1
|
Gardner RS, Ambalavanar MT, Gold PE, Korol DL. Enhancement of response learning in male rats with intrastriatal infusions of a BDNF - TrkB agonist, 7,8-dihydroxyflavone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606692. [PMID: 39211174 PMCID: PMC11360987 DOI: 10.1101/2024.08.08.606692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Enhancement of learning and memory by cognitive and physical exercise may be mediated by brain-derived neurotrophic factor (BDNF) acting at tropomyosin receptor kinase B (TrkB). Upregulation of BDNF and systemic administration of a TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF), enhance learning of several hippocampus-sensitive tasks in rodents. Although BDNF and 7,8-DHF enhance functions of other brain areas too, these effects have mainly targeted non-cognitive functions. One goal of the present study was to determine whether 7,8-DHF would act beyond the hippocampus to enhance cognitive functions sensitive to manipulations of the striatum. Here, we examined the effects of intrastriatal infusions of 7,8-DHF on learning a striatum-sensitive response maze and on phosphorylation of TrkB receptors in 3-month-old male Sprague Dawley rats. Most prior studies of BDNF and 7,8-DHF effects on learning and memory have administered the drugs for days to months before assessing effects on cognition. A second goal of the present study was to determine whether a single drug treatment near the time of training would effectively enhance learning. Moreover, 7,8-DHF is often tested for its ability to reverse impairments in learning and memory rather than to enhance these functions in the absence of impairments. Thus, a third goal of this experiment was to evaluate the efficacy of 7,8-DHF in enhancing learning in unimpaired rats. In untrained rats, intrastriatal infusions of 7,8-DHF resulted in phosphorylation of TrkB receptors, suggesting that 7,8-DHF acted as a TrkB agonist and BDNF mimic. The findings that a single, intra-striatal infusion of 7,8-DHF 20 min before training enhanced response learning in rats suggest that, in addition to its trophic effects, BDNF modulates learning and memory through receptor mediated cell signaling events.
Collapse
|
2
|
Lucas ME, Hemsworth LM, Butler KL, Morrison RS, Tilbrook AJ, Marchant JN, Rault JL, Galea RY, Hemsworth PH. Early human contact and housing for pigs - part 3: ability to cope with the environment. Animal 2024; 18:101166. [PMID: 38772077 DOI: 10.1016/j.animal.2024.101166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/23/2024] Open
Abstract
Early experiences can have long-term impacts on stress adaptability. This paper is the last of three in a series on early experiences and stress in pigs, and reports on the effects of early human contact and housing on the ability of pigs to cope with their general environment. Using a 2 × 2 factorial design, 48 litters of pigs were reared in either a farrowing crate (FC) or a loose farrowing pen (LP; PigSAFE pen) which was larger, more physically complex and allowed the sow to move freely. Piglets were provided with either routine contact from stockpeople (C), or routine contact plus regular opportunities for positive human contact (+HC) involving 5 min of scratching, patting and stroking imposed to the litter 5 days/week from 0 to 4 weeks of age. At 4 weeks of age (preweaning), C piglets that were reared in FC had considerably lower concentrations of serum brain-derived neurotrophic factor (BDNF) than piglets from the other treatment combinations. Compared to C pigs, +HC pigs had fewer injuries at 4 weeks of age. There were no clear effects of human contact on BDNF concentrations or injuries after weaning, or on basal cortisol or immunoglobulin-A concentrations, behavioural time budgets, tear staining, growth, or piglet survival. Compared to FC piglets, LP piglets showed more play behaviour and interactions with the dam and less repetitive nosing towards pen mates during lactation. There was no evidence that early housing affected pigs' behavioural time budgets or physiology after weaning. Tear staining severity was greater in LP piglets at 4 weeks of age, but this may have been associated with the higher growth rates of LP piglets preweaning. There was no effect of lactation housing on growth after weaning. Preweaning piglet mortality was higher in the loose system. The findings on BDNF concentrations, injuries and play behaviour suggest improved welfare during the treatment period in +HC and LP piglets compared to C and FC piglets, respectively. These results together with those from the other papers in this series indicate that positive human interaction early in life promotes stress adaptability in pigs. Furthermore, while the farrowing crate environment deprives piglets of opportunities for play behaviour and sow interaction, there was no evidence that rearing in crates negatively affected pig welfare or stress resilience after weaning. Whether these findings are specific to the two housing systems studied here, or can be generalised to other housing designs, warrants further research.
Collapse
Affiliation(s)
- M E Lucas
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - L M Hemsworth
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - K L Butler
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - R S Morrison
- Rivalea Australia Pty Ltd, Corowa, Victoria 2464, Australia
| | - A J Tilbrook
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia 4072, Australia; School of Veterinary Science, The University of Queensland, Gatton Campus, Gatton, Queensland 4343, Australia
| | - J N Marchant
- Organic Plus Trust, Alexandria, VA 22302, USA; A World of Good Initiative Inc., Dover, DE 19901, USA
| | - J-L Rault
- Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna A-1210, Austria
| | - R Y Galea
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - P H Hemsworth
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
3
|
Zhong Y, Du Q, Wang Z, Zheng Q, Yang M, Hu P, Yang Q, Xu H, Wu Z, Huang X, Li H, Tang M, Zeng H, Zhu L, Ren G, Cao M, Liu Y, Wang H. Antidepressant effect of Perilla frutescens essential oil through monoamine neurotransmitters and BDNF/TrkB signal pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116840. [PMID: 37355083 DOI: 10.1016/j.jep.2023.116840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine posits that affect-mind ill-being is the primary cause of depression, with Qi movement stagnation as its pathogenesis. As such, clinical treatment for depression should prioritize regulating Qi and relieving depressive symptoms. The pharmacological properties of traditional Chinese medicine indicate that Perilla frutescens may have potential therapeutic effects on depression and other neuropsychiatric diseases due to its ability to regulate Qi and alleviate depressive symptoms. Although previous studies have reported the antidepressant effects of Perilla frutescens, the mechanism underlying PFEO inhalation-mediated antidepressant effect remains unclear. AIM OF THE STUDY The aim of this investigation is to elucidate the antidepressant mechanisms of PFEO by examining its effects on monoamine neurotransmitters and the BDNF/TrkB signaling pathway. MATERIALS AND METHODS The CUMS rat model of depression was established, and the depressive state of the animals was assessed through sucrose preference and forced swim tests. ELISA assays were conducted to determine monoamine neurotransmitter levels in the hippocampus and cerebral cortex of rats. Immunohistochemistry, western blotting, and RT-PCR experiments were employed to investigate the BDNF/TrkB signaling pathway's regulation of depression via PFEO inhalation. RESULTS It has been observed that inhalation administration of PFEO can significantly enhance the preference for sugar water in CUMS rats and reduce their immobility time during forced swimming. Additionally, there was an increase in the levels of monoamine transmitters in both the hippocampus and cerebral cortex of these rats. Furthermore, there was an upregulation in the expression levels of BDNF and TrkB positive cells as well as BDNF and TrkB proteins within both regions, along with increased BDNF mRNA and TrkB mRNA expression levels. CONCLUSION The antidepressant effect of PFEO via inhalation administration is speculated to be mediated through the monoamine neurotransmitters and BDNF/TrkB signaling pathway.
Collapse
Affiliation(s)
- Yu Zhong
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qing Du
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Ziqian Wang
- Jiangxi Drug Inspection Center, Nanchang, 330000, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Ming Yang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Qiyue Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Huanhua Xu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Xiaoying Huang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Huiting Li
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Mingxia Tang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Huiming Zeng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Liyun Zhu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Guilin Ren
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ming Cao
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yu Liu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Hongbo Wang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab.of Innovation Drug and Effcient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| |
Collapse
|
4
|
Nachtigall EG, de C Myskiw J, Izquierdo I, Furini CRG. Cellular mechanisms of contextual fear memory reconsolidation: Role of hippocampal SFKs, TrkB receptors and GluN2B-containing NMDA receptors. Psychopharmacology (Berl) 2024; 241:61-73. [PMID: 37700085 DOI: 10.1007/s00213-023-06463-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023]
Abstract
Memories are stored into long-term representations through a process that depends on protein synthesis. However, a consolidated memory is not static and inflexible and can be reactivated under certain circumstances, the retrieval is able to reactivate memories and destabilize them engaging a process of restabilization known as reconsolidation. Although the molecular mechanisms that mediate fear memory reconsolidation are not entirely known, so here we investigated the molecular mechanisms in the hippocampus involved in contextual fear conditioning memory (CFC) reconsolidation in male Wistar rats. We demonstrated that the blockade of Src family kinases (SFKs), GluN2B-containing NMDA receptors and TrkB receptors (TrkBR) in the CA1 region of the hippocampus immediately after the reactivation session impaired contextual fear memory reconsolidation. These impairments were blocked by the neurotrophin BDNF and the NMDAR agonist, D-Serine. Considering that the study of the link between synaptic proteins is crucial for understanding memory processes, targeting the reconsolidation process may provide new ways of disrupting maladaptive memories, such as those seen in post-traumatic stress disorder. Here we provide new insights into the cellular mechanisms involved in contextual fear memory reconsolidation, demonstrating that SFKs, GluN2B-containing NMDAR, and TrkBR are necessary for the reconsolidation process. Our findings suggest a link between BDNF and SFKs and GluN2B-containing NMDAR as well as a link between NMDAR and SFKs and TrkBR in fear memory reconsolidation. These preliminary pharmacological findings provide new evidence of the mechanisms involved in the reconsolidation of fear memory and have the potential to contribute to the development of treatments for psychiatric disorders involving maladaptive memories.
Collapse
Affiliation(s)
- Eduarda G Nachtigall
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, Porto Alegre, RS, 90610-000, Brazil
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil
| | - Jociane de C Myskiw
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil
| | - Ivan Izquierdo
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil
| | - Cristiane R G Furini
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, Porto Alegre, RS, 90610-000, Brazil.
