51
|
Gall CM, Le AA, Lynch G. Sex differences in synaptic plasticity underlying learning. J Neurosci Res 2023; 101:764-782. [PMID: 33847004 PMCID: PMC10337639 DOI: 10.1002/jnr.24844] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 11/09/2022]
Abstract
Although sex differences in learning behaviors are well documented, sexual dimorphism in the synaptic processes of encoding is only recently appreciated. Studies in male rodents have built upon the discovery of long-term potentiation (LTP), and acceptance of this activity-dependent increase in synaptic strength as a mechanism of encoding, to identify synaptic receptors and signaling activities that coordinate the activity-dependent remodeling of the subsynaptic actin cytoskeleton that is critical for enduring potentiation and memory. These molecular substrates together with other features of LTP, as characterized in males, have provided an explanation for a range of memory phenomena including multiple stages of consolidation, the efficacy of spaced training, and the location of engrams at the level of individual synapses. In the present report, we summarize these findings and describe more recent results from our laboratories showing that in females the same actin regulatory mechanisms are required for hippocampal LTP and memory but, in females only, the engagement of both modulatory receptors such as TrkB and synaptic signaling intermediaries including Src and ERK1/2 requires neuron-derived estrogen and signaling through membrane-associated estrogen receptor α (ERα). Moreover, in association with the additional ERα involvement, females exhibit a higher threshold for hippocampal LTP and spatial learning. We propose that the distinct LTP threshold in females contributes to as yet unappreciated sex differences in information processing and features of learning and memory.
Collapse
Affiliation(s)
- Christine M. Gall
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Aliza A. Le
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| |
Collapse
|
52
|
Petter EA, Fallon IP, Hughes RN, Watson GDR, Meck WH, Ulloa Severino FP, Yin HH. Elucidating a locus coeruleus-dentate gyrus dopamine pathway for operant reinforcement. eLife 2023; 12:e83600. [PMID: 37083584 PMCID: PMC10162798 DOI: 10.7554/elife.83600] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/20/2023] [Indexed: 04/22/2023] Open
Abstract
Animals can learn to repeat behaviors to earn desired rewards, a process commonly known as reinforcement learning. While previous work has implicated the ascending dopaminergic projections to the basal ganglia in reinforcement learning, little is known about the role of the hippocampus. Here, we report that a specific population of hippocampal neurons and their dopaminergic innervation contribute to operant self-stimulation. These neurons are located in the dentate gyrus, receive dopaminergic projections from the locus coeruleus, and express D1 dopamine receptors. Activation of D1 + dentate neurons is sufficient for self-stimulation: mice will press a lever to earn optogenetic activation of these neurons. A similar effect is also observed with selective activation of the locus coeruleus projections to the dentate gyrus, and blocked by D1 receptor antagonism. Calcium imaging of D1 + dentate neurons revealed significant activity at the time of action selection, but not during passive reward delivery. These results reveal the role of dopaminergic innervation of the dentate gyrus in supporting operant reinforcement.
Collapse
Affiliation(s)
- Elijah A Petter
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Isabella P Fallon
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Ryan N Hughes
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Glenn DR Watson
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Warren H Meck
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Francesco Paolo Ulloa Severino
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Henry H Yin
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
- Department of Neurobiology, Duke University School of MedicineDurhamUnited States
| |
Collapse
|
53
|
Usmani MT, Krattli RP, El-Khatib SM, Le ACD, Smith SM, Baulch JE, Ng DQ, Acharya MM, Chan A. BDNF Augmentation Using Riluzole Reverses Doxorubicin-Induced Decline in Cognitive Function and Neurogenesis. Neurotherapeutics 2023; 20:838-852. [PMID: 36720792 PMCID: PMC10275819 DOI: 10.1007/s13311-022-01339-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2022] [Indexed: 02/02/2023] Open
Abstract
Cancer-related cognitive impairment (CRCI) considerably affects the quality of life of millions of cancer survivors. Brain-derived neurotrophic factor (BDNF) has been shown to promote survival, differentiation, and maintenance of in vivo dentate neurogenesis, and chemotherapy induces a plethora of physiological and cellular alterations, including a decline in neurogenesis and increased neuroinflammation linked with cognitive impairments. In our clinical studies, breast cancer patients treated with doxorubicin (Adriamycin®, ADR) experienced a significant reduction in the blood levels of BDNF that was associated with a higher risk of CRCI. Our past rodent studies in CRCI have also shown a significant reduction in dentate neurogenesis accompanied by cognitive impairment. In this study, using a female mouse model of ADR-induced cognitive decline, we tested the impact of riluzole (RZ), an orally active BDNF-enhancing medication that is FDA-approved for amyotrophic lateral sclerosis. ADR-treated mice receiving RZ in the drinking water for 1 month showed significant improvements in hippocampal-dependent learning and memory function (spatial recognition), fear extinction memory consolidation, and reduced anxiety-like behavior. RZ prevented chemotherapy-induced reductions of BDNF levels in the hippocampus. Importantly, RZ mitigated chemotherapy-induced loss of newly born, immature neurons, dentate neurogenesis, and neuroinflammation. In conclusion, this data provides pre-clinical evidence for a translationally feasible approach to enhance the neuroprotective effects of RZ treatment to prevent CRCI.
Collapse
Affiliation(s)
- Manal T Usmani
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Robert P Krattli
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Sanad M El-Khatib
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Anh C D Le
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Sarah M Smith
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA
| | - Janet E Baulch
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA.
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA.
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
54
|
Ehtezazi T, Rahman K, Davies R, Leach AG. The Pathological Effects of Circulating Hydrophobic Bile Acids in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:173-211. [PMID: 36994114 PMCID: PMC10041467 DOI: 10.3233/adr-220071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Recent clinical studies have revealed that the serum levels of toxic hydrophobic bile acids (deoxy cholic acid, lithocholic acid [LCA], and glycoursodeoxycholic acid) are significantly higher in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) when compared to control subjects. The elevated serum bile acids may be the result of hepatic peroxisomal dysfunction. Circulating hydrophobic bile acids are able to disrupt the blood-brain barrier and promote the formation of amyloid-β plaques through enhancing the oxidation of docosahexaenoic acid. Hydrophobic bile acid may find their ways into the neurons via the apical sodium-dependent bile acid transporter. It has been shown that hydrophobic bile acids impose their pathological effects by activating farnesoid X receptor and suppressing bile acid synthesis in the brain, blocking NMDA receptors, lowering brain oxysterol levels, and interfering with 17β-estradiol actions such as LCA by binding to E2 receptors (molecular modelling data exclusive to this paper). Hydrophobic bile acids may interfere with the sonic hedgehog signaling through alteration of cell membrane rafts and reducing brain 24(S)-hydroxycholesterol. This article will 1) analyze the pathological roles of circulating hydrophobic bile acids in the brain, 2) propose therapeutic approaches, and 3) conclude that consideration be given to reducing/monitoring toxic bile acid levels in patients with AD or aMCI, prior/in combination with other treatments.
Collapse
Affiliation(s)
- Touraj Ehtezazi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rhys Davies
- The Walton Centre, NHS Foundation Trust, Liverpool, UK
| | - Andrew G Leach
- School of Pharmacy, University of Manchester, Manchester, UK
| |
Collapse
|
55
|
Rajão-Saraiva J, Dunot J, Ribera A, Temido-Ferreira M, Coelho JE, König S, Moreno S, Enguita FJ, Willem M, Kins S, Marie H, Lopes LV, Pousinha PA. Age-dependent NMDA receptor function is regulated by the amyloid precursor protein. Aging Cell 2023; 22:e13778. [PMID: 36704841 PMCID: PMC10014064 DOI: 10.1111/acel.13778] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/13/2022] [Accepted: 12/21/2022] [Indexed: 01/28/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are critical for the maturation and plasticity of glutamatergic synapses. In the hippocampus, NMDARs mainly contain GluN2A and/or GluN2B regulatory subunits. The amyloid precursor protein (APP) has emerged as a putative regulator of NMDARs, but the impact of this interaction to their function is largely unknown. By combining patch-clamp electrophysiology and molecular approaches, we unravel a dual mechanism by which APP controls GluN2B-NMDARs, depending on the life stage. We show that APP is highly abundant specifically at the postnatal postsynapse. It interacts with GluN2B-NMDARs, controlling its synaptic content and mediated currents, both in infant mice and primary neuronal cultures. Upon aging, the APP amyloidogenic-derived C-terminal fragments, rather than APP full-length, contribute to aberrant GluN2B-NMDAR currents. Accordingly, we found that the APP processing is increased upon aging, both in mice and human brain. Interfering with stability or production of the APP intracellular domain normalized the GluN2B-NMDARs currents. While the first mechanism might be essential for synaptic maturation during development, the latter could contribute to age-related synaptic impairments.
Collapse
Affiliation(s)
- Joana Rajão-Saraiva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Jade Dunot
- University Côte d' Azur, Centre National de la Recherche Scientifique (CNRS) UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Aurore Ribera
- University Côte d' Azur, Centre National de la Recherche Scientifique (CNRS) UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Mariana Temido-Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Svenja König
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Sébastien Moreno
- University Côte d' Azur, Centre National de la Recherche Scientifique (CNRS) UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Francisco J Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Michael Willem
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan Kins
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Hélène Marie
- University Côte d' Azur, Centre National de la Recherche Scientifique (CNRS) UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Luísa V Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Paula A Pousinha
- University Côte d' Azur, Centre National de la Recherche Scientifique (CNRS) UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| |
Collapse
|
56
|
Roux CM, Lecouflet P, Billard JM, Esneault E, Leger M, Schumann-Bard P, Freret T. Genetic Background Influence on Hippocampal Synaptic Plasticity: Frequency-Dependent Variations between an Inbred and an Outbred Mice Strain. Int J Mol Sci 2023; 24:ijms24054304. [PMID: 36901735 PMCID: PMC10001449 DOI: 10.3390/ijms24054304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
For almost half a century, acute hippocampal slice preparations have been widely used to investigate anti-amnesic (or promnesic) properties of drug candidates on long-term potentiation (LTP)-a cellular substrate that supports some forms of learning and memory. The large variety of transgenic mice models now available makes the choice of the genetic background when designing experiments crucially important. Furthermore, different behavioral phenotypes were reported between inbred and outbred strains. Notably, some differences in memory performance were emphasized. Despite this, investigations, unfortunately, did not explore electrophysiological properties. In this study, two stimulation paradigms were used to compare LTP in the hippocampal CA1 area of both inbred (C57BL/6) and outbred (NMRI) mice. High-frequency stimulation (HFS) revealed no strain difference, whereas theta-burst stimulation (TBS) resulted in significantly reduced LTP magnitude in NMRI mice. Additionally, we demonstrated that this reduced LTP magnitude (exhibited by NMRI mice) was due to lower responsiveness to theta-frequency during conditioning stimuli. In this paper, we discuss the anatomo-functional correlates that may explain such hippocampal synaptic plasticity divergence, although straightforward evidence is still lacking. Overall, our results support the prime importance of considering the animal model related to the intended electrophysiological experiments and the scientific issues to be addressed.
