1
|
Escamilla S, Sáez-Valero J, Cuchillo-Ibáñez I. NMDARs in Alzheimer's Disease: Between Synaptic and Extrasynaptic Membranes. Int J Mol Sci 2024; 25:10220. [PMID: 39337704 PMCID: PMC11431980 DOI: 10.3390/ijms251810220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are glutamate receptors with key roles in synaptic communication and plasticity. The activation of synaptic NMDARs initiates plasticity and stimulates cell survival. In contrast, the activation of extrasynaptic NMDARs can promote cell death underlying a potential mechanism of neurodegeneration occurring in Alzheimer's disease (AD). The distribution of synaptic versus extrasynaptic NMDARs has emerged as an important parameter contributing to neuronal dysfunction in neurodegenerative diseases including AD. Here, we review the concept of extrasynaptic NMDARs, as this population is present in numerous neuronal cell membranes but also in the membranes of various non-neuronal cells. Previous evidence regarding the membranal distribution of synaptic versus extrasynaptic NMDRs in relation to AD mice models and in the brains of AD patients will also be reviewed.
Collapse
Affiliation(s)
- Sergio Escamilla
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Javier Sáez-Valero
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Inmaculada Cuchillo-Ibáñez
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
2
|
Casillas Martinez A, Wicki-Stordeur LE, Ariano AV, Swayne LA. Dual role for pannexin 1 at synapses: regulating functional and morphological plasticity. J Physiol 2024. [PMID: 39264228 DOI: 10.1113/jp285228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
Pannexin 1 (PANX1) is an ion and metabolite membrane channel and scaffold protein enriched in synaptic compartments of neurons in the central nervous system. In addition to a well-established link between PANX1 and synaptic plasticity, we recently identified a role for PANX1 in the regulation of dendritic spine stability. Notably, PANX1 and its interacting proteins are linked to neurological conditions involving dendritic spine loss. Understanding the dual role of PANX1 in synaptic function and morphology may help to shed light on these links. We explore potential mechanisms, including PANX1's interactions with postsynaptic receptors and cytoskeleton regulating proteins. Finally, we contextualize PANX1's dual role within neurological diseases involving dendritic spine and synapse dysfunction.
Collapse
Affiliation(s)
| | - Leigh E Wicki-Stordeur
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Annika V Ariano
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
3
|
de Carvalho MB, Teixeira-Silva B, Marques SA, Silva AA, Cossenza M, da Cunha Faria-Melibeu A, Serfaty CA, Campello-Costa P. NMDA receptor remodeling and nNOS activation in mice after unilateral striatal injury with 6-OHDA. Heliyon 2024; 10:e34120. [PMID: 39130441 PMCID: PMC11315104 DOI: 10.1016/j.heliyon.2024.e34120] [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: 01/11/2024] [Revised: 06/06/2024] [Accepted: 07/03/2024] [Indexed: 08/13/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by selective dopaminergic loss. Non dopaminergic neurotransmitters such as glutamate are also involved in PD progression. NMDA receptor/postsynaptic density protein 95 (PSD-95)/neuronal nitric oxide synthase (nNOS) activation is involved in neuronal excitability in PD. Here, we are focusing on the evaluating these post-synaptic protein levels in the 6-OHDA model of PD. Adult male C57BL/6 mice subjected to unilateral striatal injury with 6-OHDA were assessed at 1-, 2-, or 4-weeks post-lesion. Animals were subjected to an apomorphine-induced rotation test followed by the analysis of protein content, synaptic structure, and NOx production. All biochemical analysis was performed comparing the control versus lesioned sides of the same animal. 6-OHDA mice exhibited contralateral rotation activity, difficulties in coordinating movements, and changes in Iba-1 and glial fibrillary acidic protein (GFAP) expression during the whole period. At one week of survival, the mice showed a shift in NMDA composition, favoring the GluN2A subunit and increased PSD95 and nNOS expression and NOx formation. After two-weeks, a decrease in the total number of synapses was observed in the lesioned side. However, the number of excitatory synapses was increased with a higher content of GluN1 subunit and PSD95. After four weeks, NMDA receptor subunits restored to control levels. Interestingly, NOx formation in the serum increased. This study reveals, for the first time, the temporal course of behavioral deficits and glutamatergic synaptic plasticity through NMDAr subunit shift. Together, these data demonstrate that dopamine depletion leads to a fine adaptive response over time, which can be used for further studies of therapeutic management adjustments with the progression of PD.
Collapse
Affiliation(s)
- Michele Barboza de Carvalho
- Laboratory of Neuroplasticity, Postgraduate Program in Neurosciences, Biology Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Bruna Teixeira-Silva
- Laboratory of Neuroplasticity, Postgraduate Program in Neurosciences, Biology Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Suelen Adriani Marques
- Laboratory of Neural Regeneration and Function, Department of Neurobiology, Federal Fluminense University, Niteroi, RJ, Brazil
- Postgraduate School in Pathological Anatomy, Federal University of the State of Rio de Janeiro, Brazil
| | - Andrea Alice Silva
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences (LAMAP), Graduate Program in Pathology, Faculty of Medicine, Universidade Federal Fluminense, Niterói, 24033-900, Rio de Janeiro, Brazil
| | - Marcelo Cossenza
- Laboratory of Molecular Pharmacology, Physiology and Pharmacology Department, Biomedical Institute, Postgraduate Program in Neurosciences, Biology Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Adriana da Cunha Faria-Melibeu
- Laboratory of Neurobiology of Development, Postgraduate Program in Neurosciences, Biology Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Claudio Alberto Serfaty
- Laboratory of Neural Plasticity, Postgraduate Program in Neurosciences, Biology Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Paula Campello-Costa
- Laboratory of Neuroplasticity, Postgraduate Program in Neurosciences, Biology Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| |
Collapse
|
4
|
Sun M, Zheng Q, Wang L, Wang R, Cui H, Zhang X, Xu C, Yin F, Yan H, Qiao X. Alcohol Consumption During Adolescence Alters the Cognitive Function in Adult Male Mice by Persistently Increasing Levels of DUSP6. Mol Neurobiol 2024; 61:3161-3178. [PMID: 37978157 DOI: 10.1007/s12035-023-03794-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023]
Abstract
Binge alcohol drinking during adolescence has long-term effects on the adult brain that alter brain structure and behaviors, but the underlying mechanisms remain poorly understood. Extracellular signal-regulated kinase (ERK) is involved in the synaptic plasticity and pathological brain injury by regulating the expression of cyclic adenosine monophosphate response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF). Dual-specificity phosphatase 6 (DUSP6) is a critical effector that dephosphorylates ERK1/2 to control the basal tone, amplitude, and duration of ERK signaling. To explore DUSP6 as a regulator of ERK signaling in the mPFC and its impact on long-term effects of alcohol, a male mouse model of adolescent intermittent alcohol (AIA) exposure was established. Behavioral experiments showed that AIA did not affect anxiety-like behavior or sociability in adulthood, but significantly damaged new object recognition and social recognition memory. Molecular studies further found that AIA reduced the levels of pERK-pCREB-BDNF-PSD95/NR2A involved in synaptic plasticity, while DUSP6 was significantly increased. Intra-mPFC infusion of AAV-DUSP6-shRNA restored the dendritic spine density and postsynaptic density thickness by reversing the level of p-ERK and its downstream molecular expression, and ultimately repaired adult cognitive impairment caused by chronic alcohol exposure during adolescence. These findings indicate that AIA exposure inhibits ERK-CREB-BDNF-PSD95/NR2A by increasing DUSP6 in the mPFC in adulthood that may be associated with long-lasting cognitive deficits.
