective immuno-depletion of total PrP from cM1000 (dM1000); modestly PK-treated cM1000 (PK+M1000) retaining full synaptotoxicity; and restoration of the LTP impairment when employing reconstituted, PK-eluted, immuno-precipitated M1000 preparations (PK+IP-M1000). Additional detailed electrophysiological analyses exemplified by impairment of post-tetanic potentiation (PTP) suggest possible heightened pre-synaptic vulnerability to the acute synaptotoxicity. This dysfunction correlated with cumulative insufficiency of replenishment of the readily releasable pool (RRP) of vesicles during repeated high-frequency stimulation utilised for induction of LTP. Broadly comparable results with LTP and PTP impairment were obtained utilizing hippocampal slices from PrPC knockout (PrPo/o) mice, with cM1000 serial dilution assessments revealing similar sensitivity of PrPo/o and wild type (WT) slices. Size fractionation chromatography demonstrated that synaptotoxic PrP correlated with PK-resistant species >100kDa, consistent with multimeric PrPSc, with levels of these species >6 ng/ml appearing sufficient to induce synaptic dysfunction. Biochemical analyses of hippocampal slices manifesting acute synaptotoxicity demonstrated reduced levels of multiple key synaptic proteins, albeit with noteworthy differences in PrPo/o slices, while such changes were absent in hippocampi demonstrating rescued LTP through treatment with dM1000. Our findings offer important new mechanistic insights into the synaptic impairment underlying prion disease, enhancing prospects for development of targeted effective therapies. Misfolding of the normal prion protein (PrPC) into disease-associated conformations (PrPSc) is the critical initiating step for prion diseases. Similar to other neurodegenerative disorders, progressive failure of brain synapses is considered a primary deleterious event underpinning prion disease evolution. Our current understanding of the underlying mechanisms associated with synaptic failure is rudimentary contributing to difficulties in developing effective treatments. Herein we report the use of an electrophysiology paradigm that allowed us to demonstrate that at least modestly proteinase K (PK)-resistant PrPSc species from two mouse-adapted prion strains (M1000 and MU02) are directly synaptotoxic causing significant acute impairment of hippocampal CA1 region long-term potentiation (LTP). Of note, the LTP disruption approximated that reported in prion animal models. Additional detailed analyses provided novel pathophysiological insights suggesting possible heightened pre-synaptic vulnerability to the acute synaptotoxicity through impairment of replenishment of the readily releasable pool of neurotransmitter vesicles, while biochemical analyses demonstrated reduced levels of multiple key pre-and post-synaptic proteins. Broadly similar acute synaptic dysfunction and dose-response susceptibility were observed in slices from mice not expressing PrPC albeit with minor but noteworthy differences in electrophysiological and biochemical findings. Our study offers important new mechanistic insights into the synaptic impairment underlying prion disease, enhancing prospects for development effective therapies.
Collapse
|
39
|
Qiu J, Rivera HM, Bosch MA, Padilla SL, Stincic TL, Palmiter RD, Kelly MJ, Rønnekleiv OK. Estrogenic-dependent glutamatergic neurotransmission from kisspeptin neurons governs feeding circuits in females. eLife 2018; 7:e35656. [PMID: 30079889 PMCID: PMC6103748 DOI: 10.7554/elife.35656] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 07/24/2018] [Indexed: 11/13/2022] Open
Abstract
The neuropeptides tachykinin2 (Tac2) and kisspeptin (Kiss1) in hypothalamic arcuate nucleus Kiss1 (Kiss1ARH) neurons are essential for pulsatile release of GnRH and reproduction. Since 17β-estradiol (E2) decreases Kiss1 and Tac2 mRNA expression in Kiss1ARH neurons, the role of Kiss1ARH neurons during E2-driven anorexigenic states and their coordination of POMC and NPY/AgRP feeding circuits have been largely ignored. Presently, we show that E2 augmented the excitability of Kiss1ARH neurons by amplifying Cacna1g, Hcn1 and Hcn2 mRNA expression and T-type calcium and h-currents. E2 increased Slc17a6 mRNA expression and glutamatergic synaptic input to arcuate neurons, which excited POMC and inhibited NPY/AgRP neurons via metabotropic receptors. Deleting Slc17a6 in Kiss1 neurons eliminated glutamate release and led to conditioned place preference for sucrose in E2-treated KO female mice. Therefore, the E2-driven increase in Kiss1 neuronal excitability and glutamate neurotransmission may play a key role in governing the motivational drive for palatable food in females.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Physiology and PharmacologyOregon Health and Science UniversityPortlandUnited States
| | - Heidi M Rivera