Collapse
Affiliation(s)
- Candice M. Roux
- Department of Health, UNICAEN, INSERM, COMETE, CYCERON, Normandie University, 14000 Caen, France
- PORSOLT, 53940 Le Genest Saint-Isle, France
| | - Pierre Lecouflet
- Department of Health, UNICAEN, INSERM, COMETE, CYCERON, Normandie University, 14000 Caen, France
| | - Jean-Marie Billard
- Department of Health, UNICAEN, INSERM, COMETE, CYCERON, Normandie University, 14000 Caen, France
| | | | - Marianne Leger
- Department of Health, UNICAEN, INSERM, COMETE, CYCERON, Normandie University, 14000 Caen, France
| | - Pascale Schumann-Bard
- Department of Health, UNICAEN, INSERM, COMETE, CYCERON, Normandie University, 14000 Caen, France
| | - Thomas Freret
- Department of Health, UNICAEN, INSERM, COMETE, CYCERON, Normandie University, 14000 Caen, France
- Correspondence: ; Tel.: +33-2-31-56-68-77
| |
Collapse
|
57
|
Yin L, Zhang J, Ma H, Zhang X, Fan Z, Yang Y, Li M, Han J, Zhang X. Selective activation of cholinergic neurotransmission from the medial septal nucleus to hippocampal pyramidal neurones improves sepsis-induced cognitive deficits in mice. Br J Anaesth 2023; 130:573-584. [PMID: 36813621 DOI: 10.1016/j.bja.2023.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/15/2022] [Accepted: 01/07/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy is characterised by cognitive dysfunction, and might be mediated by deficits in neurotransmission. Reduced cholinergic neurotransmission in the hippocampus impairs memory function. We assessed real-time alterations of acetylcholine neurotransmission from the medial septal nucleus to the hippocampus, and explored whether sepsis-induced cognitive deficits can be relieved by activating upstream cholinergic projections. METHOD Lipopolysaccharide (LPS) injection or caecal ligation and puncture (CLP) was used to induce sepsis and associated neuroinflammation in wild-type and mutant mice. Adeno-associated viruses for calcium and acetylcholine imaging, and for optogenetic and chemogenetic modulation of cholinergic neurones were injected into the hippocampus or medial septum, and a 200-μm-diameter optical fibre was implanted to collect acetylcholine and calcium signals. Cholinergic activity of the medial septum was manipulated and combined with cognitive assessment after LPS injection or CLP. RESULTS Intracerebroventricular LPS injection reduced postsynaptic acetylcholine (from 0.146 [0.001] to 0.0047 [0.0005]; p=0.004) and calcium (from 0.0236 [0.0075] to 0.0054 [0.0026]; p=0.0388) signals in hippocampal Vglut2-positive glutamatergic neurones, whereas optogenetic activation of cholinergic neurones in the medial septum reversed LPS-induced reductions in these two signals. Intraperitoneal LPS injection decreased acetylcholine concentration in the hippocampus (476 [20] pg ml-1 to 382 [14] pg ml-1; p=0.0001). Reduction in long-term potentiation (238 [23] % to 150 [12] %; p=0.0082) and enhancement of hippocampal pyramidal neurone action potential frequency (5.8 [1.5] Hz to 8.2 [1.8] Hz; p=0.0343) were relieved, and neurocognitive performance was improved by chemogenetic activation of cholinergic innervation of the hippocampus 3 days after LPS injection in septic mice. CONCLUSIONS Systemic or local LPS reduced cholinergic neurotransmission from the medial septum to hippocampal pyramidal neurones, and their selective activation alleviated defects in hippocampal neuronal function and synaptic plasticity and ameliorated memory deficits in sepsis model mice through enhanced cholinergic neurotransmission. This provides a basis for targeting cholinergic signalling to the hippocampus in sepsis-induced encephalopathy.
Collapse
Affiliation(s)
- Lu Yin
- Department of Critical Care Medicine and Department of Anaesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinming Zhang
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Hongwei Ma
- Department of Critical Care Medicine and Department of Anaesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xinxin Zhang
- Department of Critical Care Medicine and Department of Anaesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhongmin Fan
- Department of Critical Care Medicine and Department of Anaesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yongheng Yang
- Department of Critical Care Medicine and Department of Anaesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mengyun Li
- Department of Critical Care Medicine and Department of Anaesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jing Han
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China.
| | - Xijing Zhang
- Department of Critical Care Medicine and Department of Anaesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
58
|
Fan LZ, Kim DK, Jennings JH, Tian H, Wang PY, Ramakrishnan C, Randles S, Sun Y, Thadhani E, Kim YS, Quirin S, Giocomo L, Cohen AE, Deisseroth K. All-optical physiology resolves a synaptic basis for behavioral timescale plasticity. Cell 2023; 186:543-559.e19. [PMID: 36669484 PMCID: PMC10327443 DOI: 10.1016/j.cell.2022.12.035] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 10/19/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023]
Abstract
Learning has been associated with modifications of synaptic and circuit properties, but the precise changes storing information in mammals have remained largely unclear. We combined genetically targeted voltage imaging with targeted optogenetic activation and silencing of pre- and post-synaptic neurons to study the mechanisms underlying hippocampal behavioral timescale plasticity. In mice navigating a virtual-reality environment, targeted optogenetic activation of individual CA1 cells at specific places induced stable representations of these places in the targeted cells. Optical elicitation, recording, and modulation of synaptic transmission in behaving mice revealed that activity in presynaptic CA2/3 cells was required for the induction of plasticity in CA1 and, furthermore, that during induction of these place fields in single CA1 cells, synaptic input from CA2/3 onto these same cells was potentiated. These results reveal synaptic implementation of hippocampal behavioral timescale plasticity and define a methodology to resolve synaptic plasticity during learning and memory in behaving mammals.
Collapse
Affiliation(s)
- Linlin Z Fan
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Doo Kyung Kim
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Joshua H Jennings
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - He Tian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Peter Y Wang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Sawyer Randles
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yanjun Sun
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | - Elina Thadhani
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Sean Quirin
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Lisa Giocomo
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA; Department of Physics, Harvard University, Cambridge, MA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford, CA, USA.
| |
Collapse
|
59
|
Basak S, Duttaroy AK. Maternal PUFAs, Placental Epigenetics, and Their Relevance to Fetal Growth and Brain Development. Reprod Sci 2023; 30:408-427. [PMID: 35676498 DOI: 10.1007/s43032-022-00989-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/24/2022] [Indexed: 12/17/2022]
Abstract
Dietary polyunsaturated fatty acids (PUFAs), especially omega-3 (n-3) and n-6 long-chain (LC) PUFAs, are indispensable for the fetus' brain supplied by the placenta. Despite being highly unsaturated, n-3 LCPUFA-docosahexaenoic acid (DHA) plays a protective role as an antioxidant in the brain. Deficiency of DHA during fetal development may cause irreversible damages in neurodevelopment programming. Dietary PUFAs can impact placental structure and functions by regulating early placentation processes, such as angiogenesis. They promote remodeling of uteroplacental architecture to facilitate increased blood flow and surface area for nutrient exchange. The placenta's fatty acid transfer depends on the uteroplacental vascular development, ensuring adequate maternal circulatory fatty acids transport to fulfill the fetus' rapid growth and development requirements. Maternal n-3 PUFA deficiency predominantly leads to placental epigenetic changes than other fetal developing organs. A global shift in DNA methylation possibly transmits epigenetic instability in developing fetuses due to n-3 PUFA deficiency. Thus, an optimal level of maternal omega-3 (n-3) PUFAs may protect the placenta's structural and functional integrity and allow fetal growth by controlling the aberrant placental epigenetic changes. This narrative review summarizes the recent advances and underpins the roles of maternal PUFAs on the structure and functions of the placenta and their relevance to fetal growth and brain development.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India.
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
60
|
The molecular memory code and synaptic plasticity: A synthesis. Biosystems 2023; 224:104825. [PMID: 36610586 DOI: 10.1016/j.biosystems.2022.104825] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
The most widely accepted view of memory in the brain holds that synapses are the storage sites of memory, and that memories are formed through associative modification of synapses. This view has been challenged on conceptual and empirical grounds. As an alternative, it has been proposed that molecules within the cell body are the storage sites of memory, and that memories are formed through biochemical operations on these molecules. This paper proposes a synthesis of these two views, grounded in a computational model of memory. Synapses are conceived as storage sites for the parameters of an approximate posterior probability distribution over latent causes. Intracellular molecules are conceived as storage sites for the parameters of a generative model. The model stipulates how these two components work together as part of an integrated algorithm for learning and inference.
Collapse
|
61
|
Küpper M, Porath K, Sellmann T, Bien CG, Köhling R, Kirschstein T. GluN2B inhibition rescues impaired potentiation and epileptogenicity at associational-commissural CA3 synapses in a model of anti-NMDAR encephalitis. Neurosci Lett 2023; 795:137031. [PMID: 36574811 DOI: 10.1016/j.neulet.2022.137031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Anti-N-methyl-d-aspartate receptor (anti-NMDAR) encephalitis is an autoimmune epilepsy associated with memory deficits. Research has demonstrated that anti-NMDAR inhibit long-term potentiation, and, at the same time, lead to disinhibition in the form of epileptiform afterpotentials in the potentiated state. While both effects may give rise to the key symptoms of the disease, the molecular basis of being simultaneously inhibitory and disinhibitory is difficult to explain. Here, we explored a possible involvement of the GluN2B subunit. To this aim, we injected cerebrospinal fluid from anti-NMDAR encephalitis patients into the rat hippocampus and prepared brain slices for in vitro field potential recordings. Associational-commissural-fiber-CA3 synapses from anti-NMDAR-treated animals showed increased field potential amplitudes with concomitantly enhanced paired-pulse ratios as compared to control tissue. GluN2B inhibition by Ro25-6981 mimicked these effects in controls but had no effect in anti-NMDAR tissues indicating a presynaptic and occluding effect of anti-NMDAR. We then induced potentiation of associational-commissural-fiber-CA3 synapses, and confirmed that slices from anti-NMDAR-treated animals showed reduced potentiation and pronounced epileptiform afterpotentials. Intriguingly, both effects were absent when Ro25-6981 was added in vitro before inducing potentiation. These results indicate that GluN2B-containing NMDARs, partially expressed presynaptically, show differential sensitivity to anti-NMDAR, and that altered GluN2B function is particularly apparent in the potentiated state rather than under baseline conditions. Since GluN2B inhibition rescued the effects of anti-NMDAR in the potentiated state, this opens the possibility that at least a subgroup of patients could benefit from a GluN2B antagonist.
Collapse
Affiliation(s)
- Maraike Küpper
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany
| | - Tina Sellmann
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany
| | - Christian G Bien
- Department of Epileptology (Krankenhaus Mara), Bielefeld University, Medical School, Bielefeld, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany; Center of Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Germany; Center of Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Germany.
| |
Collapse
|
62
|
Obot P, Subah G, Schonwald A, Pan J, Velíšek L, Velíšková J, Stanton PK, Scemes E. Astrocyte and neuronal Panx1 support long-term reference memory in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524236. [PMID: 36711845 PMCID: PMC9882221 DOI: 10.1101/2023.01.16.524236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Pannexin 1 (Panx1) are ubiquitously expressed proteins that form plasma membrane channels permeable to anions and moderate sized signaling molecules (e.g., ATP, glutamate). In the nervous system, activation of Panx1 channels have been extensively shown to contribute to distinct neurological disorders (epilepsy, chronic pain, migraine, neuroAIDS, etc.) but knowledge of extent to which these channels have a physiological role remains restricted to three studies supporting their involvement in hippocampus dependent learning. Given that Panx1 channels may provide an important mechanism for activity-dependent neuron-glia interaction, we used Panx1 transgenic mice with global and cell-type specific deletions of Panx1 to interrogate their participation in working and reference memory. Using the 8-arm radial maze, we show that long-term spatial reference memory, but not spatial working memory, is deficient in Panx1-null mice and that both astrocyte and neuronal Panx1 contribute to the consolidation of long-term spatial memory. Field potential recordings in hippocampal slices of Panx1-null mice revealed an attenuation of both long-term potentiation (LTP) of synaptic strength and long-term depression (LTD) at Schaffer collateral - CA1 synapses without alterations basal synaptic transmission or pre-synaptic paired-pulse facilitation. Our results implicate both neuronal and astrocyte Panx1 channels as critical players for the development and maintenance of long-term spatial reference memory in mice.