Collapse
Affiliation(s)
- Mizhu Sun
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Qingmeng Zheng
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Lulu Wang
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Runzhi Wang
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Hengzhen Cui
- Basic Medicine, School of Medicine, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Xinlei Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Chen Xu
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Fangyuan Yin
- College of Forensic Science, School of Medicine, Xi'an Jiaotong University, No. 76, Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Hongtao Yan
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China
| | - Xiaomeng Qiao
- Department of Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
5
|
Nayan NM, Kadir SHSA, Husin A, Siran R. Neurodevelopmental effects of prenatal Bisphenol A exposure on the role of microRNA regulating NMDA receptor subunits in the male rat hippocampus. Physiol Behav 2024; 280:114546. [PMID: 38583549 DOI: 10.1016/j.physbeh.2024.114546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/09/2024]
Abstract
Maternal bisphenol A (BPA) exposure has been reported to cause learning and memory deficits in born offspring. However, little is known that this impairment is potentially caused by epigenetic modulation on the development of NMDA receptor subunits. This study investigates the effect of prenatal BPA exposure on the hippocampal miR-19a and miR-539, which are responsible for regulating NMDA receptor subunits as well as learning and memory functions. Pregnant Sprague Dawley rats were orally administered with 5 mg/kg/day of BPA from pregnancy day 1 (PD1) until gestation day 21 (GD21), while control mothers received no BPA. The mothers were observed daily until GD21 for either a cesarean section or spontaneous delivery. The male offspring were sacrificed when reaching GD21 (fetus), postnatal days 7, 14, 21 (PND7, 14, 21) and adolescent age 35 (AD35) where their hippocampi were dissected from the brain. The expression of targeted miR-19a, miR-539, GRIN2A, and GRIN2B were determined by qRT-PCR while the level of GluN2A and GluN2B were estimated by western blot. At AD35, the rats were assessed with neurobehavioral tests to evaluate their learning and memory function. The findings showed that prenatal BPA exposure at 5 mg/kg/day significantly reduces the expression of miR-19a, miR-539, GRIN2A, and GRIN2B genes in the male rat hippocampus at all ages. The level of GluN2A and GluN2B proteins is also significantly reduced when reaching adolescent age. Consequently, the rats showed spatial and fear memory impairments when reaching AD35. In conclusion, prenatal BPA exposure disrupts the role of miR-19a and miR-539 in regulating the NMDA receptor subunit in the hippocampus which may be one of the causes of memory and learning impairment in adolescent rats.
Collapse
Affiliation(s)
- Norazirah Mat Nayan
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Laboratory Animal Care Unit (LACU), Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Institute for Molecular Medicine and Biotechnology (IMMB) Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia
| | - Siti Hamimah Sheikh Abd Kadir
- Institute for Molecular Medicine and Biotechnology (IMMB) Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia
| | - Andrean Husin
- Faculty of Dentistry, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia
| | - Rosfaiizah Siran
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA 47000, Sungai Buloh, Selangor, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia..
| |
Collapse
|
6
|
Poole VN, Ridwan AR, Arfanakis K, Dawe RJ, Seyfried NT, De Jager PL, Schneider JA, Leurgans SE, Yu L, Bennett DA. Associations of brain morphology with cortical proteins of cognitive resilience. Neurobiol Aging 2024; 137:1-7. [PMID: 38394722 PMCID: PMC10949968 DOI: 10.1016/j.neurobiolaging.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024]
Abstract
In a recent proteome-wide study, we identified several candidate proteins for drug discovery whose cortical abundance was associated with cognitive resilience to late-life brain pathologies. This study examines the extent to which these proteins are associated with the brain structures of cognitive resilience in decedents from the Religious Orders Study and Memory and Aging Project. Six proteins were associated with brain morphometric characteristics related to higher resilience (i.e., larger anterior and medial temporal lobe volumes), and five were associated with morphometric characteristics related to lower resilience (i.e., enlarged ventricles). Two synaptic proteins, RPH3A and CPLX1, remained inversely associated with the lower resilience signature, after further controlling for 10 neuropathologic indices. These findings suggest preserved brain structure in periventricular regions as a potential mechanism by which RPH3A and CPLX1 are associated with cognitive resilience. Further work is needed to elucidate other mechanisms by which targeting these proteins can circumvent the effects of pathology on individuals at risk for cognitive decline.
Collapse
Affiliation(s)
- Victoria N Poole
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA.
| | - Abdur R Ridwan
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Robert J Dawe
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | | | - Philip L De Jager
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA; Cell Circuits Program, Broad Institute, Cambridge, MA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Sue E Leurgans
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Family and Preventive Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
7
|
Yakout DW, Shroff A, Wei W, Thaker V, Allen ZD, Sajish M, Nazarko TY, Mabb AM. Tau regulates Arc stability in neuronal dendrites via a proteasome-sensitive but ubiquitin-independent pathway. J Biol Chem 2024; 300:107237. [PMID: 38552740 PMCID: PMC11061231 DOI: 10.1016/j.jbc.2024.107237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 02/23/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
Tauopathies are neurodegenerative disorders characterized by the deposition of aggregates of the microtubule-associated protein tau, a main component of neurofibrillary tangles. Alzheimer's disease (AD) is the most common type of tauopathy and dementia, with amyloid-beta pathology as an additional hallmark feature of the disease. Besides its role in stabilizing microtubules, tau is localized at postsynaptic sites and can regulate synaptic plasticity. The activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that plays a key role in synaptic plasticity, learning, and memory. Arc has been implicated in AD pathogenesis and regulates the release of amyloid-beta. We found that decreased Arc levels correlate with AD status and disease severity. Importantly, Arc protein was upregulated in the hippocampus of Tau KO mice and dendrites of Tau KO primary hippocampal neurons. Overexpression of tau decreased Arc stability in an activity-dependent manner, exclusively in neuronal dendrites, which was coupled to an increase in the expression of dendritic and somatic surface GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. The tau-dependent decrease in Arc was found to be proteasome-sensitive, yet independent of Arc ubiquitination and required the endophilin-binding domain of Arc. Importantly, these effects on Arc stability and GluA1 localization were not observed in the commonly studied tau mutant, P301L. These observations provide a potential molecular basis for synaptic dysfunction mediated through the accumulation of tau in dendrites. Our findings confirm that Arc is misregulated in AD and further show a physiological role for tau in regulating Arc stability and AMPA receptor targeting.
Collapse
Affiliation(s)
- Dina W Yakout
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Ankit Shroff
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Wei Wei
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Vishrut Thaker
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Zachary D Allen
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Mathew Sajish
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Taras Y Nazarko
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Angela M Mabb
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
8
|
Kim HJ, Ko EA, Kwon OB, Jung SC. Prenatal treatment with corticosterone via maternal injection induces learning and memory impairments via delaying postsynaptic development in hippocampal CA1 neurons of rats. J Neurosci Res 2024; 102:e25323. [PMID: 38553948 DOI: 10.1002/jnr.25323] [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: 02/02/2023] [Revised: 02/16/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Previously, we reported that prenatal exposure to high corticosterone induced attention-deficit hyperactivity disorder (ADHD)-like behaviors with cognitive deficits after weaning. In the present study, cellular mechanisms underlying cortisol-induced cognitive dysfunction were investigated using rat pups (Corti.Pups) born from rat mothers that were repetitively injected with corticosterone during pregnancy. In results, Corti.Pups exhibited the failure of behavioral memory formation in the Morris water maze (MWM) test and the incomplete long-term potentiation (LTP) of hippocampal CA1 neurons. Additionally, glutamatergic excitatory postsynaptic currents (EPSCs) were remarkably suppressed in Corti.Pups compared to normal rat pups. Incomplete LTP and weaker EPSCs in Corti.Pups were attributed to the delayed postsynaptic development of CA1 neurons, showing a higher expression of NR2B subunits and lower expression of PSD-95 and BDNF. These results indicated that the prenatal treatment with corticosterone to elevate cortisol level might potently downregulate the BDNF-mediated signaling critical for the synaptic development of hippocampal CA1 neurons during brain development, and subsequently, induce learning and memory impairment. Our findings suggest a possibility that the prenatal dysregulation of cortisol triggers the epigenetic pathogenesis of neurodevelopmental psychiatric disorders, such as ADHD and autism.