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil.
| |
Collapse
|
5
|
Eyford BA, Lazarczyk MJ, Choi KB, Varghese M, Arora H, Kari S, Munro L, Pfeifer CG, Sowa A, Dickstein DR, Dickstein DL, Jefferies WA. Outside-in signaling through the major histocompatibility complex class-I cytoplasmic tail modulates glutamate receptor expression in neurons. Sci Rep 2023; 13:13079. [PMID: 37567897 PMCID: PMC10421907 DOI: 10.1038/s41598-023-38663-z] [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: 03/16/2023] [Accepted: 07/12/2023] [Indexed: 08/13/2023] Open
Abstract
The interplay between AMPA-type glutamate receptors (AMPARs) and major histocompatibility complex class I (MHC-I) proteins in regulating synaptic signaling is a crucial aspect of central nervous system (CNS) function. In this study, we investigate the significance of the cytoplasmic tail of MHC-I in synaptic signaling within the CNS and its impact on the modulation of synaptic glutamate receptor expression. Specifically, we focus on the Y321 to F substitution (Y321F) within the conserved cytoplasmic tyrosine YXXΦ motif, known for its dual role in endocytosis and cellular signaling of MHC-I. Our findings reveal that the Y321F substitution influences the expression of AMPAR subunits GluA2/3 and leads to alterations in the phosphorylation of key kinases, including Fyn, Lyn, p38, ERK1/2, JNK1/2/3, and p70 S6 kinase. These data illuminate the crucial role of MHC-I in AMPAR function and present a novel mechanism by which MHC-I integrates extracellular cues to modulate synaptic plasticity in neurons, which ultimately underpins learning and memory.
Collapse
Affiliation(s)
- Brett A Eyford
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Maciej J Lazarczyk
- Division of Institutional Measures, Department of Medical Direction and Quality, University Hospitals of Geneva, Geneva, Switzerland
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Kyung Bok Choi
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Merina Varghese
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Hitesh Arora
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Suresh Kari
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lonna Munro
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Cheryl G Pfeifer
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Allison Sowa
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Daniel R Dickstein
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Dara L Dickstein
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA.
- Department of Pathology, Uniformed Services University of Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), 6720A Rockledge Drive, Bethesda, MD, 20817, USA.
| | - Wilfred A Jefferies
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada.
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada.
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
6
|
Ma T, Feng L, Wei S, Wang Y, Li G, Lu Y, Zhang Y, Chu Y, Wang W, Zhang H. Antisense oligonucleotides targeting basal forebrain ATXN2 enhances spatial memory and ameliorates sleep deprivation-induced fear memory impairment in mice. Brain Behav 2023; 13:e3013. [PMID: 37072935 PMCID: PMC10275523 DOI: 10.1002/brb3.3013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/04/2022] [Accepted: 12/24/2022] [Indexed: 04/20/2023] Open
Abstract
INTRODUCTION Regulation of brain-derived neurotrophic factor (BDNF) in the basal forebrain ameliorates sleep deprivation-induced fear memory impairments in rodents. Antisense oligonucleotides (ASOs) targeting ATXN2 was a potential therapy for spinocerebellar ataxia, whose pathogenic mechanism associates with reduced BDNF expression. We tested the hypothesis that ASO7 targeting ATXN2 could affect BDNF levels in mouse basal forebrain and ameliorate sleep deprivation-induced fear memory impairments. METHODS Adult male C57BL/6 mice were used to evaluate the effects of ASO7 targeting ATXN2 microinjected into the bilateral basal forebrain (1 μg, 0.5 μL, each side) on spatial memory, fear memory and sleep deprivation-induced fear memory impairments. Spatial memory and fear memory were detected by the Morris water maze and step-down inhibitory avoidance test, respectively. Immunohistochemistry, RT-PCR, and Western blot were used to evaluate the changes of levels of BDNF, ATXN2, and postsynaptic density 95 (PSD95) protein as well as ATXN2 mRNA. The morphological changes in neurons in the hippocampal CA1 region were detected by HE staining and Nissl staining. RESULTS ASO7 targeting ATXN2 microinjected into the basal forebrain could suppress ATXN2 mRNA and protein expression for more than 1 month and enhance spatial memory but not fear memory in mice. BDNF mRNA and protein expression in basal forebrain and hippocampus was increased by ASO7. Moreover, PSD95 expression and synapse formation were increased in the hippocampus. Furthermore, ASO7 microinjected into the basal forebrain increased BDNF and PSD95 protein expression in the basal forebrain of sleep-deprived mice and counteracted sleep deprivation-induced fear memory impairments. CONCLUSION ASOs targeting ATXN2 may provide effective interventions for sleep deprivation-induced cognitive impairments.
Collapse
Affiliation(s)
- Tao Ma
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Long Feng
- Department of AnesthesiologyPLA General Hospital of Hainan HospitalHainanChina
| | - Shi‐Nan Wei
- PLA Rocket Force Characteristic Medical Center, Postgraduate Training Base of Jinzhou Medical UniversityBeijingChina
| | - Ying‐Ying Wang
- Department of AnesthesiologyBeijing Ditan Hospital, Capital Medical UniversityBeijingChina
| | - Guan‐Hua Li
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Yan Lu
- Department of NeurologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Ying‐Xin Zhang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Yang Chu
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Wei Wang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Hao Zhang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| |
Collapse
|
7
|
Electroacupuncture stimulation of HT7 alleviates sleep disruption following acute caffeine exposure by regulating BDNF-mediated endoplasmic reticulum stress in the rat medial septum. Biomed Pharmacother 2022; 155:113724. [PMID: 36156370 DOI: 10.1016/j.biopha.2022.113724] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
Acupuncture stimulation can protect the brain against caffeine-induced sleep disruption. This study investigated whether electroacupuncture stimulation acupuncture point HT7 alleviates sleep disruption by regulating mBDNF and ER stress in the medial septum. Acute exposure to caffeine (15 mg/kg, i.p.) increased the wake time and decreased REM sleep, which HT7 stimulation alleviated. HT7 stimulation ameliorated the acute caffeine exposure-induced increase in the expression of BiP, an endoplasmic reticulum stress response protein, in the rat medial septum. Interestingly, HT7 stimulation induced the expression of mBDNF and pTrkB in the medial septum. The next experiment investigated whether TrkB phosphorylated by HT7 stimulation induced BiP expression in the rat medial septum. Before electroacupuncture stimulation at HT7, ANA-12 was administered to caffeine-treated rats. In rats administered ANA-12 in the medial septum, HT7 stimulation did not reduce BiP expression. These findings suggest that HT7 stimulation improves wake time and REM sleep dysfunction by regulating the BDNF-mediated endoplasmic reticulum stress response in the medial septum. These results indicate that the alleviation of endoplasmic reticulum stress in the medial septum by HT7 stimulation and the subsequent amelioration of insomnia may depend on phosphorylated TrkB activation.
Collapse
|
8
|
Williams RA, Johnson KW, Lee FS, Hemmings HC, Platholi J. A Common Human Brain-Derived Neurotrophic Factor Polymorphism Leads to Prolonged Depression of Excitatory Synaptic Transmission by Isoflurane in Hippocampal Cultures. Front Mol Neurosci 2022; 15:927149. [PMID: 35813074 PMCID: PMC9260310 DOI: 10.3389/fnmol.2022.927149] [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: 04/23/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Multiple presynaptic and postsynaptic targets have been identified for the reversible neurophysiological effects of general anesthetics on synaptic transmission and neuronal excitability. However, the synaptic mechanisms involved in persistent depression of synaptic transmission resulting in more prolonged neurological dysfunction following anesthesia are less clear. Here, we show that brain-derived neurotrophic factor (BDNF), a growth factor implicated in synaptic plasticity and dysfunction, enhances glutamate synaptic vesicle exocytosis, and that attenuation of vesicular BDNF release by isoflurane contributes to transient depression of excitatory synaptic transmission in mice. This reduction in synaptic vesicle exocytosis by isoflurane was acutely irreversible in neurons that release less endogenous BDNF due to a polymorphism (BDNF Val66Met; rs6265) compared to neurons from wild-type mice. These effects were prevented by exogenous application of BDNF. Our findings identify a role for a common human BDNF single nucleotide polymorphism in persistent changes of synaptic function following isoflurane exposure. These short-term persistent alterations in excitatory synaptic transmission indicate a role for human genetic variation in anesthetic effects on synaptic plasticity and neurocognitive function.