- Department of Physiology and PharmacologyOregon Health and Science UniversityPortlandUnited States
| | - Martha A Bosch
- Department of Physiology and PharmacologyOregon Health and Science UniversityPortlandUnited States
| | - Stephanie L Padilla
- Department of BiochemistryHoward Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Todd L Stincic
- Department of Physiology and PharmacologyOregon Health and Science UniversityPortlandUnited States
| | - Richard D Palmiter
- Department of BiochemistryHoward Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Martin J Kelly
- Department of Physiology and PharmacologyOregon Health and Science UniversityPortlandUnited States
- Division of NeuroscienceOregon National Primate Research Center, Oregon Health and Science UniversityBeavertonUnited States
| | - Oline K Rønnekleiv
- Department of Physiology and PharmacologyOregon Health and Science UniversityPortlandUnited States
- Division of NeuroscienceOregon National Primate Research Center, Oregon Health and Science UniversityBeavertonUnited States
| |
Collapse
|
40
|
Chang R, Eriksen J, Edwards RH. The dual role of chloride in synaptic vesicle glutamate transport. eLife 2018; 7:e34896. [PMID: 30040066 PMCID: PMC6057745 DOI: 10.7554/elife.34896] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/07/2018] [Indexed: 01/23/2023] Open
Abstract
The transport of glutamate into synaptic vesicles exhibits an unusual form of regulation by Cl- as well as an associated Cl- conductance. To distinguish direct effects of Cl- on the transporter from indirect effects via the driving force Δψ, we used whole endosome recording and report the first currents due to glutamate flux by the vesicular glutamate transporters (VGLUTs). Chloride allosterically activates the VGLUTs from both sides of the membrane, and we find that neutralization of an arginine in transmembrane domain four suffices for the lumenal activation. The dose dependence suggests that Cl- permeates through a channel and glutamate through a transporter. Competition between the anions nonetheless indicates that they use a similar permeation pathway. By controlling both ionic gradients and Δψ, endosome recording isolates different steps in the process of synaptic vesicle filling, suggesting distinct roles for Cl- in both allosteric activation and permeation.
Collapse
Affiliation(s)
- Roger Chang
- Department of PhysiologyUCSF School of MedicineSan FranciscoUnited States
- Department of NeurologyUCSF School of MedicineSan FranciscoUnited States
- Graduate Program in Biomedical SciencesUCSF School of MedicineSan FranciscoUnited States
| | - Jacob Eriksen
- Department of PhysiologyUCSF School of MedicineSan FranciscoUnited States
- Department of NeurologyUCSF School of MedicineSan FranciscoUnited States
| | - Robert H Edwards
- Department of PhysiologyUCSF School of MedicineSan FranciscoUnited States
- Department of NeurologyUCSF School of MedicineSan FranciscoUnited States
- Graduate Program in Biomedical SciencesUCSF School of MedicineSan FranciscoUnited States
- Kavli Institute for Fundamental NeuroscienceUCSF School of MedicineSan FranciscoUnited States
- Weill Institute for NeurosciencesUCSF School of MedicineSan FranciscoUnited States
| |
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
|
A mouse model of autism implicates endosome pH in the regulation of presynaptic calcium entry. Nat Commun 2018; 9:330. [PMID: 29362376 PMCID: PMC5780507 DOI: 10.1038/s41467-017-02716-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/19/2017] [Indexed: 11/21/2022] Open
Abstract
Psychoactive compounds such as chloroquine and amphetamine act by dissipating the pH gradient across intracellular membranes, but the physiological mechanisms that normally regulate organelle pH remain poorly understood. Interestingly, recent human genetic studies have implicated the endosomal Na+/H+ exchanger NHE9 in both autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD). Plasma membrane NHEs regulate cytosolic pH, but the role of intracellular isoforms has remained unclear. We now find that inactivation of NHE9 in mice reproduces behavioral features of ASD including impaired social interaction, repetitive behaviors, and altered sensory processing. Physiological characterization reveals hyperacidic endosomes, a cell-autonomous defect in glutamate receptor expression and impaired neurotransmitter release due to a defect in presynaptic Ca2+ entry. Acute inhibition of synaptic vesicle acidification rescues release but without affecting the primary defect due to loss of NHE9. The Na+/H+ exchanger NHE9 is proposed to regulate the H+ electrochemical gradient across endosomal membranes. Here, the authors find that NHE9 knockout mice show autism spectrum disorder-like behaviors and disrupted synaptic vesicle exocytosis due to impaired presynaptic calcium entry.