Collapse
Affiliation(s)
- Hye-Ji Kim
- Department of Physiology, College of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Eun-A Ko
- Department of Physiology, College of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Oh-Bin Kwon
- New Drug Development Center, Kmedihub, Daegu, Republic of Korea
| | - Sung-Cherl Jung
- Department of Physiology, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Institute of Medical Science, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
9
|
González-Arnay E, Pérez-Santos I, Jiménez-Sánchez L, Cid E, Gal B, de la Prida LM, Cavada C. Immunohistochemical field parcellation of the human hippocampus along its antero-posterior axis. Brain Struct Funct 2024; 229:359-385. [PMID: 38180568 PMCID: PMC10917878 DOI: 10.1007/s00429-023-02725-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 10/15/2023] [Indexed: 01/06/2024]
Abstract
The primate hippocampus includes the dentate gyrus, cornu ammonis (CA), and subiculum. CA is subdivided into four fields (CA1-CA3, plus CA3h/hilus of the dentate gyrus) with specific pyramidal cell morphology and connections. Work in non-human mammals has shown that hippocampal connectivity is precisely patterned both in the laminar and longitudinal axes. One of the main handicaps in the study of neuropathological semiology in the human hippocampus is the lack of clear laminar and longitudinal borders. The aim of this study was to explore a histochemical segmentation of the adult human hippocampus, integrating field (medio-lateral), laminar, and anteroposterior longitudinal patterning. We provide criteria for head-body-tail field and subfield parcellation of the human hippocampus based on immunodetection of Rabphilin3a (Rph3a), Purkinje-cell protein 4 (PCP4), Chromogranin A and Regulation of G protein signaling-14 (RGS-14). Notably, Rph3a and PCP4 allow to identify the border between CA3 and CA2, while Chromogranin A and RGS-14 give specific staining of CA2. We also provide novel histological data about the composition of human-specific regions of the anterior and posterior hippocampus. The data are given with stereotaxic coordinates along the longitudinal axis. This study provides novel insights for a detailed region-specific parcellation of the human hippocampus useful for human brain imaging and neuropathology.
Collapse
Affiliation(s)
- Emilio González-Arnay
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Basic Medical Science-Division of Human Anatomy, Universidad de La Laguna, Santa Cruz de Tenerife, Canary Islands, Spain
| | - Isabel Pérez-Santos
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lorena Jiménez-Sánchez
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Elena Cid
- Instituto Cajal, CSIC, Madrid, Spain
| | - Beatriz Gal
- Instituto Cajal, CSIC, Madrid, Spain
- Universidad CEU-San Pablo, Madrid, Spain
| | | | - Carmen Cavada
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
10
|
Irie K, Ohta KI, Ujihara H, Araki C, Honda K, Suzuki S, Warita K, Otabi H, Kumei H, Nakamura S, Koyano K, Miki T, Kusaka T. An enriched environment ameliorates the reduction of parvalbumin-positive interneurons in the medial prefrontal cortex caused by maternal separation early in life. Front Neurosci 2024; 17:1308368. [PMID: 38292903 PMCID: PMC10825025 DOI: 10.3389/fnins.2023.1308368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/11/2023] [Indexed: 02/01/2024] Open
Abstract
Early child maltreatment, such as child abuse and neglect, is well known to affect the development of social skills. However, the mechanisms by which such an adverse environment interrupts the development of social skills remain unelucidated. Identifying the period and brain regions that are susceptible to adverse environments can lead to appropriate developmental care later in life. We recently reported an excitatory/inhibitory imbalance and low activity during social behavior in the medial prefrontal cortex (mPFC) of the maternal separation (MS) animal model of early life neglect after maturation. Based on these results, in the present study, we investigated how MS disturbs factors related to excitatory and inhibitory neurons in the mPFC until the critical period of mPFC development. Additionally, we evaluated whether the effects of MS could be recovered in an enriched environment after MS exposure. Rat pups were separated from their dams on postnatal days (PDs) 2-20 (twice daily, 3 h each) and compared with the mother-reared control (MRC) group. Gene expression analysis revealed that various factors related to excitatory and inhibitory neurons were transiently disturbed in the mPFC during MS. A similar tendency was found in the sensory cortex; however, decreased parvalbumin (PV) expression persisted until PD 35 only in the mPFC. Moreover, the number of PV+ interneurons decreased in the ventromedial prefrontal cortex (vmPFC) on PD 35 in the MS group. Additionally, perineural net formation surrounding PV+ interneurons, which is an indicator of maturity and critical period closure, was unchanged, indicating that the decreased PV+ interneurons were not simply attributable to developmental delay. This reduction of PV+ interneurons improved to the level observed in the MRC group by the enriched environment from PD 21 after the MS period. These results suggest that an early adverse environment disturbs the development of the mPFC but that these abnormalities allow room for recovery depending on the subsequent environment. Considering that PV+ interneurons in the mPFC play an important role in social skills such as empathy, an early rearing environment is likely a very important factor in the subsequent acquisition of social skills.
Collapse
Affiliation(s)
- Kanako Irie
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Ken-ichi Ohta
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hidetoshi Ujihara
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Chihiro Araki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kodai Honda
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Shingo Suzuki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Katsuhiko Warita
- Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Hikari Otabi
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Haruki Kumei
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takanori Miki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
11
|
Wei RM, Zhang YM, Zhang KX, Liu GX, Li XY, Zhang JY, Lun WZ, Liu XC, Chen GH. An enriched environment ameliorates maternal sleep deprivation-induced cognitive impairment in aged mice by improving mitochondrial function via the Sirt1/PGC-1α pathway. Aging (Albany NY) 2024; 16:1128-1144. [PMID: 38231482 PMCID: PMC10866428 DOI: 10.18632/aging.205385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/16/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Early life stress can cause cognitive impairment in aged offspring. Environmental enrichment (EE) is considered to be an effective non-pharmacological treatment for improving cognitive decline. The aim of this research was to evaluate the effect of EE, on cognitive impairment in aged offspring induced by maternal sleep deprivation (MSD) and the underlying mechanisms involved to investigate its potential value in clinical practice. METHODS CD-1 damns were subjected or not to sleep deprivation during late gestation. Twenty-one days after birth, the offspring were assigned to standard or EE cages. At 18 months-old, the learning and memory function of the offspring mice was evaluated using Morris water maze. The hippocampal and prefrontal cortical levels of protein, gene, proinflammation cytokines, and oxidative stress indicators was examined by Western blot, real-time polymerase chain reaction, enzyme linked immunosorbent assay, and biochemical assays. RESULTS Offspring in MSD group exhibited declined learning and memory abilities compared with control animals. Moreover, the hippocampal and prefrontal cortical levels of Sirtuin1 (Sirt1), peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α), postsynaptic density protein-95, and synaptophysin were lower and those of proinflammation cytokines higher in the MSD group; meanwhile, the superoxide dismutase content was higher and the malondialdehyde and reactive oxygen species contents were lower. However, these deleterious changes were ameliorated by exposure to EE. CONCLUSIONS EE attenuates MSD-induced cognitive impairment, oxidative stress, and neuroinflammation and reverses the reduction in synaptic protein levels in aged offspring mice via the Sirt1/PGC-1α pathway.
Collapse
Affiliation(s)
- Ru-Meng Wei
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| | - Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| | - Kai-Xuan Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| | - Gao-Xia Liu
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| | - Xue-Yan Li
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| | - Jing-Ya Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| | - Wei-Zhong Lun
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| | - Xue-Chun Liu
- Department of Neurology, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei 230011, Anhui, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei 238001, Anhui, China
| |
Collapse
|
12
|
Pavinato L, Stanic J, Barzasi M, Gurgone A, Chiantia G, Cipriani V, Eberini I, Palazzolo L, Di Luca M, Costa A, Marcantoni A, Biamino E, Spada M, Hiatt SM, Kelley WV, Vestito L, Sisodiya SM, Efthymiou S, Chand P, Kaiyrzhanov R, Bruselles A, Cardaropoli S, Tartaglia M, De Rubeis S, Buxbaum JD, Smedley D, Ferrero GB, Giustetto M, Gardoni F, Brusco A. Missense variants in RPH3A cause defects in excitatory synaptic function and are associated with a clinically variable neurodevelopmental disorder. Genet Med 2023; 25:100922. [PMID: 37403762 DOI: 10.1016/j.gim.2023.100922] [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: 10/19/2022] [Revised: 06/22/2023] [Accepted: 06/25/2023] [Indexed: 07/06/2023] Open
Abstract
PURPOSE RPH3A encodes a protein involved in the stabilization of GluN2A subunit of N-methyl-D-aspartate (NMDA)-type glutamate receptors at the cell surface, forming a complex essential for synaptic plasticity and cognition. We investigated the effect of variants in RPH3A in patients with neurodevelopmental disorders. METHODS By using trio-based exome sequencing, GeneMatcher, and screening of 100,000 Genomes Project data, we identified 6 heterozygous variants in RPH3A. In silico and in vitro models, including rat hippocampal neuronal cultures, have been used to characterize the effect of the variants. RESULTS Four cases had a neurodevelopmental disorder with untreatable epileptic seizures [p.(Gln73His)dn; p.(Arg209Lys); p.(Thr450Ser)dn; p.(Gln508His)], and 2 cases [p.(Arg235Ser); p.(Asn618Ser)dn] showed high-functioning autism spectrum disorder. Using neuronal cultures, we demonstrated that p.(Thr450Ser) and p.(Asn618Ser) reduce the synaptic localization of GluN2A; p.(Thr450Ser) also increased the surface levels of GluN2A. Electrophysiological recordings showed increased GluN2A-dependent NMDA ionotropic glutamate receptor currents for both variants and alteration of postsynaptic calcium levels. Finally, expression of the Rph3AThr450Ser variant in neurons affected dendritic spine morphology. CONCLUSION Overall, we provide evidence that missense gain-of-function variants in RPH3A increase GluN2A-containing NMDA ionotropic glutamate receptors at extrasynaptic sites, altering synaptic function and leading to a clinically variable neurodevelopmental presentation ranging from untreatable epilepsy to autism spectrum disorder.