Collapse
Affiliation(s)
- Riley A. Williams
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
| | - Kenneth W. Johnson
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Francis S. Lee
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States,Department of Psychiatry, Sackler Institute for Developmental Psychobiology, Weill Cornell Medicine, New York, NY, United States,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Hugh C. Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States,Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Jimcy Platholi
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States,*Correspondence: Jimcy Platholi,
| |
Collapse
|
9
|
Platholi J, Hemmings HC. Effects of general anesthetics on synaptic transmission and plasticity. Curr Neuropharmacol 2021; 20:27-54. [PMID: 34344292 PMCID: PMC9199550 DOI: 10.2174/1570159x19666210803105232] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022] Open
Abstract
General anesthetics depress excitatory and/or enhance inhibitory synaptic transmission principally by modulating the function of glutamatergic or GABAergic synapses, respectively, with relative anesthetic agent-specific mechanisms. Synaptic signaling proteins, including ligand- and voltage-gated ion channels, are targeted by general anesthetics to modulate various synaptic mechanisms, including presynaptic neurotransmitter release, postsynaptic receptor signaling, and dendritic spine dynamics to produce their characteristic acute neurophysiological effects. As synaptic structure and plasticity mediate higher-order functions such as learning and memory, long-term synaptic dysfunction following anesthesia may lead to undesirable neurocognitive consequences depending on the specific anesthetic agent and the vulnerability of the population. Here we review the cellular and molecular mechanisms of transient and persistent general anesthetic alterations of synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Jimcy Platholi
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| | - Hugh C Hemmings
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| |
Collapse
|
10
|
Ehinger R, Kuret A, Matt L, Frank N, Wild K, Kabagema-Bilan C, Bischof H, Malli R, Ruth P, Bausch AE, Lukowski R. Slack K + channels attenuate NMDA-induced excitotoxic brain damage and neuronal cell death. FASEB J 2021; 35:e21568. [PMID: 33817875 DOI: 10.1096/fj.202002308rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
The neuronal Na+ -activated K+ channel Slack (aka Slo2.2, KNa 1.1, or Kcnt1) has been implicated in setting and maintaining the resting membrane potential and defining excitability and firing patterns, as well as in the generation of the slow afterhyperpolarization following bursts of action potentials. Slack activity increases significantly under conditions of high intracellular Na+ levels, suggesting this channel may exert important pathophysiological functions. To address these putative roles, we studied whether Slack K+ channels contribute to pathological changes and excitotoxic cell death caused by glutamatergic overstimulation of Ca2+ - and Na+ -permeable N-methyl-D-aspartic acid receptors (NMDAR). Slack-deficient (Slack KO) and wild-type (WT) mice were subjected to intrastriatal microinjections of the NMDAR agonist NMDA. NMDA-induced brain lesions were significantly increased in Slack KO vs WT mice, suggesting that the lack of Slack renders neurons particularly susceptible to excitotoxicity. Accordingly, excessive neuronal cell death was seen in Slack-deficient primary cerebellar granule cell (CGC) cultures exposed to glutamate and NMDA. Differences in neuronal survival between WT and Slack KO CGCs were largely abolished by the NMDAR antagonist MK-801, but not by NBQX, a potent and highly selective competitive antagonist of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors. Interestingly, NMDAR-evoked Ca2+ signals did not differ with regard to Slack genotype in CGCs. However, real-time monitoring of K+ following NMDAR activation revealed a significant contribution of this channel to the intracellular drop in K+ . Finally, TrkB and TrkC neurotrophin receptor transcript levels were elevated in NMDA-exposed Slack-proficient CGCs, suggesting a mechanism by which this K+ channel contributes to the activation of the extracellular-signal-regulated kinase (Erk) pathway and thereby to neuroprotection. Combined, our findings suggest that Slack-dependent K+ signals oppose the NMDAR-mediated excitotoxic neuronal injury by promoting pro-survival signaling via the BDNF/TrkB and Erk axis.
Collapse
Affiliation(s)
- Rebekka Ehinger
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lucas Matt
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Nadine Frank
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Katharina Wild
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Clement Kabagema-Bilan
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Helmut Bischof
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Anne E Bausch
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Sun W, Cheng H, Yang Y, Tang D, Li X, An L. Requirements of Postnatal proBDNF in the Hippocampus for Spatial Memory Consolidation and Neural Function. Front Cell Dev Biol 2021; 9:678182. [PMID: 34336832 PMCID: PMC8319730 DOI: 10.3389/fcell.2021.678182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Mature brain-derived neurotrophic factor (BDNF) and its downstream signaling pathways have been implicated in regulating postnatal development and functioning of rodent brain. However, the biological role of its precursor pro-brain-derived neurotrophic factor (proBDNF) in the postnatal brain remains unknown. The expression of hippocampal proBDNF was blocked in postnatal weeks, and multiple behavioral tests, Western blot and morphological techniques, and neural recordings were employed to investigate how proBDNF played a role in spatial cognition in adults. The peak expression and its crucial effects were found in the fourth but not in the second or eighth postnatal week. Blocking proBDNF expression disrupted spatial memory consolidation rather than learning or memory retrieval. Structurally, blocking proBDNF led to the reduction in spine density and proportion of mature spines. Although blocking proBDNF did not affect N-methyl-D-aspartate (NMDA) receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, the learning-induced phosphorylation of the GluN2B subunit level declined significantly. Functionally, paired-pulse facilitation, post-low-frequency stimulation (LFS) transiently enhanced depression, and GluN2B-dependent short-lasting long-term depression in the Schaffer collateral-CA1 pathway were weakened. The firing rate of pyramidal neurons was significantly suppressed around the target region during the memory test. Furthermore, the activation of GluN2B-mediated signaling could effectively facilitate neural function and mitigate memory impairment. The findings were consistent with the hypothesis that postnatal proBDNF played an essential role in synaptic and cognitive functions.
Collapse
Affiliation(s)
- Wei Sun
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hong Cheng
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dongxin Tang
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaolian Li
- Department of Neurology, Jinan Geriatric Hospital, Jinan, China
| | - Lei An
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
12
|
Rajani V, Sengar AS, Salter MW. Src and Fyn regulation of NMDA receptors in health and disease. Neuropharmacology 2021; 193:108615. [PMID: 34051267 DOI: 10.1016/j.neuropharm.2021.108615] [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: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022]
Abstract
The Src family kinases (SFKs) are cytoplasmic non-receptor tyrosine kinases involved in multiple signalling pathways. In the central nervous system (CNS), SFKs are key regulators of N-methyl-d-aspartate receptor (NMDAR) function and major points of convergence for neuronal transduction pathways. Physiological upregulation of NMDAR activity by members of the SFKs, namely Src and Fyn, is crucial for induction of plasticity at Schaffer collateral-CA1 synapses of the hippocampus. Aberrant SFK regulation of NMDARs is implicated in several pathological conditions in the CNS including schizophrenia and pain hypersensitivity. Here, evidence is presented to highlight the current understanding of the intermolecular interactions of SFKs within the NMDAR macromolecular complex, the upstream regulators of SFK activity on NMDAR function and the role Src and Fyn have in synaptic plasticity and metaplasticity. The targeting of SFK protein-protein interactions is discussed as a potential therapeutic strategy to restore signalling activity underlying glutamatergic dysregulation in CNS disease pathophysiology.
Collapse
Affiliation(s)
- Vishaal Rajani
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada; Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Ameet S Sengar
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
13
|
Daily oscillation of cognitive factors is modified in the temporal cortex of an amyloid β(1-42)-induced rat model of Alzheimer's disease. Brain Res Bull 2021; 170:106-114. [PMID: 33508401 DOI: 10.1016/j.brainresbull.2021.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/28/2020] [Accepted: 01/20/2021] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD) is a devastating disease characterized by loss of synapses and neurons in the elderly. Accumulation of the β-amyloid peptide (Aβ) in the brain is thought to be central to the pathogenesis of AD. ApoE plays a key role in normal and physiological clearance of Aß, since it facilitates the peptide intra- and extracellular proteolytic degradation. Besides the cognitive deficit, AD patients also show alterations in their circadian rhythms. The objective of this study was to investigate the effects of an i.c.v. injection of Aβ (1-42) peptide on the 24 h rhythms of Apo E, BMAL1, RORα, Bdnf and trkB mRNA and Aβ levels in the rat temporal cortex. We found that an i.c.v. injection of Aβ aggregates phase shifts daily Bdnf expression as well as Apo E, BMAL1, RORα, Aβ and decreased the mesor of TrkB rhythms. Thus, elevated Aβ peptide levels might modify the temporal patterns of cognition-related factors, probably; by affecting the clock factors rhythms as well as in the 24 h rhythms of Apo E.
Collapse
|
14
|
Choung JS, Kim JM, Ko MH, Cho DS, Kim M. Therapeutic efficacy of repetitive transcranial magnetic stimulation in an animal model of Alzheimer's disease. Sci Rep 2021; 11:437. [PMID: 33432077 PMCID: PMC7801521 DOI: 10.1038/s41598-020-80147-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 12/14/2020] [Indexed: 12/02/2022] Open
Abstract
Previous studies on repetitive transcranial magnetic stimulation (rTMS) suggested potential neurorestorative properties in Alzheimer's disease (AD). This study aimed to investigate therapeutic effects of rTMS on an AD mouse model at high and low frequencies. The subject mice were allocated into the AD model group (AD induced by intracerebroventricular amyloid beta 42 oligomer [Aβ42] injection) and the saline-injected control group. Each group was subdivided according to rTMS treatment: high frequency (20 Hz), low frequency (1 Hz), and not rTMS-treated. Behavioural assessments with Y-maze test and novel object recognition task were performed; the results indicated cognition recovery by both the frequencies of rTMS after treatment in the AD model (Ps < 0.01). Tendency of further effects by high frequency compared to low frequency rTMS was also shown in Y-maze test. Neurotransmitter assay showed increment in dopamine concentration and upregulation of dopamine-receptor 4 (DR4) by rTMS in AD mice with higher response by high frequency stimulation (Ps < 0.05). Only high-frequency rTMS induced an elevation of brain-derived neurotrophic factor (BDNF) levels and enhanced the expression of Nestin and NeuN in the brain tissue (Ps < 0.05). Under in vitro conditions, Aβ42 incubated mouse hippocampal cell showed an increase in dopamine levels and BDNF by application of high-frequency rTMS treatment. In conclusion, rTMS might have a potential therapeutic effect on AD, and it seems to be related with dopaminergic activation. High frequency of stimulation seems to induce higher efficacy than that induced by low frequency, with elevated expressions of DR4 gene and neurogenic proteins.
Collapse
Affiliation(s)
- Jin Seung Choung
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea
| | - Jong Moon Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, 59 Yatap-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13496, Republic of Korea
| | - Myoung-Hwan Ko
- Department of Physical Medicine and Rehabilitation, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Dong Sik Cho
- R&D Center, Remed Co., Ltd., Seongnam, Republic of Korea
| | - MinYoung Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam, Republic of Korea.