Collapse
|
43
|
Horváth HR, Fazekas CL, Balázsfi D, Jain SK, Haller J, Zelena D. Contribution of Vesicular Glutamate Transporters to Stress Response and Related Psychopathologies: Studies in VGluT3 Knockout Mice. Cell Mol Neurobiol 2018; 38:37-52. [PMID: 28776199 DOI: 10.1007/s10571-017-0528-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/28/2017] [Indexed: 10/19/2022]
Abstract
Maintenance of the homeostasis in a constantly changing environment is a fundamental process of life. Disturbances of the homeostatic balance is defined as stress response and is induced by wide variety of challenges called stressors. Being the main excitatory neurotransmitter of the central nervous system glutamate is important in the adaptation process of stress regulating both the catecholaminergic system and the hypothalamic-pituitary-adrenocortical axis. Data are accumulating about the role of different glutamatergic receptors at all levels of these axes, but little is known about the contribution of different vesicular glutamate transporters (VGluT1-3) characterizing the glutamatergic neurons. Here we summarize basic knowledge about VGluTs, their role in physiological regulation of stress adaptation, as well as their contribution to stress-related psychopathology. Most of our knowledge comes from the VGluT3 knockout mice, as VGluT1 and 2 knockouts are not viable. VGluT3 was discovered later than, and is not as widespread as the VGluT1 and 2. It may co-localize with other transmitters, and participate in retrograde signaling; as such its role might be unique. Previous reports using VGluT3 knockout mice showed enhanced anxiety and innate fear compared to wild type. Moreover, these knockout animals had enhanced resting corticotropin-releasing hormone mRNA levels in the hypothalamus and disturbed glucocorticoid stress responses. In conclusion, VGluT3 participates in stress adaptation regulation. The neuroendocrine changes observed in VGluT3 knockout mice may contribute to their anxious, fearful phenotype.
Collapse
Affiliation(s)
- Hanga Réka Horváth
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
| | - Diána Balázsfi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 26, Üllői út, 1085, Budapest, Hungary
| | | | - József Haller
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
| | - Dóra Zelena
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary.
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary.
| |
Collapse
|
44
|
Yu W, Shi X, Cui X, Niu Y, Zhang W, Bai X, Wang Q, Hu L, Wang S. Jian-Pi-Zhi-Dong-Decoction regulates the expression of glutamate transporters to attenuate glutamate excitotoxicity and exerts anti-tics effects in Tourette syndrome model rats. Neuropsychiatr Dis Treat 2018; 14:3381-3392. [PMID: 30587990 PMCID: PMC6301307 DOI: 10.2147/ndt.s185169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE This study explored whether Jian-Pi-Zhi-Dong-Decoction (JPZDD) could regulate the metabolism of glutamate (GLU) and its transporters in the striatum to exert anti-tics effects in Tourette syndrome (TS) rats. MATERIALS AND METHODS We randomly assigned 56 Sprague Dawley rats into four groups, each with 14 rats: control, model, tiapride (Tia), and JPZDD. TS groups (model, Tia, and JPZDD) received intraperitoneal injection of 3,3'-iminodipropionitrile for 7 days to establish TS model. Thereafter, rats in the four groups were treated differently once a day for 6 weeks. Behavioral evaluation was performed each week by using stereotypy recording and autonomic activity test. The level of GLU in the striatum was examined by high-performance liquid chromatography. Expression of EAAT1 and VGLUT1 were measured by quantitative real-time PCR (qRT-PCR) and laser scanning confocal microscope. RESULTS Compared with the model group, the stereotypy score and autonomic activity were decreased in Tia and JPZDD groups. Notably, the model group had increased concentration of GLU, which decreased after JPZDD and Tia treatments. In the model group, EAAT1 and glial cells were highly co-expressed and the relative fluorescence intensity (FI) of EAAT1 was significantly lower than that in the control group. Treatment with JPZDD and Tia increased the relative FI of EAAT1. The mRNA level of EAAT1 decreased in the model group compared to that in the control group, although it was significantly elevated following JPZDD or Tia treatment. In the model group, there was low co-expression of VGLUT1 and axon cells and the FI of VGLUT1 was remarkably increased relative to that in the control group and reduced following treatment with JPZDD and Tia. A similar trend was observed in the mRNA and protein expression of VGLUT1, although it was not statistically significant. CONCLUSION The mechanism by which JPZDD alleviated behavioral dysfunction of TS rats may be associated with maintaining normal GLU transport by upregulating EAAT1 and down-regulating VGLUT1 in the striatum.