Collapse
Affiliation(s)
- Lisa Pavinato
- Department of Medical Sciences, University of Turin, Turin, Italy; Institute of Oncology Research (IOR), Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland
| | - Jennifer Stanic
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Marta Barzasi
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Antonia Gurgone
- Department of Neuroscience, University of Turin, Turin, Italy
| | | | - Valentina Cipriani
- William Harvey Research Institute, Clinical Pharmacology Precision Medicine, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Ivano Eberini
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Luca Palazzolo
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Alex Costa
- Department of Biosciences, University of the Studies of Milan, Milan, Italy; Institute of Biophysics, Consiglio Nazionale delle Ricerche (CNR), Milan, Italy
| | - Andrea Marcantoni
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Elisa Biamino
- Department of Pediatrics, Regina Margherita Children Hospital, Turin, Italy
| | - Marco Spada
- Department of Pediatrics, Regina Margherita Children Hospital, Turin, Italy
| | - Susan M Hiatt
- HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | | | - Letizia Vestito
- William Harvey Research Institute, Clinical Pharmacology Precision Medicine, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom; Chalfont Centre for Epilepsy Bucks, Chalfont St Peter, United Kingdom
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Prem Chand
- Department of Paediatric and Child Health, Aga Khan University Hospital, Karachi, Pakistan
| | - Rauan Kaiyrzhanov
- University College London, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Cardaropoli
- Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Damian Smedley
- William Harvey Research Institute, Clinical Pharmacology Precision Medicine, Queen Mary University of London, Charterhouse Square, United Kingdom
| | | | | | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Turin, Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy.
| |
Collapse
|
13
|
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
|
14
|
Saadati H, Ghaheri S, Sadegzadeh F, Sakhaie N, Abdollahzadeh M. Beneficial effects of enriched environment on behavior, cognitive functions, and hippocampal brain-derived neurotrophic factor level following postnatal serotonin depletion in male rats. Int J Dev Neurosci 2023; 83:67-79. [PMID: 36342785 DOI: 10.1002/jdn.10238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/04/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
The neurotransmitter serotonin (5-HT) is one of the most important modulators of neural circuitry and has a critical role in neural development and functions. Previous studies indicated that changes in serotonergic system signaling in early life critically impact mental health, behavior, the morphology of hippocampal neurons, and cognitive functions across the lifespan. The enriched environment (EE) has indicated beneficial effects on behavior and cognitive functions in the developmental period of life, but its impacts on cognitive impairments and behavioral changes following postnatal serotonin depletion are unknown. Therefore, the present study aimed to evaluate the influences of the EE housing (postnatal days [PNDs] 21-60) following postnatal serotonin depletion (by para-chlorophenylalanine [PCPA], 100 mg/kg, s.c, in PNDs 10-20) on anxiety-related behaviors, cognitive functions, and brain-derived neurotrophic factor (BDNF) mRNA expression in the hippocampus of male rats. Memory and behavioral parameters were examined in early adulthood and after that, the hippocampi of rats were removed to determine the BDNF mRNA expression by PCR (PNDs 60-70). The findings of the present work indicated that adolescent EE exposure alleviated memory impairment, decreased BDNF levels, and anxiety disorders induced by experimental depletion of serotonin. Overall, these results indicate that serotonergic system dysregulation during the developmental periods can be alleviated by adolescent EE exposure.
Collapse
Affiliation(s)
- Hakimeh Saadati
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Safa Ghaheri
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farshid Sadegzadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nona Sakhaie
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Abdollahzadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
15
|
Yang L, Wei M, Wang Y, Zhang J, Liu S, Liu M, Wang S, Li K, Dong Z, Zhang C. Rabphilin-3A undergoes phase separation to regulate GluN2A mobility and surface clustering. Nat Commun 2023; 14:379. [PMID: 36693856 PMCID: PMC9873702 DOI: 10.1038/s41467-023-36046-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are essential for excitatory neurotransmission and synaptic plasticity. GluN2A and GluN2B, two predominant Glu2N subunits of NMDARs in the hippocampus and the cortex, display distinct clustered distribution patterns and mobility at synaptic and extrasynaptic sites. However, how GluN2A clusters are specifically organized and stabilized remains poorly understood. Here, we found that the previously reported GluN2A-specific binding partner Rabphilin-3A (Rph3A) has the ability to undergo phase separation, which relies on arginine residues in its N-terminal domain. Rph3A phase separation promotes GluN2A clustering by binding GluN2A's C-terminal domain. A complex formed by Rph3A, GluN2A, and the scaffolding protein PSD95 promoted Rph3A phase separation. Disrupting Rph3A's phase separation suppressed the synaptic and extrasynaptic surface clustering, synaptic localization, stability, and synaptic response of GluN2A in hippocampal neurons. Together, our results reveal the critical role of Rph3A phase separation in determining the organization and stability of GluN2A in the neuronal surface.
Collapse
Affiliation(s)
- Lei Yang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Yangzhen Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jingtao Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Sen Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Mengna Liu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Shanshan Wang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Ke Li
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zhaoqi Dong
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China. .,Chinese Institute for Brain Research, Beijing, 102206, China. .,State Key Laboratory of Translational Medicine and Innovative Drug Development, Nanjing, 210000, Jiangsu, China. .,Beijing Laboratory of Oral Health, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
16
|
Angelini C, Morellato A, Alfieri A, Pavinato L, Cravero T, Bianciotto OT, Salemme V, Natalini D, Centonze G, Raspanti A, Garofalo T, Valdembri D, Serini G, Marcantoni A, Becchetti A, Giustetto M, Turco E, Defilippi P. p140Cap Regulates the Composition and Localization of the NMDAR Complex in Synaptic Lipid Rafts. J Neurosci 2022; 42:7183-7200. [PMID: 35953295 PMCID: PMC9512579 DOI: 10.1523/jneurosci.1775-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 06/07/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022] Open
Abstract
The NMDARs are key players in both physiological and pathologic synaptic plasticity because of their involvement in many aspects of neuronal transmission as well as learning and memory. The contribution in these events of different types of GluN2A-interacting proteins is still unclear. The p140Cap scaffold protein acts as a hub for postsynaptic complexes relevant to psychiatric and neurologic disorders and regulates synaptic functions, such as the stabilization of mature dendritic spine, memory consolidation, LTP, and LTD. Here we demonstrate that p140Cap directly binds the GluN2A subunit of NMDAR and modulates GluN2A-associated molecular network. Indeed, in p140Cap KO male mice, GluN2A is less associated with PSD95 both in ex vivo synaptosomes and in cultured hippocampal neurons, and p140Cap expression in KO neurons can rescue GluN2A and PSD95 colocalization. p140Cap is crucial in the recruitment of GluN2A-containing NMDARs and, consequently, in regulating NMDARs' intrinsic properties. p140Cap is associated to synaptic lipid-raft (LR) and to soluble postsynaptic membranes, and GluN2A and PSD95 are less recruited into synaptic LR of p140Cap KO male mice. Gated-stimulated emission depletion microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in LR in an activity-dependent fashion. In the synaptic compartment, p140Cap influences the association between GluN2A and PSD95 and modulates GluN2A enrichment into LR. Overall, such increase in these membrane domains rich in signaling molecules results in improved signal transduction efficiency.SIGNIFICANCE STATEMENT Here we originally show that the adaptor protein p140Cap directly binds the GluN2A subunit of NMDAR and modulates the GluN2A-associated molecular network. Moreover, we show, for the first time, that p140Cap also associates to synaptic lipid rafts and controls the selective recruitment of GluN2A and PSD95 to this specific compartment. Finally, gated-stimulated emission depletion microscopy on hippocampal neurons confirmed that p140Cap is required for embedding GluN2A clusters in lipid rafts in an activity-dependent fashion. Overall, our findings provide the molecular and functional dissection of p140Cap as a new active member of a highly dynamic synaptic network involved in memory consolidation, LTP, and LTD, which are known to be altered in neurologic and psychiatric disorders.