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, 59 Yatap-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
15
|
Alzoubi KH, Abdel-Hafiz L, Khabour OF, El-Elimat T, Alzubi MA, Alali FQ. Evaluation of the Effect of Hypericum triquetrifolium Turra on Memory Impairment Induced by Chronic Psychosocial Stress in Rats: Role of BDNF. Drug Des Devel Ther 2020; 14:5299-5314. [PMID: 33299301 PMCID: PMC7720289 DOI: 10.2147/dddt.s278153] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/14/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Chronic psychosocial stress impairs memory function and leads to a depression-like phenotype induced by a persistent status of oxidative stress. Hypericum perforatum L. (St. John's wort) is widely used to relieve symptoms of anxiety and depression; however, its long-term use is associated with adverse effects. Hypericum triquetrifolium Turra is closely related to H. perforatum. Both plants belong to Hypericaceae family and share many biologically active compounds. Previous work by our group showed that methanolic extracts of H. triquetrifolium have potent antioxidant activity as well as high hypericin content, a component that proved to have stress-relieving and antidepressant effects by other studies. Therefore, we hypothesized that H. triquetrifolium would reduce stress-induced cognitive impairment in a rat model of chronic stress. OBJECTIVE To determine whether chronic treatment with H. triquetrifolium protects against stress-associated memory deficits and to investigate a possible mechanism. METHODS The radial arm water maze (RAWM) was used to test learning and memory in rats exposed to daily stress using the resident-intruder paradigm. Stressed and unstressed rats received chronic H. triquetrifolium or vehicle. We also measured levels of brain-derived neurotrophic factor (BDNF) in the hippocampus, cortex and cerebellum. RESULTS Neither chronic stress nor chronic H. triquetrifolium administration affected performance during acquisition. However, memory tests in the RAWM showed that chronic stress impaired different post-encoding memory stages. H. triquetrifolium prevented this impairment. Furthermore, hippocampal BDNF levels were markedly lower in stressed animals than in unstressed animals, and chronic administration of H triquetrifolium chronic administration protected against this reduction. No significant difference was observed in the effects of chronic stress and/or H. triquetrifolium treatment on BDNF levels in the cerebellum and cortex. CONCLUSION H. triquetrifolium extract can oppose stress-associated hippocampus-dependent memory deficits in a mechanism that may involve BDNF in the hippocampus.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid22110, Jordan
| | - Laila Abdel-Hafiz
- Institute of Anatomy II, Medical Faculty, Heinrich Heine Universität, Düsseldorf, Germany
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid22110, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid22110, Jordan
| | - Tamam El-Elimat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid22110, Jordan
| | - Mohammad A Alzubi
- Integrative Life Sciences Doctoral Program, Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Feras Q Alali
- College of Pharmacy, QU Health, Qatar University, DohaQatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
16
|
Chen W, Marvizón JC. A Src family kinase maintains latent sensitization in rats, a model of inflammatory and neuropathic pain. Brain Res 2020; 1746:146999. [PMID: 32579948 PMCID: PMC10866137 DOI: 10.1016/j.brainres.2020.146999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Latent sensitization is a long-term model of chronic pain in which hyperalgesia is continuously suppressed by opioid receptors, as demonstrated by the induction of mechanical allodynia by opioid antagonists. Different intracellular signals may mediate the initiation, maintenance and expression of latent sensitization. Our criterion for the involvement of a signal in the maintenance of latent sensitization is that inhibitors should permanently eliminate the allodynia produced by an opioid antagonist. We hypothesized that Src family kinases (SFKs) maintain latent sensitization and tested this hypothesis by inducing latent sensitization in rats with complete Freund's adjuvant (CFA) or spared nerve injury. After measures of mechanical allodynia returned to baseline, vehicle or the SFK inhibitor PP2 were injected intrathecally. The opioid antagonist naltrexone injected intrathecally 15 min later produced allodynia in control rats but not in rats injected with PP2. Vehicle or PP2 were injected daily for two more days and naltrexone was injected five days later. Again, naltrexone induced allodynia in the control rats but not in the rats injected with PP2. Results were similar when latent sensitization was induced with CFA or spared nerve injury. We concluded that an SFK, likely Fyn, maintains latent sensitization induced by inflammation or nerve injury.
Collapse
Affiliation(s)
- Wenling Chen
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Juan Carlos Marvizón
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
17
|
IL-4R alpha deficiency influences hippocampal-BDNF signaling pathway to impair reference memory. Sci Rep 2020; 10:16506. [PMID: 33020569 PMCID: PMC7536433 DOI: 10.1038/s41598-020-73574-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/18/2020] [Indexed: 11/08/2022] Open
Abstract
Like pro-inflammatory cytokines, the role of anti-inflammatory cytokines in both learning and memory has been investigated, revealing beneficial effects for both interleukin-4 and interleukin-13 via the common interleukin-4 receptor alpha chain complex. In this study, using the Morris water maze spatial task for cognition, we compared interleukin-4 receptor alpha- deficient mice and their ligands interleukin-4/ interleukin-13 double deficient mice, on a Balb/c background. We demonstrate that while interleukin-4/ interleukin-13 double deficient mice are significantly impaired in both learning and reference memory, interleukin-4 receptor alpha-deficiency impairs only reference memory, compared to the wild-type control mice. In order to better understand how interleukin-4 receptor alpha- deficient mice are able to learn but not remember, we investigated the BDNF/TrkB- and the ARC-signaling pathways. We show that interleukin-4 receptor alpha-deficiency disrupts activation of BDNF/TrkB- and ARC-signaling pathways during reference memory, while the pathway for spatial learning is spared.
Collapse
|
18
|
Cappoli N, Tabolacci E, Aceto P, Dello Russo C. The emerging role of the BDNF-TrkB signaling pathway in the modulation of pain perception. J Neuroimmunol 2020; 349:577406. [PMID: 33002723 DOI: 10.1016/j.jneuroim.2020.577406] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
The brain derived neurotrophic factor (BDNF) is a crucial neuromodulator in pain transmission both in peripheral and central nervous system (CNS). Despite evidence of a pro-nociceptive role of BDNF, recent studies have reported contrasting results, including anti-nociceptive and anti-inflammatory activities. Moreover, BDNF polymorphisms can interfere with BDNF role in pain perception. In Val66Met carriers, the Met allele may have a dual role, with anti-nociceptive actions in normal condition and pro-nociceptive effects during chronic pain. In order to elucidate the main effects of BDNF in nociception, we reviewed the main characteristics of this neurotrophin, focusing on its involvement in pain.
Collapse
Affiliation(s)
- Natalia Cappoli
- Università Cattolica del Sacro Cuore, Dipartimento di Sicurezza e Bioetica, Sezione di Farmacologia, Rome, Italy
| | - Elisabetta Tabolacci
- Università Cattolica del Sacro Cuore, Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Aceto
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Dipartimento di Scienze dell'Emergenza, Anestesiologiche e della Rianimazione, Rome, Italy; Università Cattolica del Sacro Cuore, Dipartimento di Scienze biotecnologiche di base, cliniche intensivologiche e perioperatorie, Rome, Italy.
| | - Cinzia Dello Russo
- Università Cattolica del Sacro Cuore, Dipartimento di Sicurezza e Bioetica, Sezione di Farmacologia, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
19
|
Berger T, Lee H, Young AH, Aarsland D, Thuret S. Adult Hippocampal Neurogenesis in Major Depressive Disorder and Alzheimer's Disease. Trends Mol Med 2020; 26:803-818. [PMID: 32418723 DOI: 10.1016/j.molmed.2020.03.010] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/16/2020] [Accepted: 03/31/2020] [Indexed: 12/25/2022]
Abstract
Depression and dementia are major public health problems. Major depressive disorder (MDD) and Alzheimer's disease (AD) reciprocally elevate the risk for one another. No effective drug is available to treat AD and about one-third of depressive patients show treatment resistance. The biological connection between MDD and AD is still unclear. Uncovering this link might open novel ways of treatment and prevention to improve patient healthcare. Here, we discuss recent studies specifically on the role of human adult hippocampal neurogenesis (AHN) in MDD and AD. We compare diverse approaches to analyse the effect of MDD and AD on human AHN and analyse different studies implicating the role of human AHN as a potential converging mechanism in MDD and AD.
Collapse
Affiliation(s)
- Thomas Berger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Hyunah Lee
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
20
|
Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int J Mol Sci 2020; 21:ijms21124444. [PMID: 32580508 PMCID: PMC7352836 DOI: 10.3390/ijms21124444] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/25/2022] Open
Abstract
Fyn is a non-receptor or cytoplasmatic tyrosine kinase (TK) belonging to the Src family kinases (SFKs) involved in multiple transduction pathways in the central nervous system (CNS) including synaptic transmission, myelination, axon guidance, and oligodendrocyte formation. Almost one hundred years after the original description of Fyn, this protein continues to attract extreme interest because of its multiplicity of actions in the molecular signaling pathways underlying neurodevelopmental as well as neuropathologic events. This review highlights and summarizes the most relevant recent findings pertinent to the role that Fyn exerts in the brain, emphasizing aspects related to neurodevelopment and synaptic plasticity. Fyn is a common factor in healthy and diseased brains that targets different proteins and shapes different transduction signals according to the neurological conditions. We will primarily focus on Fyn-mediated signaling pathways involved in neuronal differentiation and plasticity that have been subjected to considerable attention lately, opening the fascinating scenario to target Fyn TK for the development of potential therapeutic interventions for the treatment of CNS injuries and certain neurodegenerative disorders like Alzheimer’s disease.