Collapse
Affiliation(s)
- Wenjing Yu
- Department of Pediatrics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowei Shi
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xia Cui
- Department of Pediatrics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Niu
- Department of Pediatrics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wen Zhang
- Department of Pediatrics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Bai
- Department of Pediatrics, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qian Wang
- Department of Pediatrics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lijun Hu
- Department of Pediatrics, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sumei Wang
- Department of Pediatrics, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China,
| |
Collapse
|
45
|
Martineau M, Guzman RE, Fahlke C, Klingauf J. VGLUT1 functions as a glutamate/proton exchanger with chloride channel activity in hippocampal glutamatergic synapses. Nat Commun 2017; 8:2279. [PMID: 29273736 PMCID: PMC5741633 DOI: 10.1038/s41467-017-02367-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/24/2017] [Indexed: 12/18/2022] Open
Abstract
Glutamate is the major excitatory transmitter in the vertebrate nervous system. To maintain synaptic efficacy, recycling synaptic vesicles (SV) are refilled with glutamate by vesicular glutamate transporters (VGLUTs). The dynamics and mechanism of glutamate uptake in intact neurons are still largely unknown. Here, we show by live-cell imaging with pH- and chloride-sensitive fluorescent probes in cultured hippocampal neurons of wild-type and VGLUT1-deficient mice that in SVs VGLUT functions as a glutamate/proton exchanger associated with a channel-like chloride conductance. After endocytosis most internalized Cl− is substituted by glutamate in an electrically, and presumably osmotically, neutral manner, and this process is driven by both the Cl− gradient itself and the proton motive force provided by the vacuolar H+-ATPase. Our results shed light on the transport mechanism of VGLUT under physiological conditions and provide a framework for how modulation of glutamate transport via Cl− and pH can change synaptic strength. During neurotransmission synaptic vesicles are filled with glutamate by vesicular glutamate transporters (VGLUTs). Here, authors image intact neurons and show that in synaptic vesicles VGLUT functions as a glutamate/proton exchanger associated with a channel-like chloride conductance.
Collapse
Affiliation(s)
- Magalie Martineau
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Muenster, 48149, Muenster, Germany. .,University of Bordeaux and Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France.
| | - Raul E Guzman
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Christoph Fahlke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Jürgen Klingauf
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Muenster, 48149, Muenster, Germany. .,IZKF Münster and Cluster of Excellence EXC 1003, Cells in Motion (CiM), 48149, Muenster, Germany.
| |
Collapse
|
46
|
Kawano H, Oyabu K, Yamamoto H, Eto K, Adaniya Y, Kubota K, Watanabe T, Hirano-Iwata A, Nabekura J, Katsurabayashi S, Iwasaki K. Astrocytes with previous chronic exposure to amyloid β-peptide fragment 1-40 suppress excitatory synaptic transmission. J Neurochem 2017; 143:624-634. [PMID: 29076533 DOI: 10.1111/jnc.14247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/27/2017] [Accepted: 10/13/2017] [Indexed: 11/28/2022]
Abstract
Synaptic dysfunction and neuronal death are responsible for cognitive and behavioral deficits in Alzheimer's disease (AD). It is well known that such neurological abnormalities are preceded by long-term exposure of amyloid β-peptide (Aβ) and/or hyperphosphorylated tau prior. In addition to the neurological deficit, astrocytes as a major glial cell type in the brain, significantly participate in the neuropathogenic mechanisms underlying synaptic modulation. Although astrocytes play a significant key role in modulating synaptic transmission, little is known on whether astrocyte dysfunction caused by such long-term Aβ exposure affects synapse formation and function. Here, we show that synapse formation and synaptic transmission are attenuated in hippocampal-naïve neurons co-cultured with astrocytes that have previously experienced chronic Aβ1-40 exposure. In this abnormal astrocytic condition, hippocampal neurons exhibit decrements of evoked excitatory post-synaptic currents (EPSCs) and miniature EPSC frequency. Furthermore, size of readily releasable synaptic pools and number of excitatory synapses were also significantly decreased. Contrary to these negative effects, release probability at individual synapses was significantly increased in the same astrocytic condition. Taken together, our data indicate that lower synaptic transmission caused by astrocytes previously, and chronically, exposed to Aβ1-40 is attributable to a small number of synapses with higher release probability.