Collapse
Affiliation(s)
- Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Annalisa Alfieri
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Lisa Pavinato
- Department of Medical Sciences, Medical Genetics Unit, University of Torino, Torino, 10126, Italy
| | - Tiziana Cravero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Olga Teresa Bianciotto
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Alessandra Raspanti
- Neuroscience Department "Rita Levi Montalcini," University of Torino, Torino, 10125, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, Sapienza University, Roma, 00161, Italy
| | - Donatella Valdembri
- Department of Oncology, University of Torino School of Medicine, Regione Gonzole, 10, 10043, Orbassano, TO, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, 10060, Italy
| | - Guido Serini
- Department of Oncology, University of Torino School of Medicine, Regione Gonzole, 10, 10043, Orbassano, TO, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, 10060, Italy
| | - Andrea Marcantoni
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, University of Torino, Torino, 10126, Italy
- Department of Biotechnology and Biosciences and NeuroMI, University of Milano-Bicocca, Milano, 20126, Italy
| | - Andrea Becchetti
- Department of Biotechnology and Biosciences and NeuroMI, University of Milano-Bicocca, Milano, 20126, Italy
| | - Maurizio Giustetto
- Neuroscience Department "Rita Levi Montalcini," University of Torino, Torino, 10125, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, 10126, Italy
| |
Collapse
|
17
|
Prikas E, Paric E, Asih PR, Stefanoska K, Stefen H, Fath T, Poljak A, Ittner A. Tau target identification reveals NSF-dependent effects on AMPA receptor trafficking and memory formation. EMBO J 2022; 41:e10242. [PMID: 35993331 PMCID: PMC9475529 DOI: 10.15252/embj.2021110242] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 11/09/2022] Open
Abstract
Microtubule-associated protein tau is a central factor in Alzheimer's disease and other tauopathies. However, the physiological functions of tau are unclear. Here, we used proximity-labelling proteomics to chart tau interactomes in primary neurons and mouse brains in vivo. Tau interactors map onto pathways of cytoskeletal, synaptic vesicle and postsynaptic receptor regulation and show significant enrichment for Parkinson's, Alzheimer's and prion disease. We find that tau interacts with and dose-dependently reduces the activity of N-ethylmaleimide sensitive fusion protein (NSF), a vesicular ATPase essential for AMPA-type glutamate receptor (AMPAR) trafficking. Tau-deficient (tau-/- ) neurons showed mislocalised expression of NSF and enhanced synaptic AMPAR surface levels, reversible through the expression of human tau or inhibition of NSF. Consequently, enhanced AMPAR-mediated associative and object recognition memory in tau-/- mice is suppressed by both hippocampal tau and infusion with an NSF-inhibiting peptide. Pathologic mutant tau from mouse models or Alzheimer's disease significantly enhances NSF inhibition. Our results map neuronal tau interactomes and delineate a functional link of tau with NSF in plasticity-associated AMPAR-trafficking and memory.
Collapse
Affiliation(s)
- Emmanuel Prikas
- Flinders Health & Medical Research Institute, College of Medicine and Public HealthFlinders UniversityAdelaideSAAustralia
| | - Esmeralda Paric
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNSWAustralia
| | - Prita R Asih
- Flinders Health & Medical Research Institute, College of Medicine and Public HealthFlinders UniversityAdelaideSAAustralia
| | - Kristie Stefanoska
- Flinders Health & Medical Research Institute, College of Medicine and Public HealthFlinders UniversityAdelaideSAAustralia
| | - Holly Stefen
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNSWAustralia
| | - Thomas Fath
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNSWAustralia
| | - Anne Poljak
- Mark Wainwright Analytical CentreUniversity of New South WalesSydneyNSWAustralia
| | - Arne Ittner
- Flinders Health & Medical Research Institute, College of Medicine and Public HealthFlinders UniversityAdelaideSAAustralia
| |
Collapse
|
18
|
Identification and Validation of Prognostic Markers for Lung Squamous Cell Carcinoma Associated with Chronic Obstructive Pulmonary Disease. JOURNAL OF ONCOLOGY 2022; 2022:4254195. [PMID: 36035311 PMCID: PMC9402374 DOI: 10.1155/2022/4254195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/28/2022] [Accepted: 07/03/2022] [Indexed: 12/04/2022]
Abstract
Background Globally, the incidence and associated mortality of chronic obstructive pulmonary disease (COPD) and lung carcinoma are showing a worsening trend. There is increasing evidence that COPD is an independent risk factor for the occurrence and progression of lung carcinoma. This study aimed to identify and validate the gene signatures associated with COPD, which may serve as potential new biomarkers for the prediction of prognosis in patients with lung carcinoma. Methods A total of 111 COPD patient samples and 40 control samples were obtained from the GSE76925 cohort, and a total of 4933 genes were included in the study. The weighted gene coexpression network analysis (WGCNA) was performed to identify the modular genes that were significantly associated with COPD. The KEGG pathway and GO functional enrichment analyses were also performed. The RNAseq and clinicopathological data of 490 lung squamous cell carcinoma patients were obtained from the TCGA database. Further, univariate Cox regression and Lasso analyses were performed to screen for marker genes and construct a survival analysis model. Finally, the Human Protein Atlas (HPA) database was used to assess the gene expression in normal and tumor tissues of the lungs. Results A 6-gene signature (DVL1, MRPL4, NRTN, NSUN3, RPH3A, and SNX32) was identified based on the Cox proportional risk analysis to construct the prognostic RiskScore survival model associated with COPD. Kaplan–Meier survival analysis indicated that the model could significantly differentiate between the prognoses of patients with lung carcinoma, wherein higher RiskScore samples were associated with a worse prognosis. Additionally, the model had a good predictive performance and reliability, as indicated by a high AUC, and these were validated in both internal and external sets. The 6-gene signature had a good predictive ability across clinical signs and could be considered an independent factor of prognostic risk. Finally, the protein expressions of the six genes were analyzed based on the HPA database. The expressions of DVL1, MRPL4, and NSUN3 were relatively higher, while that of RPH3A was relatively lower in the tumor tissues. The expression of SNX32 was high in both the tumor and paracarcinoma tissues. Results of the analyses using TCGA and GSE31446 databases were consistent with the expressions reported in the HPA database. Conclusion Novel COPD-associated gene markers for lung carcinoma were identified and validated in this study. The genes may be considered potential biomarkers to evaluate the prognostic risk of patients with lung carcinoma. Furthermore, some of these genes may have implications as new therapeutic targets and can be used to guide clinical applications.
Collapse
|
19
|
Ferrari E, Scheggia D, Zianni E, Italia M, Brumana M, Palazzolo L, Parravicini C, Pilotto A, Padovani A, Marcello E, Eberini I, Calabresi P, Diluca M, Gardoni F. Rabphilin-3A as a Novel Target to Reverse α-synuclein-induced Synaptic Loss in Parkinson's Disease. Pharmacol Res 2022; 183:106375. [PMID: 35918045 DOI: 10.1016/j.phrs.2022.106375] [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: 07/04/2022] [Revised: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 10/16/2022]
Abstract
Toxic aggregates of α-synuclein (αsyn) are considered key drivers of Parkinson's disease (PD) pathology. In early PD, αsyn induces synaptic dysfunction also modulating the glutamatergic neurotransmission. However, a more detailed understanding of the molecular mechanisms underlying αsyn-triggered synaptic failure is required to design novel therapeutic interventions. Here, we described the role of Rabphilin-3A (Rph3A) as novel target to counteract αsyn-induced synaptic loss in PD. Rph3A is a synaptic protein interacting with αsyn and involved in stabilizing dendritic spines and in promoting the synaptic retention of NMDA-type glutamate receptors. We found that in vivo intrastriatal injection of αsyn-preformed fibrils in mice induces the early loss of striatal synapses associated with decreased synaptic levels of Rph3A and impaired Rph3A/NMDA receptors interaction. Modulating Rph3A striatal expression or interfering with the Rph3A/αsyn complex with a small molecule prevented dendritic spine loss and rescued associated early motor defects in αsyn-injected mice. Notably, the same experimental approaches prevented αsyn-induced synaptic loss in vitro in primary hippocampal neurons. Overall, these findings indicate that approaches aimed at restoring Rph3A synaptic functions can slow down the early synaptic detrimental effects of αsyn aggregates in PD.