Collapse
|
21
|
Sanders O, Rajagopal L. Phosphodiesterase Inhibitors for Alzheimer's Disease: A Systematic Review of Clinical Trials and Epidemiology with a Mechanistic Rationale. J Alzheimers Dis Rep 2020; 4:185-215. [PMID: 32715279 PMCID: PMC7369141 DOI: 10.3233/adr-200191] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preclinical studies, clinical trials, and reviews suggest increasing 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) with phosphodiesterase inhibitors is disease-modifying in Alzheimer's disease (AD). cAMP/protein kinase A (PKA) and cGMP/protein kinase G (PKG) signaling are disrupted in AD. cAMP/PKA and cGMP/PKG activate cAMP response element binding protein (CREB). CREB binds mitochondrial and nuclear DNA, inducing synaptogenesis, memory, and neuronal survival gene (e.g., brain-derived neurotrophic factor) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α). cAMP/PKA and cGMP/PKG activate Sirtuin-1, which activates PGC1α. PGC1α induces mitochondrial biogenesis and antioxidant genes (e.g.,Nrf2) and represses BACE1. cAMP and cGMP inhibit BACE1-inducing NFκB and tau-phosphorylating GSK3β. OBJECTIVE AND METHODS We review efficacy-testing clinical trials, epidemiology, and meta-analyses to critically investigate whether phosphodiesteraseinhibitors prevent or treat AD. RESULTS Caffeine and cilostazol may lower AD risk. Denbufylline and sildenafil clinical trials are promising but preliminary and inconclusive. PF-04447943 and BI 409,306 are ineffective. Vinpocetine, cilostazol, and nicergoline trials are mixed. Deprenyl/selegiline trials show only short-term benefits. Broad-spectrum phosphodiesterase inhibitor propentofylline has been shown in five phase III trials to improve cognition, dementia severity, activities of daily living, and global assessment in mild-to-moderate AD patients on multiple scales, including the ADAS-Cogand the CIBIC-Plus in an 18-month phase III clinical trial. However, two books claimed based on a MedScape article an 18-month phase III trial failed, so propentofylline was discontinued. Now, propentofylline is used to treat canine cognitive dysfunction, which, like AD, involves age-associated wild-type Aβ deposition. CONCLUSION Phosphodiesterase inhibitors may prevent and treat AD.
Collapse
|
22
|
Yousuf H, Smies CW, Hafenbreidel M, Tuscher JJ, Fortress AM, Frick KM, Mueller D. Infralimbic Estradiol Enhances Neuronal Excitability and Facilitates Extinction of Cocaine Seeking in Female Rats via a BDNF/TrkB Mechanism. Front Behav Neurosci 2019; 13:168. [PMID: 31417375 PMCID: PMC6684748 DOI: 10.3389/fnbeh.2019.00168] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/11/2019] [Indexed: 11/13/2022] Open
Abstract
Women are more susceptible to developing cocaine dependence than men, but paradoxically, are more responsive to treatment. The potent estrogen, 17β-estradiol (E2), mediates these effects by augmenting cocaine seeking but also promoting extinction of cocaine seeking through E2's memory-enhancing functions. Although we have previously shown that E2 facilitates extinction, the neuroanatomical locus of action and underlying mechanisms are unknown. Here we demonstrate that E2 infused directly into the infralimbic-medial prefrontal cortex (IL-mPFC), a region critical for extinction consolidation, enhances extinction of cocaine seeking in ovariectomized (OVX) female rats. Using patch-clamp electrophysiology, we show that E2 may facilitate extinction by potentiating intrinsic excitability of IL-mPFC neurons. Because the mnemonic effects of E2 are known to be regulated by brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), we examined whether BDNF/TrkB signaling was necessary for E2-induced enhancement of excitability and extinction. We found that E2-mediated increases in excitability of IL-mPFC neurons were abolished by Trk receptor blockade. Moreover, blockade of TrkB signaling impaired E2-facilitated extinction of cocaine seeking in OVX female rats. Thus, E2 enhances IL-mPFC neuronal excitability in a TrkB-dependent manner to support extinction of cocaine seeking. Our findings suggest that pharmacological enhancement of E2 or BDNF/TrkB signaling during extinction-based therapies would improve therapeutic outcome in cocaine-addicted women.
Collapse
Affiliation(s)
- Hanna Yousuf
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Chad W Smies
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Madalyn Hafenbreidel
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Devin Mueller
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States.,Department of Biological Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
23
|
The Counteracting Effects of Exercise on High-Fat Diet-Induced Memory Impairment: A Systematic Review. Brain Sci 2019; 9:brainsci9060145. [PMID: 31226771 PMCID: PMC6627483 DOI: 10.3390/brainsci9060145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022] Open
Abstract
The objective of the present review was to evaluate whether exercise can counteract a potential high-fat diet-induced memory impairment effect. The evaluated databases included: Google Scholar, Sports Discus, Embase/PubMed, Web of Science, and PsychInfo. Studies were included if: (1) an experimental/intervention study was conducted, (2) the experiment/intervention included both a high-fat diet and exercise group, and evaluated whether exercise could counteract the negative effects of a high-fat diet on memory, and (3) evaluated memory function (any type) as the outcome measure. In total, 17 articles met the inclusionary criteria. All 17 studies (conducted in rodents) demonstrated that the high-fat diet protocol impaired memory function and all 17 studies demonstrated a counteracting effect with chronic exercise engagement. Mechanisms of these robust effects are discussed herein.
Collapse
|
24
|
Innes S, Pariante CM, Borsini A. Microglial-driven changes in synaptic plasticity: A possible role in major depressive disorder. Psychoneuroendocrinology 2019; 102:236-247. [PMID: 30594100 DOI: 10.1016/j.psyneuen.2018.12.233] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022]
Abstract
Recent data gathered from both in vitro and in vivo models of Major Depressive Disorder (MDD) have indicated that microglia play an active role in modifying some of the most important sources for neuronal plasticity, specifically long-term potentiation (LTP) and long-term depression (LTD). In addition, microglia have been implicated in neuro-immune interaction dysregulations, which are considered a core constituent of MDD pathology. While prior studies have investigated the diverse effects activated microglia can have in the context of depression, including regulation of inflammatory cytokine production and structural changes, recent evidence has revealed a more direct relationship between microglial activation and changes in synaptic function and plasticity, including LTP and LTD. Here we review these findings from animal models, as well as discuss how current preclinical evidence might shed light on novel therapeutic targets for patients with depressive disorder.
Collapse
Affiliation(s)
- Stuart Innes
- Guy's King's and St Thomas' School of Life Science and Medicine, King's College London, UK
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Alessandra Borsini
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| |
Collapse
|
25
|
Harrington MO, Klaus K, Vaht M, Harro J, Pennington K, Durrant SJ. Overnight retention of emotional memories is influenced by BDNF Val66Met but not 5-HTTLPR. Behav Brain Res 2019; 359:17-27. [DOI: 10.1016/j.bbr.2018.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/24/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023]
|
26
|
More JY, Bruna BA, Lobos PE, Galaz JL, Figueroa PL, Namias S, Sánchez GL, Barrientos GC, Valdés JL, Paula-Lima AC, Hidalgo C, Adasme T. Calcium Release Mediated by Redox-Sensitive RyR2 Channels Has a Central Role in Hippocampal Structural Plasticity and Spatial Memory. Antioxid Redox Signal 2018; 29:1125-1146. [PMID: 29357673 DOI: 10.1089/ars.2017.7277] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Previous studies indicate that hippocampal synaptic plasticity and spatial memory processes entail calcium release from intracellular stores mediated by ryanodine receptor (RyR) channels. In particular, RyR-mediated Ca2+ release is central for the dendritic spine remodeling induced by brain-derived neurotrophic factor (BDNF), a neurotrophin that stimulates complex signaling pathways leading to memory-associated protein synthesis and structural plasticity. To examine if upregulation of ryanodine receptor type-2 (RyR2) channels and the spine remodeling induced by BDNF entail reactive oxygen species (ROS) generation, and to test if RyR2 downregulation affects BDNF-induced spine remodeling and spatial memory. RESULTS Downregulation of RyR2 expression (short hairpin RNA [shRNA]) in primary hippocampal neurons, or inhibition of nitric oxide synthase (NOS) or NADPH oxidase, prevented agonist-mediated RyR-mediated Ca2+ release, whereas BDNF promoted cytoplasmic ROS generation. RyR2 downregulation or inhibitors of N-methyl-d-aspartate (NMDA) receptors, or NOS or of NADPH oxidase type-2 (NOX2) prevented RyR2 upregulation and the spine remodeling induced by BDNF, as did incubation with the antioxidant agent N-acetyl l-cysteine. In addition, intrahippocampal injection of RyR2-directed antisense oligodeoxynucleotides, which caused significant RyR2 downregulation, caused conspicuous defects in a memorized spatial memory task. INNOVATION The present novel results emphasize the key role of redox-sensitive Ca2+ release mediated by RyR2 channels for hippocampal structural plasticity and spatial memory. CONCLUSION Based on these combined results, we propose (i) that BDNF-induced RyR2-mediated Ca2+ release and ROS generation via NOS/NOX2 are strictly required for the dendritic spine remodeling and the RyR2 upregulation induced by BDNF, and (ii) that RyR2 channel expression is crucial for spatial memory processes. Antioxid. Redox Signal. 29, 1125-1146.
Collapse
Affiliation(s)
- Jamileth Y More
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Barbara A Bruna
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pedro E Lobos
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Galaz
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Paula L Figueroa
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Silvia Namias
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Gina L Sánchez
- 2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Genaro C Barrientos
- 2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Valdés
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,3 Department of Neuroscience, Faculty of Medicine, Universidad de Chile , Santiago, Chile
| | - Andrea C Paula-Lima
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,4 Institute for Research in Dental Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile .,3 Department of Neuroscience, Faculty of Medicine, Universidad de Chile , Santiago, Chile .,5 Center for Exercise , Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Tatiana Adasme
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,6 Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins , Santiago, Chile
| |
Collapse
|
27
|
Miranda M, Kent BA, Morici JF, Gallo F, Saksida LM, Bussey TJ, Weisstaub N, Bekinschtein P. NMDA receptors and BDNF are necessary for discrimination of overlapping spatial and non-spatial memories in perirhinal cortex and hippocampus. Neurobiol Learn Mem 2018; 155:337-343. [PMID: 30172952 DOI: 10.1016/j.nlm.2018.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/03/2018] [Accepted: 08/29/2018] [Indexed: 01/06/2023]
Abstract
Successful memory involves not only remembering information over time but also keeping memories distinct and less confusable. Discrimination of overlapping representations has been investigated in the dentate gyrus (DG) of the hippocampus and largely in the perirhinal cortex (Prh). In particular, the DG was shown to be important for discrimination of overlapping spatial memories and Prh was shown to be important for discrimination of overlapping object memories. In the present study, we used both a DG-dependent and a Prh-dependent task and manipulated the load of similarity between either spatial or object stimuli during information encoding. We showed that N-methyl-D-aspartate-type glutamate receptors (NMDAr) and BDNF participate of the same cellular network during consolidation of both overlapping object and spatial memories in the Prh and DG, respectively. This argues in favor of conserved cellular mechanisms across regions despite anatomical differences.