Collapse
Affiliation(s)
- Hiroyuki Kawano
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kohei Oyabu
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Hideaki Yamamoto
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aoba-ku, Sendai, Japan
| | - Kei Eto
- Division of Homeostatic Development, Department of Fundamental Neuroscience, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Yuna Adaniya
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| | - Ayumi Hirano-Iwata
- Advanced Institute for Materials Research, Tohoku University, Aoba-ku, Sendai, Japan.,Research Institute of Electrical Communication, Tohoku University, Aoba-ku, Sendai, Japan
| | - Junichi Nabekura
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan.,Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki, Japan.,CREST, Japan Science and Technology Agency (JST), Kawaguchi, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.,A.I.G. Collaborative Research Institute for Aging and Brain Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
47
|
Pecoraro-Bisogni F, Lignani G, Contestabile A, Castroflorio E, Pozzi D, Rocchi A, Prestigio C, Orlando M, Valente P, Massacesi M, Benfenati F, Baldelli P. REST-Dependent Presynaptic Homeostasis Induced by Chronic Neuronal Hyperactivity. Mol Neurobiol 2017; 55:4959-4972. [PMID: 28786015 DOI: 10.1007/s12035-017-0698-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022]
Abstract
Homeostatic plasticity is a regulatory feedback response in which either synaptic strength or intrinsic excitability can be adjusted up or down to offset sustained changes in neuronal activity. Although a growing number of evidences constantly provide new insights into these two apparently distinct homeostatic processes, a unified molecular model remains unknown. We recently demonstrated that REST is a transcriptional repressor critical for the downscaling of intrinsic excitability in cultured hippocampal neurons subjected to prolonged elevation of electrical activity. Here, we report that, in the same experimental system, REST also participates in synaptic homeostasis by reducing the strength of excitatory synapses by specifically acting at the presynaptic level. Indeed, chronic hyperactivity triggers a REST-dependent decrease of the size of synaptic vesicle pools through the transcriptional and translational repression of specific presynaptic REST target genes. Together with our previous report, the data identify REST as a fundamental molecular player for neuronal homeostasis able to downscale simultaneously both intrinsic excitability and presynaptic efficiency in response to elevated neuronal activity. This experimental evidence adds new insights to the complex activity-dependent transcriptional regulation of the homeostatic plasticity processes mediated by REST.
Collapse
Affiliation(s)
- F Pecoraro-Bisogni
- Department of Experimental Medicine, Section of Physiology, University of Genova, Viale Benedetto XV 3, 16132, Genoa, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Gabriele Lignani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy. .,Institute of Neurology, University College of London, WC1N 3BG, London, UK.