Collapse
Affiliation(s)
- Elena Ferrari
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Diego Scheggia
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Elisa Zianni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Maria Italia
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Marta Brumana
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Luca Palazzolo
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Chiara Parravicini
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy.
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy.
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Ivano Eberini
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Paolo Calabresi
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy; Clinica Neurologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| | - Monica Diluca
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy.
| |
Collapse
|
20
|
Zhu X, Li H, You W, Yu Z, Wang Z, Shen H, Li X, Yu H, Wang Z, Chen G. Role of Rph3A in brain injury induced by experimental cerebral ischemia-reperfusion model in rats. CNS Neurosci Ther 2022; 28:1124-1138. [PMID: 35467084 PMCID: PMC9160444 DOI: 10.1111/cns.13850] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 01/01/2023] Open
Abstract
Aim The aim was to study the role of Rph3A in neuronal injury induced by cerebral ischemia‐reperfusion. Methods The protein and mRNA levels of Rph3A in penumbra were detected by Western blot. The localization of Rph3A in different cell types in penumbra was detected by immunofluorescence. Apoptosis in the brain was detected by TUNEL staining. We tested neurobehavioral evaluation using rotarod test, adhesive‐removal test, and Morris Water maze test. We examined the expression and localization of Rph3A in cultured neurons and astrocytes in vitro by Western blot and ELISA, respectively. Results The mRNA and protein levels of Rph3A had significantly increased in brain penumbra of the rat MCAO/R model. Rph3A was mainly distributed in neurons and astrocytes and was significantly increased by MCAO/R. We downregulated Rph3A and found that it further worsened the cerebral infarct, neuronal death and behavioral, cognitive, and memory impairments in rats after MCAO/R. We also found that ischemia‐reperfusion upregulated the in vitro protein level and secretion of Rph3A in astrocytes but led to a decrease in the protein level of Rph3A in neurons. Conclusion The increase in Rph3A in the brain penumbra may be an endogenous protective mechanism against ischemia‐reperfusion injury, which is mainly dominated by astrocytes.
Collapse
Affiliation(s)
- Xianlong Zhu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China.,Department of Neurosurgery, The Second People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Wanchun You
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Hao Yu
- Department of Neurosurgery, The First People's Hospital of Nantong city, Nantong, Jiangsu, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Institute of Stroke Research, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
21
|
Franchini L, Stanic J, Barzasi M, Zianni E, Mauceri D, Diluca M, Gardoni F. Rabphilin-3A Drives Structural Modifications of Dendritic Spines Induced by Long-Term Potentiation. Cells 2022; 11:1616. [PMID: 35626653 PMCID: PMC9139176 DOI: 10.3390/cells11101616] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/01/2022] [Accepted: 05/10/2022] [Indexed: 01/09/2023] Open
Abstract
The interaction of Rabphilin-3A (Rph3A) with the NMDA receptor (NMDAR) in hippocampal neurons plays a pivotal role in the synaptic retention of this receptor. The formation of a Rph3A/NMDAR complex is needed for the induction of long-term potentiation and NMDAR-dependent hippocampal behaviors, such as spatial learning. Moreover, Rph3A can also interact with AMPA receptors (AMPARs) through the formation of a complex with myosin Va. Here, we used a confocal imaging approach to show that Rph3A overexpression in primary hippocampal neuronal cultures is sufficient to promote increased dendritic spine density. This morphological event is correlated with an increase in GluN2A-containing NMDARs at synaptic membranes and a decrease in the surface levels of GluA1-containing AMPARs. These molecular and morphological modifications of dendritic spines are sufficient to occlude the spine formation induced by long-term potentiation, but do not prevent the spine loss induced by long-term depression. Overall, our results demonstrate a key role for Rph3A in the modulation of structural synaptic plasticity at hippocampal synapses that correlates with its interactions with both NMDARs and AMPARs.
Collapse
Affiliation(s)
- Luca Franchini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.F.); (J.S.); (M.B.); (E.Z.); (M.D.)
| | - Jennifer Stanic
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.F.); (J.S.); (M.B.); (E.Z.); (M.D.)
| | - Marta Barzasi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.F.); (J.S.); (M.B.); (E.Z.); (M.D.)
| | - Elisa Zianni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.F.); (J.S.); (M.B.); (E.Z.); (M.D.)
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 366, 69120 Heidelberg, Germany;
| | - Monica Diluca
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.F.); (J.S.); (M.B.); (E.Z.); (M.D.)
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (L.F.); (J.S.); (M.B.); (E.Z.); (M.D.)
| |
Collapse
|
22
|
Chen T, Mandal A, Zhu H, Liu R. Imaging Genetic Based Mediation Analysis for Human Cognition. Front Neurosci 2022; 16:824069. [PMID: 35573299 PMCID: PMC9097855 DOI: 10.3389/fnins.2022.824069] [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: 11/28/2021] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
The brain connectome maps the structural and functional connectivity that forms an important neurobiological basis for the analysis of human cognitive traits while the genetic predisposition and our cognition ability are frequently found in close association. The issue of how genetic architecture and brain connectome causally affect human behaviors remains unknown. To seek for the potential causal relationship, in this paper, we carried out the causal pathway analysis from single nucleotide polymorphism (SNP) data to four common human cognitive traits, mediated by the brain connectome. Specifically, we selected 942 SNPs that are significantly associated with the brain connectome, and then estimated the direct and indirect effect on the human traits for each SNP. We found out that a majority of the selected SNPs have significant direct effects on human traits and discussed the trait-related brain regions and their implications.
Collapse
Affiliation(s)
- Tingan Chen
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Abhishek Mandal
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rongjie Liu
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
23
|
McGrath T, Baskerville R, Rogero M, Castell L. Emerging Evidence for the Widespread Role of Glutamatergic Dysfunction in Neuropsychiatric Diseases. Nutrients 2022; 14:nu14050917. [PMID: 35267893 PMCID: PMC8912368 DOI: 10.3390/nu14050917] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
The monoamine model of depression has long formed the basis of drug development but fails to explain treatment resistance or associations with stress or inflammation. Recent animal research, clinical trials of ketamine (a glutamate receptor antagonist), neuroimaging research, and microbiome studies provide increasing evidence of glutamatergic dysfunction in depression and other disorders. Glutamatergic involvement across diverse neuropathologies including psychoses, neurodevelopmental, neurodegenerative conditions, and brain injury forms the rationale for this review. Glutamate is the brain's principal excitatory neurotransmitter (NT), a metabolic and synthesis substrate, and an immune mediator. These overlapping roles and multiple glutamate NT receptor types complicate research into glutamate neurotransmission. The glutamate microcircuit comprises excitatory glutamatergic neurons, astrocytes controlling synaptic space levels, through glutamate reuptake, and inhibitory GABA interneurons. Astroglia generate and respond to inflammatory mediators. Glutamatergic microcircuits also act at the brain/body interface via the microbiome, kynurenine pathway, and hypothalamus-pituitary-adrenal axis. Disruption of excitatory/inhibitory homeostasis causing neuro-excitotoxicity, with neuronal impairment, causes depression and cognition symptoms via limbic and prefrontal regions, respectively. Persistent dysfunction reduces neuronal plasticity and growth causing neuronal death and tissue atrophy in neurodegenerative diseases. A conceptual overview of brain glutamatergic activity and peripheral interfacing is presented, including the common mechanisms that diverse diseases share when glutamate homeostasis is disrupted.