Collapse
Affiliation(s)
- Magdalena Miranda
- Laboratory of Memory Research and Molecular Cognition, Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro, Buenos Aires, Argentina
| | - Brianne A Kent
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Juan Facundo Morici
- Laboratory of Memory Research and Molecular Cognition, Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro, Buenos Aires, Argentina
| | - Francisco Gallo
- Laboratory of Memory Research and Molecular Cognition, Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro, Buenos Aires, Argentina
| | - Lisa M Saksida
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK; Molecular Medicine Research Laboratories, Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; The Brain and Mind Institute, Western University, London, ON, Canada
| | - Timothy J Bussey
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK; Molecular Medicine Research Laboratories, Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; The Brain and Mind Institute, Western University, London, ON, Canada
| | - Noelia Weisstaub
- Laboratory of Memory Research and Molecular Cognition, Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro, Buenos Aires, Argentina
| | - Pedro Bekinschtein
- Laboratory of Memory Research and Molecular Cognition, Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación INECO - Universidad Favaloro, Buenos Aires, Argentina.
| |
Collapse
|
28
|
Srivastava P, Dhuriya YK, Kumar V, Srivastava A, Gupta R, Shukla RK, Yadav RS, Dwivedi HN, Pant AB, Khanna VK. PI3K/Akt/GSK3β induced CREB activation ameliorates arsenic mediated alterations in NMDA receptors and associated signaling in rat hippocampus: Neuroprotective role of curcumin. Neurotoxicology 2018; 67:190-205. [PMID: 29723552 DOI: 10.1016/j.neuro.2018.04.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 04/26/2018] [Accepted: 04/29/2018] [Indexed: 12/20/2022]
Abstract
Protective efficacy of curcumin in arsenic induced NMDA receptor dysfunctions and PI3K/Akt/ GSK3β signalling in hippocampus has been investigated in vivo and in vitro. Exposure to sodium arsenite (in vivo - 20 mg/kg, body weight p.o. for 28 days; in vitro - 10 μM for 24 h) and curcumin (in vivo - 100 mg/kg body weight p.o. for 28 days; in vitro - 20 μM for 24 h) was carried out alone or simultaneously. Treatment with curcumin ameliorated sodium arsenite induced alterations in the levels of NMDA receptors, its receptor subunits and synaptic proteins - pCaMKIIα, PSD-95 and SynGAP both in vivo and in vitro. Decreased levels of BDNF, pAkt, pERK1/2, pGSK3β and pCREB on sodium arsenite exposure were also protected by curcumin. Curcumin was found to decrease sodium arsenite induced changes in hippocampus by modulating PI3K/Akt/GSK3β neuronal survival pathway, known to regulate various cellular events. Treatment of hippocampal cultures with pharmacological inhibitors for ERK1/2, GSK3β and Akt individually inhibited levels of CREB and proteins associated with PI3K/Akt/GSK3β pathway. Simultaneous treatment with curcumin was found to improve sodium arsenite induced learning and memory deficits in rats assessed by water maze and Y-maze. The results provide evidence that curcumin exercises its neuroprotective effect involving PI3K/Akt pathway which may affect NMDA receptors and downstream signalling through TrKβ and BDNF in arsenic induced cognitive deficits in hippocampus.
Collapse
Affiliation(s)
- Pranay Srivastava
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India; School of Pharmacy, Babu Banarsi Das University, Faizabad Road, Lucknow, 226 028, UP, India
| | - Yogesh K Dhuriya
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India
| | - Vivek Kumar
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Brazil
| | - Akriti Srivastava
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India
| | - Richa Gupta
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India; School of Pharmacy, Babu Banarsi Das University, Faizabad Road, Lucknow, 226 028, UP, India
| | - Rajendra K Shukla
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India
| | - Rajesh S Yadav
- Department of Criminology and Forensic Science, Dr. Harisingh Gour Central University, Sagar, 470003, MP, India
| | - Hari N Dwivedi
- School of Pharmacy, Babu Banarsi Das University, Faizabad Road, Lucknow, 226 028, UP, India
| | - Aditya B Pant
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India.
| | - Vinay K Khanna
- Developmental Toxicology and NeuroToxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, UP, India.
| |
Collapse
|
29
|
Nookala AR, Schwartz DC, Chaudhari NS, Glazyrin A, Stephens EB, Berman NEJ, Kumar A. Methamphetamine augment HIV-1 Tat mediated memory deficits by altering the expression of synaptic proteins and neurotrophic factors. Brain Behav Immun 2018; 71:37-51. [PMID: 29729322 PMCID: PMC6003882 DOI: 10.1016/j.bbi.2018.04.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 01/06/2023] Open
Abstract
Methamphetamine (METH) abuse is common among individuals infected with HIV-1 and has been shown to affect HIV replication and pathogenesis. These HIV-1 infected individuals also exhibit greater neuronal injury and higher cognitive decline. HIV-1 proteins, specifically gp120 and HIV-1 Tat, have been earlier shown to affect neurocognition. HIV-1 Tat, a viral protein released early during HIV-1 replication, contributes to HIV-associated neurotoxicity through various mechanisms including production of pro-inflammatory cytokines, reactive oxygen species and dysregulation of neuroplasticity. However, the combined effect of METH and HIV-1 Tat on neurocognition and its potential effect on neuroplasticity mechanisms remains largely unknown. Therefore, the present study was undertaken to investigate the combined effect of METH and HIV-1 Tat on behavior and on the expression of neuroplasticity markers by utilizing Doxycycline (DOX)-inducible HIV-1 Tat (1-86) transgenic mice. Expression of Tat in various brain regions of these mice was confirmed by RT-PCR. The mice were administered with an escalating dose of METH (0.1 mg/kg to 6 mg/kg, i.p) over a 7-day period, followed by 6 mg/kg, i.p METH twice a day for four weeks. After three weeks of METH administration, Y maze and Morris water maze assays were performed to determine the effect of Tat and METH on working and spatial memory, respectively. Compared with controls, working memory was significantly decreased in Tat mice that were administered METH. Moreover, significant deficits in spatial memory were also observed in Tat-Tg mice that were administered METH. A significant reduction in the protein expressions of synapsin 1, synaptophysin, Arg3.1, PSD-95, and BDNF in different brain regions were also observed. Expression levels of Calmodulin kinase II (CaMKII), a marker of synaptodendritic integrity, were also significantly decreased in HIV-1 Tat mice that were treated with METH. Together, this data suggests that METH enhances HIV-1 Tat-induced memory deficits by reducing the expression of pre- and postsynaptic proteins and neuroplasticity markers, thus providing novel insights into the molecular mechanisms behind neurocognitive impairments in HIV-infected amphetamine users.
Collapse
Affiliation(s)
- Anantha Ram Nookala
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Daniel C. Schwartz
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Nitish S. Chaudhari
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Alexy Glazyrin
- Department of Pathology, School of Medicine, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Edward B. Stephens
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nancy E. J. Berman
- Department of Anatomy and Cell biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anil Kumar
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA.
| |
Collapse
|
30
|
Alzoubi KH, Mokhemer E, Abuirmeileh AN. Beneficial effect of etazolate on depression-like behavior and, learning, and memory impairment in a model of Parkinson's disease. Behav Brain Res 2018; 350:109-115. [PMID: 29758248 DOI: 10.1016/j.bbr.2018.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 02/05/2023]
Abstract
The aim of this study was to evaluate etazolate against depression-like behavior and, learning and memory impairment induced by 6- hydroxydopamine (6-OHDA) rat model of Parkinson's disease (PD). This aim was achieved through comparing 6-OHDA lesioned rats in the presence and absence of etazolate. The 6-OHDA was used to induce lesion as a model of PD. Etazolate was administered at a dose of 1 mg/kg/day for 14 days, starting 7 days after lesion induction. Apomorphine-induced rotation test was used to evaluate 6-OHDA-induced motor deficits, tail suspension test was used to assess depression-like symptoms, and the radial arms water maze (RAWM) was used to evaluate special learning and memory functions. Antioxidant biomarkers and BDNF protein levels were assessed in the hippocampus. Results revealed that etazolate administration significantly improved 6-OHDA-induced PD related symptoms including motor deficits, depression-like behavior and impairment of both short- and long- term memory. Moreover, etazolate significantly prevented 6-OHDA-induced reduction in oxidative stress biomarkers (GSH/GSSG ratio, GPx) and BDNF levels. In conclusion, motor dysfunction, depressive- like behavior, and learning and memory deficits in the 6-OHDA rat model of PD can be significantly prevented by etazolate. This prevention could be attributed to etazolate's ability to prevent reduction in antioxidative stress biomarkers and BDNF levels.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan.
| | - Enas Mokhemer
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Amjad N Abuirmeileh
- Department of Applied Pharmaceutical Sciences, Faculty of Pharmacy, Al-Isra University, Amman, Jordan
| |
Collapse
|
31
|
Dhanda S, Gupta S, Halder A, Sunkaria A, Sandhir R. Systemic inflammation without gliosis mediates cognitive deficits through impaired BDNF expression in bile duct ligation model of hepatic encephalopathy. Brain Behav Immun 2018. [PMID: 29518527 DOI: 10.1016/j.bbi.2018.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease per se induces neuroinflammation that contributes to cognitive deficits in hepatic encephalopathy (HE). However, the processes by which pro-inflammatory molecules result in cognitive impairment still remains unclear. In the present study, a significant increase in the activity of liver function enzymes viz. alanine transaminase (ALT), aspartate transaminase (AST) and alkaline phosphatase (ALP) was observed along with increase in plasma ammonia levels after four weeks of bile duct ligation (BDL) in rats suggesting hepatocellular damage. A significant increase was observed in mRNA expression of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) in brain regions and liver of BDL rats. Concomitantly, IL-6, TNF-α and MCP-1 protein levels were also increased in brain regions, liver and serum of BDL rats suggesting the involvement of blood-brain-axis in inflammatory response. However, a significant decrease was observed in glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule-1 (Iba-1) expression at transcriptional and translation level in brain of BDL rats. Immunohistochemical and flowcytometric analysis revealed reduced number of GFAP-immunopositive astrocytes and Iba1-immunopositive microglia in the brain regions of BDL rats. Further, a significant decline was observed in cognitive functions in BDL rats assessed using Morris water maze and novel object recognition tests. Expression of pro and mature form of brain derived neurotrophic factor (BDNF) and its upstream transcription element showed significant reduction in brain of BDL rats. Taken together, the results of the present study suggest that systemic inflammation and reduced expression of BDNF and its upstream transcription factor plays a key role in cognitive decline in HE.