Collapse
|
63
|
Zhu X, Huang J, Wu Y, Zhao S, Chai X. Effect of Heat Stress on Hippocampal Neurogenesis: Insights into the Cellular and Molecular Basis of Neuroinflammation-Induced Deficits. Cell Mol Neurobiol 2023; 43:1-13. [PMID: 34767143 PMCID: PMC11415162 DOI: 10.1007/s10571-021-01165-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/01/2021] [Indexed: 01/07/2023]
Abstract
Heat stress is known to result in neuroinflammation, neuronal damage, and disabilities in learning and memory in animals and humans. It has previously been reported that cognitive impairment caused by neuroinflammation may at least in part be mediated by defective hippocampal neurogenesis, and defective neurogenesis has been linked to aberrantly activated microglial cells. Moreover, the release of cytokines within the brain has been shown to contribute to the disruption of cognitive functions in several conditions following neuroinflammation. In this review, we summarize evolving evidence for the current understanding of inflammation-induced deficits in hippocampal neurogenesis, and the resulting behavioral impairments after heat stress. Furthermore, we provide valuable insights into the molecular and cellular mechanisms underlying neuroinflammation-induced deficits in hippocampal neurogenesis, particularly relating to cognitive dysfunction following heat stress. Lastly, we aim to identify potential mechanisms through which neuroinflammation induces cognitive dysfunction, and elucidate how neuroinflammation contributes to defective hippocampal neurogenesis. This review may therefore help to better understand the relationship between hippocampal neurogenesis and heat stress.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China.
| | - Jian Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Xuejun Chai
- College of Basic Medicine, Xi'An Medical University, Xi'An, 710021, Shaanxi, People's Republic of China.
| |
Collapse
|
64
|
Goodman J, Leong KC, Packard MG. NMDA receptor blockade in the dorsolateral striatum impairs consolidation but not retrieval of habit memory. Neurobiol Learn Mem 2023; 197:107709. [PMID: 36503101 DOI: 10.1016/j.nlm.2022.107709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/24/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
The present study investigated whether N-methyl-d-aspartate (NMDA) receptors in the dorsolateral striatum (DLS) mediate consolidation and retrieval of habit memory. Adult male Long-Evans rats were trained in a response learning version of a water plus-maze task in which rats were reinforced to make a habitual and consistent body-turn response at the maze choice point in order to mount a hidden escape platform. Prior research indicates that acquisition, consolidation, and retrieval in this task requires DLS function. The present study consisted of two experiments. In Experiment 1, rats received intra-DLS post-training injections of the NMDA receptor antagonist 2-amino-5- phosphonopentanoic acid (AP5; 2 µg/side) to examine the role of NMDA receptors in consolidation of habit memory. In Experiment 2, different groups of rats received a single pre-retrieval injection of AP5 in the DLS (AP5; 2 µg/side) during the last day of maze training to examine the potential role of NMDA receptors in retrieval of habit memory. Results indicated that post-training intra-DLS AP5 injections impaired memory consolidation. However, administration of AP5 at the same dose that impaired consolidation had no effect on memory retrieval. The findings are consistent with previous research indicating a role for NMDA receptors in the DLS in memory consolidation, and suggest that NMDA-dependent synaptic activity in the DLS may not be a critical component of habit memory retrieval.
Collapse
Affiliation(s)
- Jarid Goodman
- Department of Psychology, Delaware State University, Dover, DE, United States
| | - Kah-Chung Leong
- Department of Psychology, Trinity University, San Antonio, TX, United States
| | - Mark G Packard
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
65
|
Kreis A, Issa F, Yerna X, Jabbour C, Schakman O, de Clippele M, Tajeddine N, Pierrot N, Octave JN, Gualdani R, Gailly P. Conditional deletion of KCC2 impairs synaptic plasticity and both spatial and nonspatial memory. Front Mol Neurosci 2023; 16:1081657. [PMID: 37168681 PMCID: PMC10164999 DOI: 10.3389/fnmol.2023.1081657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/31/2023] [Indexed: 05/13/2023] Open
Abstract
The postsynaptic inhibition through GABAA receptors (GABAAR) relies on two mechanisms, a shunting effect due to an increase in the postsynaptic membrane conductance and, in mature neurons, a hyperpolarization effect due to an entry of chloride into postsynaptic neurons. The second effect requires the action of the K+-Cl- cotransporter KCC2 which extrudes Cl- from the cell and maintains its cytosolic concentration very low. Neuronal chloride equilibrium seems to be dysregulated in several neurological and psychiatric conditions such as epilepsy, anxiety, schizophrenia, Down syndrome, or Alzheimer's disease. In the present study, we used the KCC2 Cre-lox knockdown system to investigate the role of KCC2 in synaptic plasticity and memory formation in adult mice. Tamoxifen-induced conditional deletion of KCC2 in glutamatergic neurons of the forebrain was performed at 3 months of age and resulted in spatial and nonspatial learning impairment. On brain slices, the stimulation of Schaffer collaterals by a theta burst induced long-term potentiation (LTP). The lack of KCC2 did not affect potentiation of field excitatory postsynaptic potentials (fEPSP) measured in the stratum radiatum (dendrites) but increased population spike (PS) amplitudes measured in the CA1 somatic layer, suggesting a reinforcement of the EPSP-PS potentiation, i.e., an increased ability of EPSPs to generate action potentials. At the cellular level, KCC2 deletion induced a positive shift in the reversal potential of GABAAR-driven Cl- currents (EGABA), suggesting an intracellular accumulation of chloride subsequent to the downregulation of KCC2. After treatment with bumetanide, an antagonist of the Na+-K+-Cl- cotransporter NKCC1, spatial memory impairment, chloride accumulation, and EPSP-PS potentiation were rescued in mice lacking KCC2. The presented results emphasize the importance of chloride equilibrium and GABA-inhibiting ability in synaptic plasticity and memory formation.
Collapse
|
66
|
Obot P, Subah G, Schonwald A, Pan J, Velíšek L, Velíšková J, Stanton PK, Scemes E. Astrocyte and Neuronal Panx1 Support Long-Term Reference Memory in Mice. ASN Neuro 2023; 15:17590914231184712. [PMID: 37365910 DOI: 10.1177/17590914231184712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Pannexin 1 (Panx1) is an ubiquitously expressed protein that forms plasma membrane channels permeable to anions and moderate-sized signaling molecules (e.g., ATP, glutamate). In the nervous system, activation of Panx1 channels has been extensively shown to contribute to distinct neurological disorders (epilepsy, chronic pain, migraine, neuroAIDS, etc.), but knowledge of the extent to which these channels have a physiological role remains restricted to three studies supporting their involvement in hippocampus dependent learning. Given that Panx1 channels may provide an important mechanism for activity-dependent neuron-glia interaction, we used Panx1 transgenic mice with global and cell-type specific deletions of Panx1 to interrogate their participation in working and reference memory. Using the eight-arm radial maze, we show that long-term spatial reference memory, but not spatial working memory, is deficient in Panx1-null mice and that both astrocyte and neuronal Panx1 contribute to the consolidation of long-term spatial memory. Field potential recordings in hippocampal slices of Panx1-null mice revealed an attenuation of both long-term potentiation (LTP) of synaptic strength and long-term depression (LTD) at Schaffer collateral-CA1 synapses without alterations of basal synaptic transmission or pre-synaptic paired-pulse facilitation. Our results implicate both neuronal and astrocyte Panx1 channels as critical players for the development and maintenance of long-term spatial reference memory in mice.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Emergency Medicine, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Galadu Subah
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Antonia Schonwald
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Jian Pan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Pediatrics, Penn State Hershey Medical Center, Hershey, PA, USA
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Obstetrics and Gynecology, New York Medical College, Valhalla, NY, USA
| | - Patric K Stanton
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Eliana Scemes
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
67
|
Steigerwald R, Chou T, Furukawa H, Wünsch B. GluN2A-Selective NMDA Receptor Antagonists: Mimicking the U-Shaped Bioactive Conformation of TCN-201 by a [2.2]Paracyclophane System. ChemMedChem 2022; 17:e202200484. [PMID: 36169098 PMCID: PMC9828697 DOI: 10.1002/cmdc.202200484] [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: 09/06/2022] [Revised: 09/23/2022] [Indexed: 01/19/2023]
Abstract
Under physiological conditions, N-Methyl-D-Aspartate (NMDA) receptors play a crucial role for synaptic plasticity, long-term potentiation and long-term depression. However, overactivation of NMDA receptors can result in excitotoxicity, which is associated with various neurological and neurodegenerative diseases. The physiological properties of NMDA receptors are strongly dependent on the GluN2 subunit incorporated into the heterotetrameric NMDA receptor. Therefore, subtype selective NMDA receptor modulators are of high interest. Since prototypical GluN2A-NMDA receptor antagonists TCN-201 and its MPX-analogs adopt a U-shaped conformation within the binding pocket, paracyclophanes were designed containing the phenyl rings in an already parallel orientation. Docking studies of the designed paracyclophanes show a similar binding pose as TCN-201. [2.2]Paracyclophanes with a benzoate or benzamide side chain were prepared in four-step synthesis, respectively, starting with a radical bromination in benzylic 1-position of [2.2]paracyclophane. In two-electrode voltage clamp experiments using Xenopus laevis oocytes transfected with cRNAs for the GluN1-4a and GluN2A subunits, the esters and amides (conc. 10 μM) did not show considerable inhibition of ion flux. It can be concluded that the GluN2A-NMDA receptor does not accept ligands with a paracyclophane scaffold functionalized in benzylic 1-position, although docking studies had revealed promising binding poses for benzoic acid esters and benzamides.
Collapse
Affiliation(s)
- Ruben Steigerwald
- Chemical biology of ion channels (Chembion)Westfälische Wilhelms-Universität MünsterCorrensstraße 4848149MünsterGermany,Institut für Pharmazeutische und Medizinische ChemieWestfälische Wilhelms-Universität MünsterCorrensstraße 4848149MünsterGermany
| | - Tsung‐Han Chou
- W.M. Keck Structural Biology LaboratoryCold Spring Harbor LaboratoryNew YorkNY11724USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology LaboratoryCold Spring Harbor LaboratoryNew YorkNY11724USA
| | - Bernhard Wünsch
- Chemical biology of ion channels (Chembion)Westfälische Wilhelms-Universität MünsterCorrensstraße 4848149MünsterGermany,Institut für Pharmazeutische und Medizinische ChemieWestfälische Wilhelms-Universität MünsterCorrensstraße 4848149MünsterGermany
| |
Collapse
|
68
|
Pitzer EM, Sugimoto C, Regan SL, Gudelsky GA, Williams MT, Vorhees CV. Developmental deltamethrin: Sex-specific hippocampal effects in Sprague Dawley rats. Curr Res Toxicol 2022; 3:100093. [PMID: 36393872 PMCID: PMC9661443 DOI: 10.1016/j.crtox.2022.100093] [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: 08/16/2022] [Revised: 10/04/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Pyrethroid pesticides are widely used and can cause long-term effects after early exposure. Epidemiological and animal studies reveal associations between pyrethroid exposure and altered cognition following prenatal and/or neonatal exposure. However, little is known about the cellular effects of such exposure. Sprague Dawley rats were gavaged with 0 or 1.0 mg/kg deltamethrin (DLM), a Type II pyrethroid, in corn oil (dose volume 5 mL/kg) once per day from postnatal day (P) 3-20 and assessed shortly after dosing ended or as adults. No effects of DLM exposure were found on striatal dopaminergic markers, nor on AMPA receptor subunits or on NMDA-NR1. However, DLM increased NMDA-NR2A and decreased NMDA-NR2B levels in the hippocampus, in males but not females. Additionally, adult hippocampal CA1 long-term potentiation was increased in DLM-treated males but not females. Potassium stimulated extracellular glutamate release in the hippocampus was not affected using in vivo microdialysis. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) showed increased apoptotic cells in the dentate gyrus of male rats, in the absence of changes in cleaved caspase-3 at P21. Proinflammatory cytokines interferon gamma trended up in striatum, interleukin-1β trended down in nucleus accumbens, IL-13 trended up in hippocampus, and keratinocyte chemoattractant/human growth-regulated oncogene (KC/GRO or CXCL1) was significantly increased in the hippocampus in male DLM-treated rats on P20. The data point to the developing hippocampus as a susceptible region to DLM-induced adverse effects.