Collapse
Affiliation(s)
- Thomas McGrath
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| | - Richard Baskerville
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
- Correspondence:
| | - Marcelo Rogero
- School of Public Health, University of Sao Paulo, Sao Paulo 01246-904, Brazil;
| | - Linda Castell
- Green Templeton College, University of Oxford, Oxford OX2 6HG, UK; (T.M.); (L.C.)
| |
Collapse
|
24
|
Logan RW, Xue X, Ketchesin KD, Hoffman G, Roussos P, Tseng G, McClung CA, Seney ML. Sex Differences in Molecular Rhythms in the Human Cortex. Biol Psychiatry 2022; 91:152-162. [PMID: 33934884 PMCID: PMC8423868 DOI: 10.1016/j.biopsych.2021.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Diurnal rhythms in gene expression have been detected in the human brain. Previous studies found that males and females exhibit 24-hour rhythms in known circadian genes, with earlier peak expression in females. Whether there are sex differences in large-scale transcriptional rhythms in the cortex that align with observed sex differences in physiological and behavioral rhythms is currently unknown. METHODS Diurnal rhythmicity of gene expression was determined for males and females using RNA sequencing data from human postmortem dorsolateral prefrontal cortex (DLPFC) and anterior cingulate cortex (ACC). Sex differences among rhythmic genes were determined using significance cutoffs, threshold-free analyses, and R2 difference. Phase concordance was assessed across the DLPFC and ACC for males and females. Pathway and transcription factor analyses were also conducted on significantly rhythmic genes. RESULTS Canonical circadian genes had diurnal rhythms in both sexes with similar amplitude and phase. When analyses were expanded to the entire transcriptome, significant sex differences in transcriptional rhythms emerged. There were nearly twice as many rhythmic transcripts in the DLPFC in males and nearly 4 times as many rhythmic transcripts in the ACC in females. Results suggest a diurnal rhythm in synaptic transmission specific to the ACC in females (e.g., GABAergic [gamma-aminobutyric acidergic] and cholinergic neurotransmission). For males, there was phase concordance between the DLPFC and ACC, while phase asynchrony was found in females. CONCLUSIONS There are robust sex differences in molecular rhythms of genes in the DLPFC and ACC, providing potential mechanistic insights into how neurotransmission and synaptic function are modulated in a circadian-dependent and sex-specific manner.
Collapse
Affiliation(s)
- Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts; Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyle D Ketchesin
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Gabriel Hoffman
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York
| | - Panos Roussos
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York; Mental Illness Research, Education, and Clinical Center, James J. Peters VA Medical Center, Bronx, New York
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Colleen A McClung
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine; Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania.
| |
Collapse
|
25
|
mRNA Trafficking in the Nervous System: A Key Mechanism of the Involvement of Activity-Regulated Cytoskeleton-Associated Protein (Arc) in Synaptic Plasticity. Neural Plast 2021; 2021:3468795. [PMID: 34603440 PMCID: PMC8486535 DOI: 10.1155/2021/3468795] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/15/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
Synaptic activity mediates information storage and memory consolidation in the brain and requires a fast de novo synthesis of mRNAs in the nucleus and proteins in synapses. Intracellular localization of a protein can be achieved by mRNA trafficking and localized translation. Activity-regulated cytoskeleton-associated protein (Arc) is a master regulator of synaptic plasticity and plays an important role in controlling large signaling networks implicated in learning, memory consolidation, and behavior. Transcription of the Arc gene may be induced by a short behavioral event, resulting in synaptic activation. Arc mRNA is exported into the cytoplasm and can be trafficked into the dendrite of an activated synapse where it is docked and translated. The structure of Arc is similar to the viral GAG (group-specific antigen) protein, and phylogenic analysis suggests that Arc may originate from the family of Ty3/Gypsy retrotransposons. Therefore, Arc might evolve through “domestication” of retroviruses. Arc can form a capsid-like structure that encapsulates a retrovirus-like sentence in the 3′-UTR (untranslated region) of Arc mRNA. Such complex can be loaded into extracellular vesicles and transported to other neurons or muscle cells carrying not only genetic information but also regulatory signals within neuronal networks. Therefore, Arc mRNA inter- and intramolecular trafficking is essential for the modulation of synaptic activity required for memory consolidation and cognitive functions. Recent studies with single-molecule imaging in live neurons confirmed and extended the role of Arc mRNA trafficking in synaptic plasticity.
Collapse
|
26
|
Olivero G, Cisani F, Marimpietri D, Di Paolo D, Gagliani MC, Podestà M, Cortese K, Pittaluga A. The Depolarization-Evoked, Ca 2+-Dependent Release of Exosomes From Mouse Cortical Nerve Endings: New Insights Into Synaptic Transmission. Front Pharmacol 2021; 12:670158. [PMID: 34366842 PMCID: PMC8339587 DOI: 10.3389/fphar.2021.670158] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/22/2021] [Indexed: 01/08/2023] Open
Abstract
Whether exosomes can be actively released from presynaptic nerve terminals is a matter of debate. To address the point, mouse cortical synaptosomes were incubated under basal and depolarizing (25 mM KCl-enriched medium) conditions, and extracellular vesicles were isolated from the synaptosomal supernatants to be characterized by dynamic light scattering, transmission electron microscopy, Western blot, and flow cytometry analyses. The structural and biochemical analysis unveiled that supernatants contain vesicles that have the size and the shape of exosomes, which were immunopositive for the exosomal markers TSG101, flotillin-1, CD63, and CD9. The marker content increased upon the exposure of nerve terminals to the high-KCl stimulus, consistent with an active release of the exosomes from the depolarized synaptosomes. High KCl-induced depolarization elicits the Ca2+-dependent exocytosis of glutamate. Interestingly, the depolarization-evoked release of exosomes from cortical synaptosomes also occurred in a Ca2+-dependent fashion, since the TSG101, CD63, and CD9 contents in the exosomal fraction isolated from supernatants of depolarized synaptosomes were significantly reduced when omitting external Ca2+ ions. Differently, (±)-baclofen (10 µM), which significantly reduced the glutamate exocytosis, did not affect the amount of exosomal markers, suggesting that the GABAB-mediated mechanism does not control the exosome release. Our findings suggest that the exposure of synaptosomes to a depolarizing stimulus elicits a presynaptic release of exosomes that occurs in a Ca2+-dependent fashion. The insensitivity to the presynaptic GABAB receptors, however, leaves open the question on whether the release of exosomes could be a druggable target for new therapeutic intervention for the cure of synaptopathies.
Collapse
Affiliation(s)
- Guendalina Olivero
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Francesca Cisani
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
| | - Danilo Marimpietri
- Stem Cell Laboratory and Cell Therapy Center, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Daniela Di Paolo
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Maria Cristina Gagliani
- Department of Experimental Medicine, DIMES, Human Anatomy Section, University of Genoa, Genoa, Italy
| | - Marina Podestà
- Stem Cell Laboratory and Cell Therapy Center, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Katia Cortese
- Department of Experimental Medicine, DIMES, Human Anatomy Section, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, Centre of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
27
|
Regulation of the NMDA receptor by its cytoplasmic domains: (How) is the tail wagging the dog? Neuropharmacology 2021; 195:108634. [PMID: 34097949 DOI: 10.1016/j.neuropharm.2021.108634] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 12/18/2022]
Abstract
Excitatory neurotransmission mediated by N-methyl-d-aspartate receptors (NMDARs) is critical for synapse development, function, and plasticity in the brain. NMDARs are tetra-heteromeric cation-channels that mediate synaptic transmission and plasticity. Extensive human studies show the existence of genetic variants in NMDAR subunits genes (GRIN genes) that are associated with neurodevelopmental and neuropsychiatric disorders, including autism spectrum disorders (ASD), epilepsy (EP), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), and schizophrenia (SCZ). NMDAR subunits have a unique modular architecture with four semiautonomous domains. Here we focus on the carboxyl terminal domain (CTD), also known as the intracellular C-tail, which varies in length among the glutamate receptor subunits and is the most diverse domain in terms of amino acid sequence. The CTD shows no sequence homology to any known proteins but encodes short docking motifs for intracellular binding proteins and covalent modifications. Our review will discuss the many important functions of the CTD in regulating NMDA membrane and synaptic targeting, stabilization, degradation targeting, allosteric modulation and metabotropic signaling of the receptor. This article is part of the special issue on 'Glutamate Receptors - NMDA Receptors'.