Collapse
Affiliation(s)
- Saurabh Dhanda
- Department of Biochemistry, Basic Medical Sciences Block-II, Sector-25, Panjab University, Chandigarh 160014, India
| | - Smriti Gupta
- Department of Biochemistry, Basic Medical Sciences Block-II, Sector-25, Panjab University, Chandigarh 160014, India
| | - Avishek Halder
- Department of Biochemistry, Basic Medical Sciences Block-II, Sector-25, Panjab University, Chandigarh 160014, India
| | - Aditya Sunkaria
- Department of Biochemistry, Basic Medical Sciences Block-II, Sector-25, Panjab University, Chandigarh 160014, India
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Sciences Block-II, Sector-25, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
32
|
Lindsay JH, Prosser RA. The Mammalian Circadian Clock Exhibits Chronic Ethanol Tolerance and Withdrawal-Induced Glutamate Hypersensitivity, Accompanied by Changes in Glutamate and TrkB Receptor Proteins. Alcohol Clin Exp Res 2017; 42:315-328. [PMID: 29139560 DOI: 10.1111/acer.13554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/07/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Alcohol tolerance and withdrawal-induced effects are criteria for alcohol use disorders listed by the DSM-V. Although tolerance and withdrawal have been studied over many decades, there is still uncertainty regarding mechanistic distinctions that characterize these different forms of ethanol (EtOH)-induced plasticity. Previously, we demonstrated that the suprachiasmatic nucleus (SCN) circadian clock develops both acute and rapid tolerance to EtOH inhibition of glutamate-induced circadian phase shifts. Here, we demonstrate that chronic EtOH tolerance and withdrawal-induced glutamate hypersensitivity occur in vitro and that rapid tolerance, chronic tolerance, and glutamate hypersensitivity have distinct cellular changes. METHODS We use single-unit extracellular electrophysiological recordings to determine whether chronic tolerance to EtOH inhibition of glutamatergic phase shifts and withdrawal-induced glutamate hypersensitivity develop in the SCN. We use Western blotting to compare phosphorylation state and total expression of N-methyl-D-aspartate (NMDA) receptor subunits and associated proteins in the SCN after mice were exposed to varying EtOH consumption paradigms. RESULTS Chronic tolerance developed after a minimum of 8 days of 4 h/d EtOH access, as indicated by a decreased sensitivity to EtOH inhibition of glutamate-induced phase shifts. We also observed an increased sensitivity to glutamate-induced phase shifts in SCN tissue following withdrawal. We demonstrated an increase in the ratio of NR2B:NR2A NMDA receptor subunit expression after 21 days, but not after 10 days of EtOH drinking. This increase persisted during EtOH withdrawal, along with an increase in NR2B Y1472 phosphorylation, mature brain-derived neurotrophic factor, and phosphorylated TrkB. CONCLUSIONS These results demonstrate that multiple tolerance forms and withdrawal-induced glutamate hypersensitivity occur in the SCN and that these different forms of EtOH-induced plasticity are accompanied by distinct changes in cellular physiology. Importantly, this study further demonstrates the power of using the SCN as a model system to investigate EtOH-induced plasticity.
Collapse
Affiliation(s)
- Jonathan H Lindsay
- Department of Biochemistry and Cellular and Molecular Biology (JHL, RAP), University of Tennessee Knoxville, NeuroNET Research Center, Knoxville, Tennessee
| | - Rebecca A Prosser
- Department of Biochemistry and Cellular and Molecular Biology (JHL, RAP), University of Tennessee Knoxville, NeuroNET Research Center, Knoxville, Tennessee
| |
Collapse
|
33
|
Yiannakas A, Rosenblum K. The Insula and Taste Learning. Front Mol Neurosci 2017; 10:335. [PMID: 29163022 PMCID: PMC5676397 DOI: 10.3389/fnmol.2017.00335] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/03/2017] [Indexed: 12/29/2022] Open
Abstract
The sense of taste is a key component of the sensory machinery, enabling the evaluation of both the safety as well as forming associations regarding the nutritional value of ingestible substances. Indicative of the salience of the modality, taste conditioning can be achieved in rodents upon a single pairing of a tastant with a chemical stimulus inducing malaise. This robust associative learning paradigm has been heavily linked with activity within the insular cortex (IC), among other regions, such as the amygdala and medial prefrontal cortex. A number of studies have demonstrated taste memory formation to be dependent on protein synthesis at the IC and to correlate with the induction of signaling cascades involved in synaptic plasticity. Taste learning has been shown to require the differential involvement of dopaminergic GABAergic, glutamatergic, muscarinic neurotransmission across an extended taste learning circuit. The subsequent activation of downstream protein kinases (ERK, CaMKII), transcription factors (CREB, Elk-1) and immediate early genes (c-fos, Arc), has been implicated in the regulation of the different phases of taste learning. This review discusses the relevant neurotransmission, molecular signaling pathways and genetic markers involved in novel and aversive taste learning, with a particular focus on the IC. Imaging and other studies in humans have implicated the IC in the pathophysiology of a number of cognitive disorders. We conclude that the IC participates in circuit-wide computations that modulate the interception and encoding of sensory information, as well as the formation of subjective internal representations that control the expression of motivated behaviors.
Collapse
Affiliation(s)
- Adonis Yiannakas
- Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
| | - Kobi Rosenblum
- Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| |
Collapse
|
34
|
Role of Src Family Kinases in BDNF-Mediated Suppression of Cocaine-Seeking and Prevention of Cocaine-Induced ERK, GluN2A, and GluN2B Dephosphorylation in the Prelimbic Cortex. Neuropsychopharmacology 2017; 42:1972-1980. [PMID: 28585567 PMCID: PMC5561338 DOI: 10.1038/npp.2017.114] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/23/2017] [Accepted: 05/27/2017] [Indexed: 01/17/2023]
Abstract
Models of relapse have demonstrated that neuroadaptations in reward circuits following cocaine self-administration (SA) underlie reinstatement of drug-seeking. Dysregulation of the pathway from the prelimbic (PrL) cortex to the nucleus accumbens is implicated in reinstatement. A single BDNF infusion into the PrL cortex following a final cocaine SA session results in attenuation of reinstatement of cocaine-seeking. Inhibiting BDNF's receptor, TrkB, ERK/MAP kinase activation, or NMDA receptors blocks this attenuating effect, indicating that the interaction between glutamate-mediated synaptic activity and TrkB signaling is imperative to BDNF's suppressive effect on drug-seeking. Src family kinases (SFKs) are involved in both NMDA-mediated activation of TrkB- and TrkB-mediated tyrosine phosphorylation of NMDA receptors. We hypothesized that infusion of the SFK inhibitor, PP2, into the PrL cortex prior to a BDNF infusion, immediately after the end of the last cocaine SA session, would block BDNF's ability to suppress reinstatement of cocaine-seeking in rats with a cocaine SA history. PP2, but not the negative control, PP3, blocked BDNF's suppressive effect on context-induced relapse after 1 week of abstinence and cue-induced reinstatement after extinction. As previously reported, infusion of BDNF into the PrL cortex blocked cocaine SA-induced dephosphorylation of ERK, GluN2A, and GluN2B-containing receptors. Inhibition of SFKs using PP2 blocked BDNF-mediated phosphorylation of GluN2A, GluN2B, and ERK. These data indicate that SFK activity is necessary for BDNF-mediated suppression of cocaine-seeking and reversal of cocaine-induced dephosphorylation of key phosphoproteins in the prefrontal cortex related to synaptic plasticity.
Collapse
|
35
|
Cognitive impairment and gene expression alterations in a rodent model of binge eating disorder. Physiol Behav 2017; 180:78-90. [PMID: 28821448 DOI: 10.1016/j.physbeh.2017.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
Binge eating disorder (BED) is defined as recurrent, distressing over-consumption of palatable food (PF) in a short time period. Clinical studies suggest that individuals with BED may have impairments in cognitive processes, executive functioning, impulse control, and decision-making, which may play a role in sustaining binge eating behavior. These clinical reports, however, are limited and often conflicting. In this study, we used a limited access rat model of binge-like behavior in order to further explore the effects of binge eating on cognition. In binge eating prone (BEP) rats, we found novel object recognition (NOR) as well as Barnes maze reversal learning (BM-RL) deficits. Aberrant gene expression of brain derived neurotrophic factor (Bdnf) and tropomyosin receptor kinase B (TrkB) in the hippocampus (HPC)-prefrontal cortex (PFC) network was observed in BEP rats. Additionally, the NOR deficits were correlated with reductions in the expression of TrkB and insulin receptor (Ir) in the CA3 region of the hippocampus. Furthermore, up-regulation of serotonin-2C (5-HT2C) receptors in the orbitoprefrontal cortex (OFC) was associated with BM-RL deficit. Finally, in the nucleus accumbens (NAc), we found decreased dopamine receptor 2 (Drd2) expression among BEP rats. Taken together, these data suggest that binge eating vegetable shortening may induce contextual and reversal learning deficits which may be mediated, at least in part, by the altered expression of genes in the CA3-OFC-NAc neural network.