Collapse
Affiliation(s)
- Emily M. Pitzer
- Dept. of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Chiho Sugimoto
- Dept. of Physiology, Michigan State University, 766 Service Rd. 5401 Interdisciplinary Science and Technology Building, East Lansing, MI 48824, USA
| | - Samantha L. Regan
- Dept. of Human Genetics, University of Michigan Medical Center, 3703 Med Sci II, 1241 E. Catherine St., Ann Arbor, MI 48109-5618, USA
| | - Gary A. Gudelsky
- College of Pharmacy, Div. of Pharmaceutical Sciences, 3212 Medical Sciences Building, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Michael T. Williams
- Dept. of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Charles V. Vorhees
- Dept. of Pediatrics, University of Cincinnati College of Medicine, and Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
69
|
Kelvington BA, Nickl-Jockschat T, Abel T. Neurobiological insights into twice-exceptionality: Circuits, cells, and molecules. Neurobiol Learn Mem 2022; 195:107684. [PMID: 36174887 PMCID: PMC9888516 DOI: 10.1016/j.nlm.2022.107684] [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: 10/26/2021] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 02/02/2023]
Abstract
Twice-exceptional learners face a unique set of challenges arising from the intersection of extraordinary talent and disability. Neurobiology research has the capacity to complement pedagogical research and provide support for twice-exceptional learners. Very few studies have attempted to specifically address the neurobiological underpinnings of twice-exceptionality. However, neurobiologists have built a broad base of knowledge in nervous system function spanning from the level of neural circuits to the molecular basis of behavior. It is known that distinct neural circuits mediate different neural functions, which suggests that 2e learning may result from enhancement in one circuit and disruption in another. Neural circuits are known to adapt and change in response to experience, a cellular process known as neuroplasticity. Plasticity is controlled by a bidirectional connection between the synapse, where neural signals are received, and the nucleus, where regulated gene expression can return to alter synaptic function. Complex molecular mechanisms compose this connection in distinct neural circuits, and genetic alterations in these mechanisms are associated with both memory enhancements and psychiatric disorder. Understanding the consequences of these changes at the molecular, cellular, and circuit levels will provide critical insights into the neurobiological bases of twice-exceptionality.
Collapse
Affiliation(s)
- Benjamin A Kelvington
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
70
|
Sivakumar S, Ghasemi M, Schachter SC. Targeting NMDA Receptor Complex in Management of Epilepsy. Pharmaceuticals (Basel) 2022; 15:ph15101297. [PMID: 36297409 PMCID: PMC9609646 DOI: 10.3390/ph15101297] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are widely distributed in the central nervous system (CNS) and play critical roles in neuronal excitability in the CNS. Both clinical and preclinical studies have revealed that the abnormal expression or function of these receptors can underlie the pathophysiology of seizure disorders and epilepsy. Accordingly, NMDAR modulators have been shown to exert anticonvulsive effects in various preclinical models of seizures, as well as in patients with epilepsy. In this review, we provide an update on the pathologic role of NMDARs in epilepsy and an overview of the NMDAR antagonists that have been evaluated as anticonvulsive agents in clinical studies, as well as in preclinical seizure models.
Collapse
Affiliation(s)
- Shravan Sivakumar
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- Correspondence: (M.G.); (S.C.S.)
| | - Steven C. Schachter
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02114, USA
- Consortia for Improving Medicine with Innovation & Technology (CIMIT), Boston, MA 02114, USA
- Correspondence: (M.G.); (S.C.S.)
| |
Collapse
|
71
|
Coulibaly AP. Neutrophil modulation of behavior and cognition in health and disease: The unexplored role of an innate immune cell. Immunol Rev 2022; 311:177-186. [PMID: 35924463 PMCID: PMC9804154 DOI: 10.1111/imr.13123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Behavior and cognition are multifaceted processes influenced by genetics, synaptic plasticity, and neuronal connectivity. Recent reports have demonstrated that peripheral inflammation and peripheral immune cells play important roles in the preservation and deterioration of behavior/cognition under various conditions. Indeed, several studies show that the activity of peripheral immune cells can be critical for normal cognitive function. Neutrophils are the most abundant immune cells in the mammalian system. Their activation is critical to the initiation of the inflammatory process and critical for wound healing. Neutrophils are the first cells to be activated and recruited to the central nervous system in both injury and disease. However, our understanding of the role these cells play in behavior and cognition is limited. The present review will summarize what is currently known about the effect the activation of these cells has on various behaviors and cognitive processes.
Collapse
Affiliation(s)
- Aminata P. Coulibaly
- Department of NeuroscienceRockefeller Neuroscience InstituteWest Virginia UniversityMorgantownWest VirginiaUSA
| |
Collapse
|
72
|
Mohammed RA, Sayed RH, El-Sahar AE, Khattab MA, Saad MA. Insights into the role of pERK1/2 signaling in post-cerebral ischemia reperfusion sexual dysfunction in rats. Eur J Pharmacol 2022; 933:175258. [PMID: 36096157 DOI: 10.1016/j.ejphar.2022.175258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022]
Abstract
The purpose of the present study was to investigate the effects of ERK1/2 inhibition on both the amygdala and hippocampal structures, and to investigate its role in regulating memory for sexual information. This study utilized a cerebral ischemia reperfusion (IR) model to produce a stressful brain condition that highlights the possible involvement of a hippocampal GC/pERK1/2/BDNF pathway in the resulting sexual consequences of this ailment. Male Wistar rats were divided into four groups: (1) sham; (2) IR: subjected to 45 min of ischemia followed by 48 h of reperfusion; (3) PD98059: received PD98059 at 0.3 mg/kg, i.p.; (4) IR + PD98059. This study provides new evidence for cerebral IR-induced amygdala injury and the sexual impairments that are associated with motor and cognitive deficits in rats. These findings were correlated with histopathological changes that are defined by extensive neuronal loss in both the hippocampus and the amygdala. The current study postulated that the ERK inhibitor PD98059 could reverse IR-induced injury in the amygdala as well as reversing IR-induced sexual impairments. This hypothesis is supported by the ability of PD98059 to: (1) restore luteinizing hormone and testosterone levels; (2) increase sexual arousal and copulatory performance (as evidenced by modulating mount, intromission, ejaculation latencies, and post-ejaculatory intervals); (3) improve the histological profile in the amygdala that is associated with reduced glutamate levels, c-Fos expression, and elevated gamma aminobutyric acid levels. In conclusion, the present findings introduce pERK1/2 inhibition as a possible strategy for enhancing sexual activity in survivors of IR.
Collapse
Affiliation(s)
- Reham A Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Ayman E El-Sahar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamed A Khattab
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Muhammed A Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, 4184, United Arab Emirates
| |
Collapse
|
73
|
Nicotine alleviates alcohol-induced memory and long-term potentiation impairment. Neurosci Lett 2022; 786:136813. [PMID: 35878655 DOI: 10.1016/j.neulet.2022.136813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022]
Abstract
Alcohol and nicotine are routinely abused together. There is one plausible explanation that comorbidity can alleviate alcohol-induced cognitive impairment. However, the mechanism involved is not known. The aim of this report was to evaluate the interactive effects of alcohol coadministration with nicotine on hippocampal memory and long-term potentiation (LTP). C57BL/6 mice were distributed into 4 treatment groups: control, alcohol, nicotine, and alcohol plus nicotine. All mice received tap water or alcohol solution and saline or nicotine. In water maze test, the alcohol group showed significant decreases in hippocampus function and acquisition training than control, nicotine, and combined treatment groups. The alcohol group also showed a significantly shorter latency in entering the foot-shock compartment than control, nicotine, and combined treatment groups in a passive avoidance test. Theta burst stimulation was adopted to induce concrete LTP in CA1 field recording using hippocampal slice. Alcohol alone administration failed to maintain LTP. Nicotine alone administration did not alter hippocampal LTP. There were no negative effects of alcohol on hippocampal LTP in mice administrated with nicotine. The current study successfully demonstrated beneficial effects of nicotine on alcohol induced memory impairment accompanied by hippocampal LTP impairment after one week of co-administration and one-day withdrawal.
Collapse
|
74
|
Liu R, Bai L, Liu M, Wang R, Wu Y, Li Q, Ba Y, Zhang H, Zhou G, Yu F, Huang H. Combined exposure of lead and high-fat diet enhanced cognitive decline via interacting with CREB-BDNF signaling in male rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 304:119200. [PMID: 35364187 DOI: 10.1016/j.envpol.2022.119200] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/03/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
The health risks to populations induced by lead (Pb) and high-fat diets (HFD) have become a global public health problem. Pb and HFD often co-exist and are co-occurring risk factors for cognitive impairment. This study investigates effect of combined Pb and HFD on cognitive function, and explores the underlying mechanisms in terms of regulatory components of synaptic plasticity and insulin signaling pathway. We showed that the co-exposure of Pb and HFD further increased blood Pb levels, caused body weight loss and dyslipidemia. The results from Morris water maze (MWM) test and Nissl staining disclosed that Pb and HFD each contributed to cognitive deficits and neuronal damage and combined exposure enhanced this toxic injury. Pb and HFD decreased the levels of synapsin-1, GAP-43 and PSD-95 protein related to synaptic properties and SIRT1, NMDARs, phosphorylated CREB and BDNF related to synaptic plasticity regulatory, and these decreases was greater when combined exposure. Additionally, we revealed that Pb and HFD promoted IRS-1 phosphorylation and subsequently reduced downstream PI3K-Akt kinases phosphorylation in hippocampus and cortex of rats, and this process was aggravated when co-exposure. Collectively, our data suggested that combined exposure of Pb and HFD enhanced cognitive deficits, pointing to additive effects in rats than the individual stress effects related to multiple signaling pathways with CREB-BDNF signaling as the hub. This study emphasizes the need to evaluate the effects of mixed exposures on brain function in realistic environment and to better inform prevention of neurological disorders via modulating central pathway, such as CREB/BDNF signaling.