Collapse
|
28
|
Wallis TP, Venkatesh BG, Narayana VK, Kvaskoff D, Ho A, Sullivan RK, Windels F, Sah P, Meunier FA. Saturated free fatty acids and association with memory formation. Nat Commun 2021; 12:3443. [PMID: 34103527 PMCID: PMC8187648 DOI: 10.1038/s41467-021-23840-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 05/14/2021] [Indexed: 02/05/2023] Open
Abstract
Polyunsaturated free fatty acids (FFAs) such as arachidonic acid, released by phospholipase activity on membrane phospholipids, have long been considered beneficial for learning and memory and are known modulators of neurotransmission and synaptic plasticity. However, the precise nature of other FFA and phospholipid changes in specific areas of the brain during learning is unknown. Here, using a targeted lipidomics approach to characterise FFAs and phospholipids across the rat brain, we demonstrated that the highest concentrations of these analytes were found in areas of the brain classically involved in fear learning and memory, such as the amygdala. Auditory fear conditioning led to an increase in saturated (particularly myristic and palmitic acids) and to a lesser extent unsaturated FFAs (predominantly arachidonic acid) in the amygdala and prefrontal cortex. Both fear conditioning and changes in FFA required activation of NMDA receptors. These results suggest a role for saturated FFAs in memory acquisition.
Collapse
Affiliation(s)
- Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Bharat G Venkatesh
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Vinod K Narayana
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Metabolomics Australia, Bio21 Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - David Kvaskoff
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Biberach an der Riß, Germany
| | - Alan Ho
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Robert K Sullivan
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - François Windels
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Pankaj Sah
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Joint Center for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province, P. R. China
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
29
|
Kneussel M, Sánchez-Rodríguez N, Mischak M, Heisler FF. Dynein and muskelin control myosin VI delivery towards the neuronal nucleus. iScience 2021; 24:102416. [PMID: 33997696 PMCID: PMC8099778 DOI: 10.1016/j.isci.2021.102416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/15/2021] [Accepted: 04/07/2021] [Indexed: 11/30/2022] Open
Abstract
Protein transport toward the nucleus is important for translating molecular signals into gene expression changes. Interestingly, the unconventional motor protein myosin VI regulates RNA polymerase II-dependent gene transcription. Whether actin-filament-dependent myosins are actively transported to nuclear compartments remains unknown. Here, we report that neurons also contain myosin VI inside their nucleus. Notably, nuclear appearance of this actin-dependent motor depends on functional cytoplasmic dynein, a minus end-directed microtubule motor. We find that the trafficking factor muskelin assists in the formation of dynein-myosin VI interactions and further localizes to nuclear foci, enriched in the myosin. Impairment of dynein, but not myosin VI function, reduces nuclear muskelin levels. In turn, muskelin represents a critical determinant in regulating myosin VI nuclear targeting. Our data reveal that minus end-directed microtubule transport determines myosin VI subcellular localization. They suggest a pathway of cytoplasm-to-nucleus trafficking that requires muskelin and is based on dynein-myosin cross talk.
Collapse
Affiliation(s)
- Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Noelia Sánchez-Rodríguez
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Michaela Mischak
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Frank F. Heisler
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| |
Collapse
|
30
|
Acutain MF, Griebler Luft J, Vazquez CA, Popik B, Cercato MC, Epstein A, Salvetti A, Jerusalinsky DA, de Oliveira Alvares L, Baez MV. Reduced Expression of Hippocampal GluN2A-NMDAR Increases Seizure Susceptibility and Causes Deficits in Contextual Memory. Front Neurosci 2021; 15:644100. [PMID: 33897358 PMCID: PMC8064689 DOI: 10.3389/fnins.2021.644100] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/08/2021] [Indexed: 12/23/2022] Open
Abstract
N-methyl-D-aspartate receptors are heterotetramers composed of two GluN1 obligatory subunits and two regulatory subunits. In cognitive-related brain structures, GluN2A and GluN2B are the most abundant regulatory subunits, and their expression is subjected to tight regulation. During development, GluN2B expression is characteristic of immature synapses, whereas GluN2A is present in mature ones. This change in expression induces a shift in GluN2A/GluN2B ratio known as developmental switch. Moreover, modifications in this relationship have been associated with learning and memory, as well as different pathologies. In this work, we used a specific shRNA to induce a reduction in GluN2A expression after the developmental switch, both in vitro in primary cultured hippocampal neurons and in vivo in adult male Wistar rats. After in vitro characterization, we performed a cognitive profile and evaluated seizure susceptibility in vivo. Our in vitro results showed that the decrease in the expression of GluN2A changes GluN2A/GluN2B ratio without altering the expression of other regulatory subunits. Moreover, rats expressing the anti-GluN2A shRNA in vivo displayed an impaired contextual fear-conditioning memory. In addition, these animals showed increased seizure susceptibility, in terms of both time and intensity, which led us to conclude that deregulation in GluN2A expression at the hippocampus is associated with seizure susceptibility and learning–memory mechanisms.
Collapse
Affiliation(s)
- Maria Florencia Acutain
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | - Jordana Griebler Luft
- Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cecila Alejandra Vazquez
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | - Bruno Popik
- Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Magalí C Cercato
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | | | - Anna Salvetti
- International Center for Infectiology Research (CIRI), INSERM U1111, CNRS UMR5308, Université de Lyon (UCBL1), Lyon, France
| | - Diana A Jerusalinsky
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina
| | | | - Maria Verónica Baez
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN, CONICET-UBA), Buenos Aires, Argentina.,1° U.A. Departamento de Histologia, Embriología, Biologia Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
31
|
Gardoni F, Di Luca M. Protein-protein interactions at the NMDA receptor complex: From synaptic retention to synaptonuclear protein messengers. Neuropharmacology 2021; 190:108551. [PMID: 33819458 DOI: 10.1016/j.neuropharm.2021.108551] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022]
Abstract
N-methyl-d-aspartate receptors (NMDARs) are glutamate-gated ion channels that support essential functions throughout the brain. NMDARs are tetramers composed of the GluN1 subunit in complex with GluN2- and GluN3-type regulatory subunits, resulting in the formation of various receptor subtypes throughout the central nervous system (CNS), characterised by different kinetics, biophysical and pharmacological properties, and the abilities to interact with specific partners at dendritic spines. NMDARs are expressed at high levels, are widely distributed throughout the brain, and are involved in several physiological and pathological conditions. Here, we will focus on the GluN2A- and GluN2B-containing NMDARs found at excitatory synapses and their interactions with plasticity-relevant proteins, such as the postsynaptic density family of membrane-associated guanylate kinases (PSD-MAGUKs), Ca2+/calmodulin-dependent kinase II (CaMKII) and synaptonuclear protein messengers. The dynamic interactions between NMDAR subunits and various proteins regulating synaptic receptor retention and synaptonuclear signalling mediated by protein messengers suggest that the NMDAR serves as a key molecular player that coordinates synaptic activity and cell-wide events that require gene transcription. Importantly, protein-protein interactions at the NMDAR complex can also contribute to synaptic dysfunction in several brain disorders. Therefore, the modulation of the molecular composition of the NMDAR complex might represent a novel pharmacological approach for the treatment of certain disease states.
Collapse
Affiliation(s)
- Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy.
| |
Collapse
|
32
|
Synaptic GluN2A-Containing NMDA Receptors: From Physiology to Pathological Synaptic Plasticity. Int J Mol Sci 2020; 21:ijms21041538. [PMID: 32102377 PMCID: PMC7073220 DOI: 10.3390/ijms21041538] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
N-Methyl-d-Aspartate Receptors (NMDARs) are ionotropic glutamate-gated receptors. NMDARs are tetramers composed by several homologous subunits of GluN1-, GluN2-, or GluN3-type, leading to the existence in the central nervous system of a high variety of receptor subtypes with different pharmacological and signaling properties. NMDAR subunit composition is strictly regulated during development and by activity-dependent synaptic plasticity. Given the differences between GluN2 regulatory subunits of NMDAR in several functions, here we will focus on the synaptic pool of NMDARs containing the GluN2A subunit, addressing its role in both physiology and pathological synaptic plasticity as well as the contribution in these events of different types of GluN2A-interacting proteins.
Collapse
|