| | - A Contestabile
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - E Castroflorio
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - D Pozzi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy.,Pharmacology and Brain Pathology Lab, Humanitas Clinical and Research Center, Humanitas University, Via Manzoni 56, Rozzano, Milan, Italy
| | - A Rocchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - C Prestigio
- Department of Experimental Medicine, Section of Physiology, University of Genova, Viale Benedetto XV 3, 16132, Genoa, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - M Orlando
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy.,Neurocure NWFZ, Charite Universitaetsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - P Valente
- Department of Experimental Medicine, Section of Physiology, University of Genova, Viale Benedetto XV 3, 16132, Genoa, Italy
| | - M Massacesi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy.,Laboratory of Neurosciences and Neurogenetics, Department of Head and Neck Diseases, "G. Gaslini" Institute, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - F Benfenati
- Department of Experimental Medicine, Section of Physiology, University of Genova, Viale Benedetto XV 3, 16132, Genoa, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, Section of Physiology, University of Genova, Viale Benedetto XV 3, 16132, Genoa, Italy. .,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
48
|
Schenck S, Kunz L, Sahlender D, Pardon E, Geertsma ER, Savtchouk I, Suzuki T, Neldner Y, Štefanić S, Steyaert J, Volterra A, Dutzler R. Generation and Characterization of Anti-VGLUT Nanobodies Acting as Inhibitors of Transport. Biochemistry 2017; 56:3962-3971. [DOI: 10.1021/acs.biochem.7b00436] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Stephan Schenck
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Laura Kunz
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniela Sahlender
- Department
of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Els Pardon
- VIB
Center for Structural Biology, VIB, 1050 Brussels, Belgium
- Structural
Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Eric R. Geertsma
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Institute
of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue-Straβe
9, 60438 Frankfurt
am Main, Germany
| | - Iaroslav Savtchouk
- Department
of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Toshiharu Suzuki
- Department
of Applied Chemistry, School of Engineering, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yvonne Neldner
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Saša Štefanić
- Institute
of Parasitology, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland
| | - Jan Steyaert
- VIB
Center for Structural Biology, VIB, 1050 Brussels, Belgium
- Structural
Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Andrea Volterra
- Department
of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Raimund Dutzler
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
49
|
Effect of high fat diet on phenotype, brain transcriptome and lipidome in Alzheimer's model mice. Sci Rep 2017; 7:4307. [PMID: 28655926 PMCID: PMC5487356 DOI: 10.1038/s41598-017-04412-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/15/2017] [Indexed: 02/01/2023] Open
Abstract
We examined the effect of chronic high fat diet (HFD) on amyloid deposition and cognition of 12-months old APP23 mice, and correlated the phenotype to brain transcriptome and lipidome. HFD significantly increased amyloid plaques and worsened cognitive performance compared to mice on normal diet (ND). RNA-seq results revealed that in HFD mice there was an increased expression of genes related to immune response, such as Trem2 and Tyrobp. We found a significant increase of TREM2 immunoreactivity in the cortex in response to HFD, most pronounced in female mice that correlated to the amyloid pathology. Down-regulated by HFD were genes related to neuron projections and synaptic transmission in agreement to the significantly deteriorated neurite morphology and cognition in these mice. To examine the effect of the diet on the brain lipidome, we performed Shotgun Lipidomics. While there was no difference in the total amounts of phospholipids of each class, we revealed that the levels of 24 lipid sub-species in the brain were significantly modulated by HFD. Network visualization of correlated lipids demonstrated overall imbalance with most prominent effect on cardiolipin molecular sub-species. This integrative approach demonstrates that HFD elicits a complex response at molecular, cellular and system levels in the CNS.
Collapse
|
50
|
Neocortical Chandelier Cells Developmentally Shape Axonal Arbors through Reorganization but Establish Subcellular Synapse Specificity without Refinement. eNeuro 2017; 4:eN-NWR-0057-17. [PMID: 28584877 PMCID: PMC5458751 DOI: 10.1523/eneuro.0057-17.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 11/21/2022] Open
Abstract
Diverse types of cortical interneurons (INs) mediate various kinds of inhibitory control mechanisms to balance and shape network activity. Distinct IN subtypes develop uniquely organized axonal arbors that innervate different subcellular compartments of excitatory principal neurons (PNs), which critically contribute to determining their output properties. However, it remains poorly understood how they establish this peculiar axonal organization and synaptic connectivity during development. Here, taking advantage of genetic labeling of IN progenitors, we examined developmental processes of axonal arbors and synaptic connections formed by murine chandelier cells (ChCs), which innervate axon initial segments (AISs) of PNs and thus powerfully regulate their spike generation. Our quantitative analysis by light microscopy revealed that ChCs overgrow and subsequently refine axonal branches as well as varicosities. Interestingly, we found that although a significant number of axonal varicosities are formed off AISs in addition to on AISs, presynaptic markers are predominantly colocalized with those on AISs throughout development. Immunoelectron microscopic (IEM) analysis also demonstrated that only varicosities apposed to AISs contain presynaptic profiles. These results suggest that subcellular synapse specificity of ChCs is genetically predetermined while axonal geometry is shaped through remodeling. Molecular cues localized at AISs may regulate target recognition and synapse formation by ChCs.
Collapse
|