Collapse
Affiliation(s)
- Rundong Liu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Bai
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengchen Liu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruike Wang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yingying Wu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Qiong Li
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Ba
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Huizhen Zhang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Guoyu Zhou
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Fangfang Yu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Huang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
75
|
Sambu S, Hemaram U, Murugan R, Alsofi AA. Toxicological and Teratogenic Effect of Various Food Additives: An Updated Review. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6829409. [PMID: 35782077 PMCID: PMC9249520 DOI: 10.1155/2022/6829409] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022]
Abstract
Scientific evidence is mounting that synthetic chemicals used as food additives may have harmful impacts on health. Food additives are chemicals that are added to food to keep it from spoiling, as well as to improve its colour and taste. Some are linked to negative health impacts, while others are healthy and can be ingested with little danger. According to several studies, health issues such as asthma, attention deficit hyperactivity disorder (ADHD), heart difficulties, cancer, obesity, and others are caused by harmful additives and preservatives. Some food additives may interfere with hormones and influences growth and development. It is one of the reasons why so many children are overweight. Children are more likely than adults to be exposed to these types of dietary intakes. Several food additives are used by women during pregnancy and breast feeding that are not fully safe. We must take specific precaution to avoid consuming dangerous compounds before they begin to wreak havoc on our health. This study is intended to understand how the preservatives induce different health problem in the body once it is consumed. This review focuses on some specific food additives such as sodium benzoate, aspartame, tartrazine, carrageenan, and potassium benzoate, as well as vitamin A. Long-term use of food treated with the above-mentioned food preservatives resulted in teratogenicity and other allergens, according to the study. Other health issues can be avoided in the future by using natural food additives derived from plants and other natural sources.
Collapse
Affiliation(s)
- Saseendran Sambu
- Department of Food Technology, Faculty of Life and Allied Health Sciences, Ramaiah University of Applied Sciences, Bengaluru, Karnataka, India
| | - Urmila Hemaram
- Department of Food Technology, Faculty of Life and Allied Health Sciences, Ramaiah University of Applied Sciences, Bengaluru, Karnataka, India
| | - Rajadurai Murugan
- Department of Food Technology, Faculty of Life and Allied Health Sciences, Ramaiah University of Applied Sciences, Bengaluru, Karnataka, India
| | - Ahmed A. Alsofi
- Department of Pharmacy, Faculty of Medical Sciences, Aljanad University for Science and Technology, Taiz, Yemen
| |
Collapse
|
76
|
Ng DQ, Chan D, Agrawal P, Zhao W, Xu X, Acharya M, Chan A. EVIDENCE OF BRAIN-DERIVED NEUROTROPHIC FACTOR IN AMELIORATING CANCER-RELATED COGNITIVE IMPAIRMENT: A SYSTEMATIC REVIEW OF HUMAN STUDIES. Crit Rev Oncol Hematol 2022; 176:103748. [PMID: 35718064 DOI: 10.1016/j.critrevonc.2022.103748] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 12/27/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays an essential role in neurogenesis and neuroplasticity and may be a key protein in cancer-related cognitive impairment (CRCI). This systematic review assessed the relationship between BDNF biomarkers and neurocognitive outcomes in cancer patients and survivors. A search in PubMed, Scopus, and PsycINFO yielded 638 articles, of which 26 were eligible. Fourteen (54%)studied BDNF protein levels while 15 (58%) analyzed BDNF rs6265 polymorphism. Of the nine observational studies reporting BDNF plasma/serum levels, five (56%) exhibited a positive association between BDNF and cognitive function. One study reported intra-tumoral BDNF levels that were negatively associated with memory. For rs6265, three (20%) of 15 studies reported an association with cognitive function with inconsistent directions. Among seven neuroimaging studies, three (43%) demonstrated an effect of BDNF on brain function and structure. These results suggest that BDNF is a potential monitoring biomarker and druggable target for CRCI.
Collapse
Affiliation(s)
- Ding Quan Ng
- Department of Clinical Pharmacy Practice, University of California, Irvine, Irvine, California, United States of America; Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Daniella Chan
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Parisa Agrawal
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Weian Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America; Department of Biological Chemistry, University of California, Irvine, Irvine, California, USA; Department of Biomedical Engineering, The Henry Samueli School of Engineering, University of California, Irvine, Irvine, California, USA; Institute for Immunology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, California, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, California, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California, USA; The Center for Neural Circuit Mapping, University of California Irvine, Irvine, California, USA
| | - Munjal Acharya
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California, USA; Department of Radiation Oncology, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, University of California, Irvine, Irvine, California, United States of America; Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
77
|
Gupta S, Moreno AJ, Wang D, Leon J, Chen C, Hahn O, Poon Y, Greenberg K, David N, Wyss-Coray T, Raftery D, Promislow DEL, Dubal DB. KL1 Domain of Longevity Factor Klotho Mimics the Metabolome of Cognitive Stimulation and Enhances Cognition in Young and Aging Mice. J Neurosci 2022; 42:4016-4025. [PMID: 35428698 PMCID: PMC9097772 DOI: 10.1523/jneurosci.2458-21.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/02/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
Cognitive deficits are a major biomedical challenge-and engagement of the brain in stimulating tasks improves cognition in aged individuals (Wilson et al., 2002; Gates et al., 2011) and rodents (Aidil-Carvalho et al., 2017), through unknown mechanisms. Whether cognitive stimulation alters specific metabolic pathways in the brain is unknown. Understanding which metabolic processes are involved in cognitive stimulation is important because it could lead to pharmacologic intervention that promotes biological effects of a beneficial behavior, toward the goal of effective medical treatments for cognitive deficits. Here we show using male mice that cognitive stimulation induced metabolic remodeling of the mouse hippocampus, and that pharmacologic treatment with the longevity hormone α-klotho (KL), mediated by its KL1 domain, partially mimicked this alteration. The shared, metabolic signature shared between cognitive stimulation and treatment with KL or KL1 closely correlated with individual mouse cognitive performance, indicating a link between metabolite levels and learning and memory. Importantly, the treatment of mice with KL1, an endogenous circulating factor that more closely mimicked cognitive stimulation than KL, acutely increased synaptic plasticity, a substrate of cognition. KL1 also improved cognition, itself, in young mice and countered deficits in old mice. Our data show that treatments or interventions mimicking the hippocampal metabolome of cognitive stimulation can enhance brain functions. Further, we identify the specific domain by which klotho promotes brain functions, through KL1, a metabolic mimic of cognitive stimulation.SIGNIFICANCE STATEMENT Cognitive deficits are a major biomedical challenge without truly effective pharmacologic treatments. Engaging the brain through cognitive tasks benefits cognition. Mimicking the effects of such beneficial behaviors through pharmacological treatment represents a highly valuable medical approach to treating cognitive deficits. We demonstrate that brain engagement through cognitive stimulation induces metabolic remodeling of the hippocampus that was acutely recapitulated by the longevity factor klotho, mediated by its KL1 domain. Treatment with KL1, a close mimic of cognitive stimulation, enhanced cognition and countered cognitive aging. Our findings shed light on how cognition metabolically alters the brain and provide a plausible therapeutic intervention for mimicking these alterations that, in turn, improves cognition in the young and aging brain.
Collapse
Affiliation(s)
- Shweta Gupta
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Arturo J Moreno
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Dan Wang
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Julio Leon
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Chen Chen
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5101
| | - Yan Poon
- Unity Biotechnology, Inc, South San Francisco 94080
| | | | | | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5101
- Veterans Administration Palo Alto Healthcare System, Palo Alto, California 94304-1207
- Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California 94305-5235
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California 94305-5235
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, Washington 98109-4714
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024
| | - Daniel E L Promislow
- Department of Lab Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195-7470
- Department of Biology, University of Washington, Seattle, Washington 98195-1800
| | - Dena B Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143-1207
| |
Collapse
|
78
|
Vasavda C, Semenza ER, Liew J, Kothari R, Dhindsa RS, Shanmukha S, Lin A, Tokhunts R, Ricco C, Snowman AM, Albacarys L, Pastore F, Ripoli C, Grassi C, Barone E, Kornberg MD, Dong X, Paul BD, Snyder SH. Biliverdin reductase bridges focal adhesion kinase to Src to modulate synaptic signaling. Sci Signal 2022; 15:eabh3066. [PMID: 35536885 PMCID: PMC9281001 DOI: 10.1126/scisignal.abh3066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Synapses connect discrete neurons into vast networks that send, receive, and encode diverse forms of information. Synaptic function and plasticity, the neuronal process of adapting to diverse and variable inputs, depend on the dynamic nature of synaptic molecular components, which is mediated in part by cell adhesion signaling pathways. Here, we found that the enzyme biliverdin reductase (BVR) physically links together key focal adhesion signaling molecules at the synapse. BVR-null (BVR-/-) mice exhibited substantial deficits in learning and memory on neurocognitive tests, and hippocampal slices in which BVR was postsynaptically depleted showed deficits in electrophysiological responses to stimuli. RNA sequencing, biochemistry, and pathway analyses suggested that these deficits were mediated through the loss of focal adhesion signaling at both the transcriptional and biochemical level in the hippocampus. Independently of its catalytic function, BVR acted as a bridge between the primary focal adhesion signaling kinases FAK and Pyk2 and the effector kinase Src. Without BVR, FAK and Pyk2 did not bind to and stimulate Src, which then did not phosphorylate the N-methyl-d-aspartate (NMDA) receptor, a critical posttranslational modification for synaptic plasticity. Src itself is a molecular hub on which many signaling pathways converge to stimulate NMDAR-mediated neurotransmission, thus positioning BVR at a prominent intersection of synaptic signaling.
Collapse
Affiliation(s)
- Chirag Vasavda
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Evan R. Semenza
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA
| | - Jason Liew
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ruchita Kothari
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ryan S. Dhindsa
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
| | - Shruthi Shanmukha
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anthony Lin
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Robert Tokhunts
- Department of Anesthesiology, Dartmouth–Hitchcock Medical Center, Lebanon, NH 03766, USA
| | - Cristina Ricco
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Adele M. Snowman
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren Albacarys
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Preclinical Neuroscience Lab, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Preclinical Neuroscience Lab, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Rome 00185, Italy
| | - Michael D. Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bindu D. Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Solomon H. Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
79
|
Grünenfelder DC, Navarro R, Wang H, Fastuca NJ, Butler JR, Reisman SE. Enantioselective Synthesis of (-)-10-Hydroxyacutuminine. Angew Chem Int Ed Engl 2022; 61:e202117480. [PMID: 35112449 DOI: 10.1002/anie.202117480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Indexed: 11/08/2022]
Abstract
An enantioselective synthesis of (-)-10-hydroxyacutuminine is reported. Central to our strategy is a photochemical [2+2] cycloaddition that forges two of the quaternary stereocenters present in the acutumine alkaloids. A subsequent retro-aldol/Dieckmann sequence furnishes the spirocyclic cyclopentenone. Efforts to chlorinate the acutumine scaffold at C10 under heterolytic or radical deoxychlorination conditions led to the synthesis of an unexpected cyclopropane-containing pentacycle.
Collapse
Affiliation(s)
- Denise C Grünenfelder
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Raul Navarro
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Haoxuan Wang
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Nicholas J Fastuca
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - John R Butler
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sarah E Reisman
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
80
|
He X, Wang Y, Zhou G, Yang J, Li J, Li T, Hu H, Ma H. A Critical Role for γCaMKII in Decoding NMDA Signaling to Regulate AMPA Receptors in Putative Inhibitory Interneurons. Neurosci Bull 2022; 38:916-926. [PMID: 35290589 PMCID: PMC9352831 DOI: 10.1007/s12264-022-00840-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
CaMKII is essential for long-term potentiation (LTP), a process in which synaptic strength is increased following the acquisition of information. Among the four CaMKII isoforms, γCaMKII is the one that mediates the LTP of excitatory synapses onto inhibitory interneurons (LTPE→I). However, the molecular mechanism underlying how γCaMKII mediates LTPE→I remains unclear. Here, we show that γCaMKII is highly enriched in cultured hippocampal inhibitory interneurons and opts to be activated by higher stimulating frequencies in the 10-30 Hz range. Following stimulation, γCaMKII is translocated to the synapse and becomes co-localized with the postsynaptic protein PSD-95. Knocking down γCaMKII prevents the chemical LTP-induced phosphorylation and trafficking of AMPA receptors (AMPARs) in putative inhibitory interneurons, which are restored by overexpression of γCaMKII but not its kinase-dead form. Taken together, these data suggest that γCaMKII decodes NMDAR-mediated signaling and in turn regulates AMPARs for expressing LTP in inhibitory interneurons.
Collapse
Affiliation(s)
- Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yang Wang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Guangjun Zhou
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jing Yang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jiarui Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Tao Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Hailan Hu
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
- Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China.
- Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
81
|
Collingridge GL, Monaghan DT. The continually evolving role of NMDA receptors in neurobiology and disease. Neuropharmacology 2022; 210:109042. [DOI: 10.1016/j.neuropharm.2022.109042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
82
|
Grünenfelder DC, Navarro R, Wang H, Fastuca NJ, Butler JR, Reisman SE. Enantioselective Synthesis of (−)‐10‐Hydroxyacutuminine. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202117480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Denise C. Grünenfelder
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering Division of Chemistry and Chemical Engineering California Institute of Technology Pasadena CA 91125 USA
| | - Raul Navarro
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering Division of Chemistry and Chemical Engineering California Institute of Technology Pasadena CA 91125 USA
| | - Haoxuan Wang
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering Division of Chemistry and Chemical Engineering California Institute of Technology Pasadena CA 91125 USA
| | - Nicholas J. Fastuca
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering Division of Chemistry and Chemical Engineering California Institute of Technology Pasadena CA 91125 USA
| | - John R. Butler
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering Division of Chemistry and Chemical Engineering California Institute of Technology Pasadena CA 91125 USA
| | - Sarah E. Reisman
- The Warren and Katharine Schlinger Laboratory of Chemistry and Chemical Engineering Division of Chemistry and Chemical Engineering California Institute of Technology Pasadena CA 91125 USA
| |
Collapse
|
83
|
Caruso TJ, Armstrong-Carter E, Rama A, Neiman N, Taylor K, Madill M, Lawrence K, Hemphill SF, Guo N, Domingue BW. The Physiologic and Emotional Effects of 360-Degree Video Simulation on Head-Mounted Display Versus In-Person Simulation: A Noninferiority, Randomized Controlled Trial. Simul Healthc 2022; 17:e105-e112. [PMID: 34120135 DOI: 10.1097/sih.0000000000000587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION A key simulation component is its capability to elicit physiological changes, improving recall. The primary aim was to determine whether parasympathetic responses to head-mounted display simulations (HMDs) were noninferior to in-person simulations. The secondary aims explored sympathetic and affective responses and learning effectiveness. METHODS The authors conducted a noninferiority trial. Hospital providers who did not use chronotropic medications, have motion sickness, or have seizures were included. The authors randomized participants to in-person or HMD simulation. Biometric sensors collected respiratory sinus arrhythmia and skin conductance levels to measure parasympathetic and sympathetic states at baseline, during, and after the simulation. Affect was measured using a schedule. The authors measured 3-month recall of learning points and used split-plot analysis of variance and Mann-Whitney U tests to analyze. RESULTS One hundred fifteen participants qualified, and the authors analyzed 56 in each group. Both groups experienced a significant change in mean respiratory sinus arrhythmia from baseline to during and from during to afterward. The difference of change between the groups from baseline to during was 0.134 (95% confidence interval = 0.142 to 0.410, P = 0.339). The difference of change from during the simulation to after was -0.060 (95% confidence interval = -0.337 to 0.217, P = 0.670). Noninferiority was not established for either period. Sympathetic arousal did not occur in either group. Noninferiority was not established for the changes in affect that were demonstrated. The mean scores of teaching effectiveness and achievement scores were not different. CONCLUSIONS Although a parasympathetic and affective response to the video simulation on an HMD did occur, it was not discernibly noninferior to in-person in this study.
Collapse
Affiliation(s)
- Thomas J Caruso
- From the Department of Anesthesiology, Perioperative, and Pain Medicine (T.J.C., A.R., N.N., K.T., N.G.), Stanford University School of Medicine; Stanford University Graduate School of Education (E.A.-C., B.D.), Stanford, CA; University of Pittsburgh School of Medicine (M.M.), Pittsburgh, PA; Department of Internal Medicine, Legacy Emanuel Medical Center (K.L.), Portland, OR; and Stanford University School of Medicine (S.F.H.), Stanford, CA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Keith RE, Ogoe RH, Dumas TC. Behind the scenes: Are latent memories supported by calcium independent plasticity? Hippocampus 2022; 32:73-88. [PMID: 33905147 PMCID: PMC8548406 DOI: 10.1002/hipo.23332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 02/03/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) can be considered to be the de facto "plasticity" receptors in the brain due to their central role in the activity-dependent modification of neuronal morphology and synaptic transmission. Since the 1980s, research on NMDARs has focused on the second messenger properties of calcium and the downstream signaling pathways that mediate alterations in neural form and function. Recently, NMDARs were shown to drive activity-dependent synaptic plasticity without calcium influx. How this "nonionotropic" plasticity occurs in vitro is becoming clearer, but research on its involvement in behavior and cognition is in its infancy. There is a partial overlap in the downstream signaling molecules that are involved in ionotropic and nonionotropic NMDAR-dependent plasticity. Given this, and prior studies of the cognitive impacts of ionotropic NMDAR plasticity, a preliminary model explaining how NMDAR nonionotropic plasticity affects learning and memory can be established. We hypothesize that nonionotropic NMDAR plasticity takes part in latent memory encoding in immature rodents through nonassociative depression of synaptic efficacy, and possibly shrinking of dendritic spines. Further, the late postnatal alteration in NMDAR composition in the hippocampus appears to reduce nonionotropic signaling and remove a restriction on memory retrieval. This framework substantially alters the canonical model of NMDAR involvement in spatial cognition and hippocampal maturation and provides novel and exciting inroads for future studies.
Collapse
Affiliation(s)
- Rachel E. Keith
- Interdisciplinary Program in Neuroscience, College of Science, George Mason University, Fairfax, Virginia
| | - Richard H. Ogoe
- Department of Psychology, College of Humanities and Social Sciences, George Mason University, Fairfax, Virginia
| | - Theodore C. Dumas
- Interdisciplinary Program in Neuroscience, College of Science, George Mason University, Fairfax, Virginia,Department of Psychology, College of Humanities and Social Sciences, George Mason University, Fairfax, Virginia
| |
Collapse
|
85
|
George H, Bashir ZI, Hussain S. Impaired hippocampal NMDAR-LTP in a transgenic model of NSUN2-deficiency. Neurobiol Dis 2022; 163:105597. [PMID: 34954053 DOI: 10.1016/j.nbd.2021.105597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/09/2021] [Accepted: 12/19/2021] [Indexed: 11/28/2022] Open
Abstract
Biallelic loss-of-function NSUN2 mutations have recently been associated with cases of Autism Spectrum Condition (ASC), and NSun2-deficiency was also previously shown to cause a severe autosomal recessive intellectually disability disorder syndrome in which patients can sometimes display autistic behaviour. It has been demonstrated that NSUN2 can control protein synthesis rates via direct regulation of RNA methylation, and it is therefore of interest that other studies have suggested protein synthesis-dependent synaptic plasticity dysregulation as a mechanism for learning difficulties in various other autism-expressing conditions and disorders. Here we investigated NMDAR-LTP in a murine transgenic model harbouring loss-of-function mutation in the NSun2 gene and find an impairment of a protein synthesis-dependent form of this synaptic plasticity pathway. Our findings support the idea that NMDAR-LTP mis-regulation may represent a previously underappreciated mechanism associated with autism phenotypes.
Collapse
Affiliation(s)
- Harry George
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK; Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Zafar I Bashir
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK.
| | - Shobbir Hussain
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK.
| |
Collapse
|
86
|
Lee Y, Bortolotto ZA, Bradley CA, Sanderson TM, Zhuo M, Kaang BK, Collingridge GL. The GSK-3 Inhibitor CT99021 Enhances the Acquisition of Spatial Learning and the Accuracy of Spatial Memory. Front Mol Neurosci 2022; 14:804130. [PMID: 35153671 PMCID: PMC8829050 DOI: 10.3389/fnmol.2021.804130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK-3) is a Ser/Thr protein kinase that regulates many cellular processes, including synaptic plasticity. Previously, we reported that inhibition of GSK-3 prevents the induction of one of the major forms of synaptic plasticity, N-methyl-D-aspartate receptor (NMDAR)-dependent long-term depression (LTD), in hippocampal slices. In the present study, we have investigated the effects of inhibiting GSK-3 on learning and memory in healthy naïve animals. Systemic administration of a highly selective GSK-3 inhibitor, CT99021, reversibly blocked NMDAR-dependent LTD in the CA1 region of the hippocampus in anesthetized adult mice. In behavioral tasks, CT99021 had no effect on locomotor activity, anxiety, hippocampus-dependent contextual fear memory, and hippocampus-dependent reversal learning. However, CT99021 facilitated the rate of learning in the Morris water maze (MWM) and T-maze and enhanced the accuracy of long-term spatial memory in the MWM. These findings suggest that GSK-3 regulates the accuracy of spatial memory acquisition and recall.
Collapse
Affiliation(s)
- Yeseul Lee
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Zuner A. Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Clarrisa A. Bradley
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Genes and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Thomas M. Sanderson
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Min Zhuo
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Bong-Kiun Kaang
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- *Correspondence: Bong-Kiun Kaang,
| | - Graham L. Collingridge
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
- Graham L. Collingridge,
| |
Collapse
|
87
|
Hernández-Mercado K, Zepeda A. Morris Water Maze and Contextual Fear Conditioning Tasks to Evaluate Cognitive Functions Associated With Adult Hippocampal Neurogenesis. Front Neurosci 2022; 15:782947. [PMID: 35046769 PMCID: PMC8761726 DOI: 10.3389/fnins.2021.782947] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
New neurons are continuously generated and functionally integrated into the dentate gyrus (DG) network during the adult lifespan of most mammals. The hippocampus is a crucial structure for spatial learning and memory, and the addition of new neurons into the DG circuitry of rodents seems to be a key element for these processes to occur. The Morris water maze (MWM) and contextual fear conditioning (CFC) are among the most commonly used hippocampus-dependent behavioral tasks to study episodic-like learning and memory in rodents. While the functional contribution of adult hippocampal neurogenesis (AHN) through these paradigms has been widely addressed, results have generated controversial findings. In this review, we analyze and discuss possible factors in the experimental methods that could explain the inconsistent results among AHN studies; moreover, we provide specific suggestions for the design of more sensitive protocols to assess AHN-mediated learning and memory functions.
Collapse
Affiliation(s)
- Karina Hernández-Mercado
- Departamento de Medicina Genómica y Toxicológia Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicológia Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
88
|
Da Rocha RVO, Martins MIM, Antunes FTT, Martins MG, Klein AB, Corrêa DS, de Souza AH. Behavioral, Oxidative, and Biochemical Effects of Omega-3 on an Ovariectomized Rat Model of Menopause. J Menopausal Med 2022; 27:132-140. [PMID: 34989186 PMCID: PMC8738853 DOI: 10.6118/jmm.21016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/13/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Menopause induces changes in neuronal transmission, leading to anxiety and depression. Changes in the brain's glutamate levels cause psychological behavior in postmenopausal women. Omega-3 has been studied to improve some of these behaviors. METHODS Twenty-four female Wistar rats were divided into four groups: sham-operated treated with water (SO-W), sham-operated treated with omega-3 (SO-O), ovariectomized (OVX) treated with water (OVX-W), and bilateral OVX treated with omega-3 (OVX-O). These treatments were performed for 20 days via gavage, before and after surgery, totaling 40 days. RESULTS In the forced swimming, elevated plus-maze, and open field tests to assess behaviors, such as depression and anxiety, omega-3 improved these behaviors in both treated groups. The levels of thiobarbituric acid reactive substances (TBARS) in the brain were not different between the groups; however, there was a significant decrease in the catalase activity in the SO-O group compared with the SO-W group (P < 0.05). The glutamate level in the cerebrospinal fluid (CSF) was elevated in the SO-O group (P < 0.001) but not in the OVX-W or OVX-O groups. CONCLUSIONS These results bring novel data when related to the glutamatergic system in the SO-O group. This has suggested that the action mechanism of omega-3 was not dependent on glutamate levels in the CSF of the OVX group, but it played a regulatory role in the sham-operated animals. To confirm this, more studies are needed to explore this field when relating to the estrogen and glutamate receptor changes in specific brain regions.
Collapse
Affiliation(s)
| | | | | | - Marcia Gerhardt Martins
- Graduate Program in Genetic and Applied Toxicology, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Adriane Belló Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Dione Silva Corrêa
- Graduate Program in Cellular and Molecular Biology, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Alessandra Hubner de Souza
- Graduate Program in Cellular and Molecular Biology, Lutheran University of Brazil (ULBRA), Canoas, Brazil.
| |
Collapse
|
89
|
OTHMAN MZ, HASSAN Z, CHE HAS AT. Morris water maze: a versatile and pertinent tool for assessing spatial learning and memory. Exp Anim 2022; 71:264-280. [PMID: 35314563 PMCID: PMC9388345 DOI: 10.1538/expanim.21-0120] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Since its development about 40 years ago (1981–2021), Morris water maze has turned into a very popular tool for assessing spatial learning and memory. Its many advantages have ensured its
pertinence to date. These include its effectiveness in evaluating hippocampal-dependent learning and memory, exemption from motivational differences across diverse experimental
manipulations, reliability in various cross-species studies, and adaptability to many experimental conditions with various test protocols. Nonetheless, throughout its establishment, several
experimental and analysis loopholes have galvanized researchers to assess ways in which it could be improved and adapted to fill this gap. Therefore, in this review, we briefly summarize
these developments since the early years of its establishment through to the most recent advancements in computerized analysis, offering more comprehensive analysis paradigms. In addition,
we discuss the adaptability of the Morris water maze across different test versions and analysis paradigms, providing suggestions with regard to the best paradigms for particular
experimental conditions. Hence, the proper selection of the experimental protocols, analysis paradigms, and consideration of the assay’s limitations should be carefully considered. Given
that appropriate measures are taken, with various adaptations made, the Morris water maze will likely remain a relevant tool to assess the mechanisms of spatial learning and memory.
Collapse
|
90
|
France G, Volianskis R, Ingram R, Bannister N, Rothärmel R, Irvine MW, Fang G, Burnell ES, Sapkota K, Costa BM, Chopra DA, Dravid SM, Michael-Titus AT, Monaghan DT, Georgiou J, Bortolotto ZA, Jane DE, Collingridge GL, Volianskis A. Differential regulation of STP, LTP and LTD by structurally diverse NMDA receptor subunit-specific positive allosteric modulators. Neuropharmacology 2022; 202:108840. [PMID: 34678377 PMCID: PMC8803579 DOI: 10.1016/j.neuropharm.2021.108840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 11/30/2022]
Abstract
Different types of memory are thought to rely on different types of synaptic plasticity, many of which depend on the activation of the N-Methyl-D Aspartate (NMDA) subtype of glutamate receptors. Accordingly, there is considerable interest in the possibility of using positive allosteric modulators (PAMs) of NMDA receptors (NMDARs) as cognitive enhancers. Here we firstly review the evidence that NMDA receptor-dependent forms of synaptic plasticity: short-term potentiation (STP), long-term potentiation (LTP) and long-term depression (LTD) can be pharmacologically differentiated by using NMDAR ligands. These observations suggest that PAMs of NMDAR function, depending on their subtype selectivity, might differentially regulate STP, LTP and LTD. To test this hypothesis, we secondly performed experiments in rodent hippocampal slices with UBP714 (a GluN2A/2B preferring PAM), CIQ (a GluN2C/D selective PAM) and UBP709 (a pan-PAM that potentiates all GluN2 subunits). We report here, for the first time, that: (i) UBP714 potentiates sub-maximal LTP and reduces LTD; (ii) CIQ potentiates STP without affecting LTP; (iii) UBP709 enhances LTD and decreases LTP. We conclude that PAMs can differentially regulate distinct forms of NMDAR-dependent synaptic plasticity due to their subtype selectivity. This article is part of the Neuropharmacology Special Issue on ‘Glutamate Receptors – NMDA receptors’. NMDAR-dependent STP, LTP and LTD can be dissociated pharmacologically GluN2A/2B PAM UBP714 potentiates LTP and reduces LTD GluN2C/D PAM CIQ potentiates STP without affecting LTP NMDAR pan-PAM UBP709 potentiates LTD and reduces LTP
Collapse
Affiliation(s)
- G France
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - R Volianskis
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - R Ingram
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - N Bannister
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - R Rothärmel
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - M W Irvine
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - G Fang
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - E S Burnell
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; University of Exeter, St Luke's Campus, Heavitree Road, Exeter, UK
| | - K Sapkota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - B M Costa
- Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA & Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - D A Chopra
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - S M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - A T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - D T Monaghan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - J Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Z A Bortolotto
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - D E Jane
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - G L Collingridge
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Department of Physiology, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada; TANZ Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - A Volianskis
- Schools of Clinical Sciences and Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK; School of Biosciences, Museum Avenue, Cardiff University, Cardiff, CF10 3AX, UK.
| |
Collapse
|
91
|
Abstract
Drug addiction remains a key biomedical challenge facing current neuroscience research. In addition to neural mechanisms, the focus of the vast majority of studies to date, astrocytes have been increasingly recognized as an "accomplice." According to the tripartite synapse model, astrocytes critically regulate nearby pre- and postsynaptic neuronal substrates to craft experience-dependent synaptic plasticity, including synapse formation and elimination. Astrocytes within brain regions that are implicated in drug addiction exhibit dynamic changes in activity upon exposure to cocaine and subsequently undergo adaptive changes themselves during chronic drug exposure. Recent results have identified several key astrocytic signaling pathways that are involved in cocaine-induced synaptic and circuit adaptations. In this review, we provide a brief overview of the role of astrocytes in regulating synaptic transmission and neuronal function, and discuss how cocaine influences these astrocyte-mediated mechanisms to induce persistent synaptic and circuit alterations that promote cocaine seeking and relapse. We also consider the therapeutic potential of targeting astrocytic substrates to ameliorate drug-induced neuroplasticity for behavioral benefits. While primarily focusing on cocaine-induced astrocytic responses, we also include brief discussion of other drugs of abuse where data are available.
Collapse
|
92
|
Laha K, Zhu M, Gemperline E, Rau V, Li L, Fanselow MS, Lennertz R, Pearce RA. CPP impairs contextual learning at concentrations below those that block pyramidal neuron NMDARs and LTP in the CA1 region of the hippocampus. Neuropharmacology 2022; 202:108846. [PMID: 34687710 PMCID: PMC8627488 DOI: 10.1016/j.neuropharm.2021.108846] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/29/2021] [Accepted: 10/15/2021] [Indexed: 01/03/2023]
Abstract
Drugs that block N-methyl-d-aspartate receptors (NMDARs) suppress hippocampus-dependent memory formation; they also block long-term potentiation (LTP), a cellular model of learning and memory. However, the fractional block that is required to achieve these effects is unknown. Here, we measured the dose-dependent suppression of contextual memory in vivo by systemic administration of the competitive antagonist (R,S)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP); in parallel, we measured the concentration-dependent block by CPP of NMDAR-mediated synapses and LTP of excitatory synapses in hippocampal brain slices in vitro. We found that the dose of CPP that suppresses contextual memory in vivo (EC50 = 2.3 mg/kg) corresponds to a free concentration of 53 nM. Surprisingly, applying this concentration of CPP to hippocampal brain slices had no effect on the NMDAR component of evoked field excitatory postsynaptic potentials (fEPSPNMDA), or on LTP. Rather, the IC50 for blocking the fEPSPNMDA was 434 nM, and for blocking LTP was 361 nM - both nearly an order of magnitude higher. We conclude that memory impairment produced by systemically administered CPP is not due primarily to its blockade of NMDARs on hippocampal pyramidal neurons. Rather, systemic CPP suppresses memory formation by actions elsewhere in the memory-encoding circuitry.
Collapse
Affiliation(s)
- Kurt Laha
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Mengwen Zhu
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Erin Gemperline
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
| | - Vinuta Rau
- Department of Anesthesiology, University of California-San Francisco, San Francisco, CA, USA.
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
| | - Michael S Fanselow
- Departments of Psychology and Psychiatry, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Richard Lennertz
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Robert A Pearce
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
93
|
Eapen AV, Fernández-Fernández D, Georgiou J, Bortolotto ZA, Lightman S, Jane DE, Volianskis A, Collingridge GL. Multiple roles of GluN2D-containing NMDA receptors in short-term potentiation and long-term potentiation in mouse hippocampal slices. Neuropharmacology 2021; 201:108833. [PMID: 34637787 PMCID: PMC8607330 DOI: 10.1016/j.neuropharm.2021.108833] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/28/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023]
Abstract
The GluN2 subunits of N-methyl-d-aspartate receptors (NMDARs) are key drivers of synaptic plasticity in the brain, where the particular GluN2 composition endows the NMDAR complex with distinct pharmacological and physiological properties. Compared to GluN2A and GluN2B subunits, far less is known about the role of the GluN2D subunit in synaptic plasticity. In this study, we have used a GluN2C/2D selective competitive antagonist, UBP145, in combination with a GluN2D global knockout (GluN2D KO) mouse line to study the contribution of GluN2D-containing NMDARs to short-term potentiation (STP) and long-term potentiation (LTP) in the CA1 region of mouse hippocampal slices. We made several distinct observations: First, GluN2D KO mice have higher levels of LTP compared to wild-type (WT) mice, an effect that was occluded by blockade of GABA receptor-mediated inhibition or by using a strong LTP induction protocol. Second, UBP145 partially inhibited LTP in WT but not GluN2D KO mice. Third, UBP145 inhibited a component of STP, termed STP2, in WT but not GluN2D KO mice. Taken together, these findings suggest an involvement for GluN2D-containing NMDARs in both STP and LTP in mouse hippocampus.
Collapse
Affiliation(s)
- Alen V Eapen
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada.
| | - Diego Fernández-Fernández
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Zuner A Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | | | - David E Jane
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Arturas Volianskis
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Bristol Medical School, University of Bristol, Bristol, UK; Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Graham L Collingridge
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada; TANZ Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
94
|
Collingridge GL, Abraham WC. Glutamate receptors and synaptic plasticity: The impact of Evans and Watkins. Neuropharmacology 2021; 206:108922. [PMID: 34919905 DOI: 10.1016/j.neuropharm.2021.108922] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/23/2021] [Accepted: 12/09/2021] [Indexed: 12/31/2022]
Abstract
On the occasion of the 40 year anniversary of the hugely impactful review by Richard (Dick) Evans and Jeff Watkins, we describe how their work has impacted the field of synaptic plasticity. We describe their influence in each of the major glutamate receptor subtypes: AMPARs, NMDARs, KARs and mGluRs. Particular emphasis is placed on how their work impacted our own studies in the hippocampus. For example, we describe how the tools and regulators that they identified for studying NMDARs (e.g., NMDA, D-AP5 and Mg2+) led to the understanding of the molecular basis of the induction of LTP. We also describe how other tools that they introduced (e.g., (1S,3R)-ACPD and MCPG) helped lead to the concept of metaplasticity.
Collapse
Affiliation(s)
- G L Collingridge
- Department of Psychology, Brain Health Research Centre and Brain Research New Zealand, University of Otago, New Zealand; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada; TANZ Centre for Research in Neurodegenerative Diseases, Department of Physiology, University of Toronto, Toronto, ON, Canada.
| | - W C Abraham
- Department of Psychology, Brain Health Research Centre and Brain Research New Zealand, University of Otago, New Zealand
| |
Collapse
|
95
|
Juárez-Aguilar E, Olivares-Hernández JD, Regalado-Santiago C, García-García F. The role of growth hormone in hippocampal function. VITAMINS AND HORMONES 2021; 118:289-313. [PMID: 35180930 DOI: 10.1016/bs.vh.2021.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Growth hormone is a multifunctional molecule with broad cellular targets. This pituitary hormone is currently used as a therapeutic agent against several brain injuries due to its neurotrophic activity. The hippocampus is one of the brain regions where the growth hormone plays a role in normal and pathologic conditions. This brain structure is associated with several cognitive functions such as learning, memory, and mood, which are frequently affected by brain traumatism. The present chapter describes the experimental and clinical evidence that supports a central role of growth hormone in the hippocampus functionality.
Collapse
Affiliation(s)
- Enrique Juárez-Aguilar
- Departmento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico.
| | - Juan David Olivares-Hernández
- Laboratorio D-01, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | | | - Fabio García-García
- Departmento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico
| |
Collapse
|
96
|
Quercetin exhibits potent antioxidant activity, restores motor and non-motor deficits induced by rotenone toxicity. PLoS One 2021; 16:e0258928. [PMID: 34767546 PMCID: PMC8589152 DOI: 10.1371/journal.pone.0258928] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/10/2021] [Indexed: 02/06/2023] Open
Abstract
The rotenone-induced animal model of Parkinson's disease (PD) has been used to investigate the pathogenesis of PD. Oxidative stress is one of the main contributors of neurodegeneration in PD. Flavonoids have the potential to modulate neuronal function and combat various neurodegenerative diseases. The pre- and post-supplementation of quercetin (50 mg/kg, p.o) was done in rats injected with rotenone (1.5 mg/kg, s.c). After the treatment, behavioral activities were monitored for motor activity, depression-like behavior, and cognitive changes. Rats were decapitated after behavioral analysis and the brain samples were dissected out for neurochemical and biochemical estimation. Results showed that supplementation of quercetin significantly (p<0.01) restored rotenone-induced motor and non-motor deficits (depression and cognitive impairments), enhanced antioxidant enzyme activities (p<0.01), and attenuated neurotransmitter alterations (p<0.01). It is suggested that quercetin supplementation improves neurotransmitter levels by mitigating oxidative stress via increasing antioxidant enzyme activity and hence improves motor activity, cognitive functions, and reduces depressive behavior. The results of the present study showed that quercetin pre-supplementation produced more significant results as compared to post-supplementation. These findings show that quercetin can be a potential therapeutic agent to reduce the risk and progression of PD.
Collapse
|
97
|
Memory Disorders Related to Hippocampal Function: The Interest of 5-HT 4Rs Targeting. Int J Mol Sci 2021; 22:ijms222112082. [PMID: 34769511 PMCID: PMC8584667 DOI: 10.3390/ijms222112082] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
The hippocampus has long been considered as a key structure for memory processes. Multilevel alterations of hippocampal function have been identified as a common denominator of memory impairments in a number of psychiatric and neurodegenerative diseases. For many years, the glutamatergic and cholinergic systems have been the main targets of therapeutic treatments against these symptoms. However, the high rate of drug development failures has left memory impairments on the sideline of current therapeutic strategies. This underscores the urgent need to focus on new therapeutic targets for memory disorders, such as type 4 serotonin receptors (5-HT4Rs). Ever since the discovery of their expression in the hippocampus, 5-HT4Rs have gained growing interest for potential use in the treatment of learning and memory impairments. To date, much of the researched information gathered by scientists from both animal models and humans converge on pro-mnesic and anti-amnesic properties of 5-HT4Rs activation, although the mechanisms at work require more work to be fully understood. This review addresses a fundamental, yet poorly understood set of evidence of the potential of 5-HT4Rs to re-establish or limit hippocampal alterations related to neurological diseases. Most importantly, the potential of 5-HT4Rs is translated by refining hypotheses regarding the benefits of their activation in memory disorders at the hippocampal level.
Collapse
|
98
|
Safari S, Ahmadi N, Mohammadkhani R, Ghahremani R, Khajvand-Abedeni M, Shahidi S, Komaki A, Salehi I, Karimi SA. Sex differences in spatial learning and memory and hippocampal long-term potentiation at perforant pathway-dentate gyrus (PP-DG) synapses in Wistar rats. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2021; 17:9. [PMID: 34724971 PMCID: PMC8559395 DOI: 10.1186/s12993-021-00184-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/24/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recent studies show that gender may have a significant impact on brain functions. However, the reports of sex effects on spatial ability and synaptic plasticity in rodents are divergent and controversial. Here spatial learning and memory was measured in male and female rats by using Morris water maze (MWM) task. Moreover, to assess sex difference in hippocampal synaptic plasticity we examined hippocampal long-term potentiation (LTP) at perforant pathway-dentate gyrus (PP-DG) synapses. RESULTS In MWM task, male rats outperformed female rats, as they had significantly shorter swim distance and escape latency to find the hidden platform during training days. During spatial reference memory test, female rats spent less time and traveled less distance in the target zone. Male rats also had larger LTP at PP-DG synapses, which was evident in the high magnitude of population spike (PS) potentiation and the field excitatory post synaptic potentials (fEPSP) slope. CONCLUSIONS Taken together, our results suggest that sex differences in the LTP at PP-DG synapses, possibly contribute to the observed sex difference in spatial learning and memory.
Collapse
Affiliation(s)
- Samaneh Safari
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nesa Ahmadi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Reza Ghahremani
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Birjand, Birjand, Iran
| | | | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran.
| |
Collapse
|
99
|
Pu H, Ma C, Zhao Y, Wang Y, Zhang W, Miao W, Yu F, Hu X, Shi Y, Leak RK, Hitchens TK, Dixon CE, Bennett MV, Chen J. Intranasal delivery of interleukin-4 attenuates chronic cognitive deficits via beneficial microglial responses in experimental traumatic brain injury. J Cereb Blood Flow Metab 2021; 41:2870-2886. [PMID: 34259069 PMCID: PMC8545055 DOI: 10.1177/0271678x211028680] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Traumatic brain injury (TBI) is commonly followed by long-term cognitive deficits that severely impact the quality of life in survivors. Recent studies suggest that microglial/macrophage (Mi/MΦ) polarization could have multidimensional impacts on post-TBI neurological outcomes. Here, we report that repetitive intranasal delivery of interleukin-4 (IL-4) nanoparticles for 4 weeks after controlled cortical impact improved hippocampus-dependent spatial and non-spatial cognitive functions in adult C57BL6 mice, as assessed by a battery of neurobehavioral tests for up to 5 weeks after TBI. IL-4-elicited enhancement of cognitive functions was associated with improvements in the integrity of the hippocampus at the functional (e.g., long-term potentiation) and structural levels (CA3 neuronal loss, diffusion tensor imaging of white matter tracts, etc.). Mechanistically, IL-4 increased the expression of PPARγ and arginase-1 within Mi/MΦ, thereby driving microglia toward a global inflammation-resolving phenotype. Notably, IL-4 failed to shift microglial phenotype after TBI in Mi/MΦ-specific PPARγ knockout (mKO) mice, indicating an obligatory role for PPARγ in IL-4-induced Mi/MΦ polarization. Accordingly, post-TBI treatment with IL-4 failed to improve hippocampal integrity or cognitive functions in PPARγ mKO mice. These results demonstrate that administration of exogenous IL-4 nanoparticles stimulates PPARγ-dependent beneficial Mi/MΦ responses, and improves hippocampal function after TBI.
Collapse
Affiliation(s)
- Hongjian Pu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cheng Ma
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yongfang Zhao
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yangfan Wang
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wenting Zhang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wanying Miao
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fang Yu
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yejie Shi
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rehana K Leak
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - T Kevin Hitchens
- Animal Imaging Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - C Edward Dixon
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Vl Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jun Chen
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
100
|
Lv Z, Chen Z, Ye W, Pang X, Nie L, Chang W, Long Q, Zheng J. Disruption in Surface-Based Functional Connectivity in the Right Posterior Hippocampal CA3 Subfield: A Probable Neural Basis of Visuospatial Working Memory Impairment in Patients With Right Temporal Lobe Epilepsy. Front Neurol 2021; 12:735689. [PMID: 34712198 PMCID: PMC8545809 DOI: 10.3389/fneur.2021.735689] [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: 07/03/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022] Open
Abstract
Visuospatial working memory (VSWM) impairment is common in patients with right temporal lobe epilepsy (rTLE). The posterior hippocampus is critical for spatial memory, but the contributions of the different subfields to VSWM deficits remain unclear. Forty-six rTLE patients and 42 healthy controls (HCs) were recruited. Resting-state fMRI (rsfMRI) and structural MRI scans were administered, followed by a VSWM_Nback test. The right posterior hippocampus was automatically segmented, and the surface-based functional connectivity (SBFC) of the subiculum (Sub), CA1, CA3, dentate gyrus (DG), hippocampal tail, and right entorhinal cortex (EC) were compared between groups. Correlation analysis was performed between the altered SBFC and VSWM_Nback scores for rTLE patients. The results showed that rTLE patients underperformed in the VSWM_Nback test, with longer mean reaction time of accurate response (ACCmeanRT) in 0back and 2back condition, lower hit rate (HR) and higher false alarm rate (FAR) in 2back condition. Compared with HCs, the rCA3 in the rTLE group exhibited decreased SBFC with inferior parietal cortex (IPC), temporal lateral cortex (TLC), and posterior visual cortex (PVC) in the right hemisphere as well as the bilateral dorsolateral prefrontal cortex (DLPFC). The SBFC of the rEC and right anterior cingulate cortex (rACC) increased in the rTLE group. Within the rTLE group, the decreased SBFC of the rCA3-rIPC and rCA3-rLTC were correlated with worse VSWM performance. Therefore, the decreased SBFC of the rCA3-rIPC and rCA3-rLTC might be the critical aberrant FC pattern reflecting VSWM impairment in rTLE patients. The mechanism might involve functional disruption between the core subsystem and the medial temporal subsystem of the default mode network (DMN).
Collapse
Affiliation(s)
- Zongxia Lv
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zirong Chen
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei Ye
- Department of Radiology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaomin Pang
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liluo Nie
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weiwei Chang
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qijia Long
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinou Zheng
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|