Collapse
|
36
|
Zaletel I, Filipović D, Puškaš N. Hippocampal BDNF in physiological conditions and social isolation. Rev Neurosci 2017; 28:675-692. [DOI: 10.1515/revneuro-2016-0072] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/16/2017] [Indexed: 12/19/2022]
Abstract
AbstractExposure of an organism to chronic psychosocial stress may affect brain-derived neurotrophic factor (BDNF) expression that has been implicated in the etiology of psychiatric disorders, such as depression. Given that depression in humans has been linked with social stress, the chronic social stress paradigms for modeling psychiatric disorders in animals have thus been developed. Chronic social isolation in animal models generally causes changes in hypothalamic-pituitary-adrenal axis functioning, associated with anxiety- and depressive-like behaviors. Also, this chronic stress causes downregulation of BDNF protein and mRNA in the hippocampus, a stress-sensitive brain region closely related to the pathophysiology of depression. In this review, we discuss the current knowledge regarding the structure, function, intracellular signaling, inter-individual differences and epigenetic regulation of BDNF in both physiological conditions and depression and changes in corticosterone levels, as a marker of stress response. Since BDNF levels are age dependent in humans and rodents, this review will also highlight the effects of adolescent and adult chronic social isolation models of both genders on the BDNF expression.
Collapse
Affiliation(s)
- Ivan Zaletel
- Institute of Histology and Embryology “Aleksandar Ð. Kostić”, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dragana Filipović
- Laboratory of Molecular Biology and Endocrinology, Institute of Nuclear Sciences “Vinča”, University of Belgrade, 11000 Belgrade, Serbia
| | - Nela Puškaš
- Institute of Histology and Embryology “Aleksandar Ð. Kostić”, School of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| |
Collapse
|
37
|
BDNF Contributes to Spinal Long-Term Potentiation and Mechanical Hypersensitivity Via Fyn-Mediated Phosphorylation of NMDA Receptor GluN2B Subunit at Tyrosine 1472 in Rats Following Spinal Nerve Ligation. Neurochem Res 2017; 42:2712-2729. [DOI: 10.1007/s11064-017-2274-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/01/2017] [Accepted: 04/18/2017] [Indexed: 11/26/2022]
|
38
|
Li SW, Chen YC, Sheen JM, Hsu MH, Tain YL, Chang KA, Huang LT. Minocycline restores cognitive-relative altered proteins in young bile duct-ligated rat prefrontal cortex. Life Sci 2017; 180:75-82. [PMID: 28366719 DOI: 10.1016/j.lfs.2017.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/24/2017] [Accepted: 03/29/2017] [Indexed: 01/06/2023]
Abstract
AIMS Bile duct ligation (BDL) model is used to study hepatic encephalopathy accompanied by cognitive impairment. We employed the proteomic analysis approach to evaluate cognition-related proteins in the prefrontal cortex of young BDL rats and analyzed the effect of minocycline on these proteins and spatial memory. MAIN METHODS BDL was induced in young rats at postnatal day 17. Minocycline as a slow-release pellet was implanted into the peritoneum. Morris water maze test and two-dimensional liquid chromatography-tandem mass spectrometry were used to evaluate spatial memory and prefrontal cortex protein expression, respectively. We used 2D/LC-MS/MS to analyze for affected proteins in the prefrontal cortex of young BDL rats. Results were verified with Western blotting, immunohistochemistry, and quantitative real-time PCR. The effect of minocycline in BDL rats was assessed. KEY FINDINGS BDL induced spatial deficits, while minocycline rescued it. Collapsin response mediator protein 2 (CRMP2) and manganese-dependent superoxide dismutase (MnSOD) were upregulated and nucleoside diphosphate kinase B (NME2) was downregulated in young BDL rats. BDL rats exhibited decreased levels of brain-derived neurotrophic factor (BDNF) mRNA as compared with those by the control. However, minocycline treatment restored CRMP2 and NME2 protein expression, BDNF mRNA level, and MnSOD activity to control levels. SIGNIFICANCE We demonstrated that BDL altered the expression of CRMP2, NME2, MnSOD, and BDNF in the prefrontal cortex of young BDL rats. However, minocycline treatment restored the expression of the affected mediators that are implicated in cognition.
Collapse
Affiliation(s)
- Shih-Wen Li
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Chieh Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Mei-Hsin Hsu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kow-Aung Chang
- Department of Anesthesiolgy, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Traditional Medicine, Chang Gung University, Linkow, Taiwan.
| |
Collapse
|
39
|
Strength and Aerobic Exercises Improve Spatial Memory in Aging Rats Through Stimulating Distinct Neuroplasticity Mechanisms. Mol Neurobiol 2016; 54:7928-7937. [DOI: 10.1007/s12035-016-0272-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/30/2016] [Indexed: 01/04/2023]
|
40
|
Coronas-Samano G, Baker KL, Tan WJT, Ivanova AV, Verhagen JV. Fus1 KO Mouse As a Model of Oxidative Stress-Mediated Sporadic Alzheimer's Disease: Circadian Disruption and Long-Term Spatial and Olfactory Memory Impairments. Front Aging Neurosci 2016; 8:268. [PMID: 27895577 PMCID: PMC5108791 DOI: 10.3389/fnagi.2016.00268] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022] Open
Abstract
Insufficient advances in the development of effective therapeutic treatments of sporadic Alzheimer's Disease (sAD) to date are largely due to the lack of sAD-relevant animal models. While the vast majority of models do recapitulate AD's hallmarks of plaques and tangles by virtue of tau and/or beta amyloid overexpression, these models do not reflect the fact that in sAD (unlike familial AD) these genes are not risk factors per se and that other mechanisms like oxidative stress, metabolic dysregulation and inflammation play key roles in AD etiology. Here we characterize and propose the Fus1 KO mice that lack a mitochondrial protein Fus1/Tusc2 as a new sAD model. To establish sAD relevance, we assessed sAD related deficits in Fus1 KO and WT adult mice of 4-5 months old, the equivalent human age when the earliest cognitive and olfactory sAD symptoms arise. Fus1 KO mice showed oxidative stress (increased levels of ROS, decreased levels of PRDX1), disruption of metabolic homeostasis (decreased levels of ACC2, increased phosphorylation of AMPK), autophagy (decreased levels of LC3-II), PKC (decreased levels of RACK1) and calcium signaling (decreased levels of Calb2) in the olfactory bulb and/or hippocampus. Mice were behaviorally tested using objective and accurate video tracking (Noldus), in which Fus1 KO mice showed clear deficits in olfactory memory (decreased habituation/cross-habituation in the short and long term), olfactory guided navigation memory (inability to reduce their latency to find the hidden cookie), spatial memory (learning impairments on finding the platform in the Morris water maze) and showed more sleep time during the diurnal cycle. Fus1 KO mice did not show clear deficits in olfactory perception (cross-habituation), association memory (passive avoidance) or in species-typical behavior (nest building) and no increased anxiety (open field, light-dark box) or depression/anhedonia (sucrose preference) at this relatively young age. These neurobehavioral deficits of the Fus1 KO mice at this relatively young age are highly relevant to sAD, making them suitable for effective research on pharmacological targets in the context of early intervention of sAD.
Collapse
Affiliation(s)
| | - Keeley L Baker
- The John B. Pierce LaboratoryNew Haven, CT, USA; Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA
| | - Winston J T Tan
- Department of Surgery, Yale University School of Medicine New Haven, CT, USA
| | - Alla V Ivanova
- Department of Surgery, Yale University School of Medicine New Haven, CT, USA
| | - Justus V Verhagen
- The John B. Pierce LaboratoryNew Haven, CT, USA; Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA
| |
Collapse
|
41
|
The Gut-Brain Axis, BDNF, NMDA and CNS Disorders. Neurochem Res 2016; 41:2819-2835. [PMID: 27553784 DOI: 10.1007/s11064-016-2039-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/09/2016] [Accepted: 08/17/2016] [Indexed: 02/08/2023]
Abstract
Gastro-intestinal (GI) microbiota and the 'gut-brain axis' are proving to be increasingly relevant to early brain development and the emergence of psychiatric disorders. This review focuses on the influence of the GI tract on Brain-Derived Neurotrophic Factor (BDNF) and its relationship with receptors for N-methyl-D-aspartate (NMDAR), as these are believed to be involved in synaptic plasticity and cognitive function. NMDAR may be associated with the development of schizophrenia and a range of other psychopathologies including neurodegenerative disorders, depression and dementias. An analysis of the routes and mechanisms by which the GI microbiota contribute to the pathophysiology of BDNF-induced NMDAR dysfunction could yield new insights relevant to developing novel therapeutics for schizophrenia and related disorders. In the absence of GI microbes, central BDNF levels are reduced and this inhibits the maintenance of NMDAR production. A reduction of NMDAR input onto GABA inhibitory interneurons causes disinhibition of glutamatergic output which disrupts the central signal-to-noise ratio and leads to aberrant synaptic behaviour and cognitive deficits. Gut microbiota can modulate BDNF function in the CNS, via changes in neurotransmitter function by affecting modulatory mechanisms such as the kynurenine pathway, or by changes in the availability and actions of short chain fatty acids (SCFAs) in the brain. Interrupting these cycles by inducing changes in the gut microbiota using probiotics, prebiotics or antimicrobial drugs has been found promising as a preventative or therapeutic measure to counteract behavioural deficits and these may be useful to supplement the actions of drugs in the treatment of CNS disorders.
Collapse
|
42
|
Stucky A, Bakshi KP, Friedman E, Wang HY. Prenatal Cocaine Exposure Upregulates BDNF-TrkB Signaling. PLoS One 2016; 11:e0160585. [PMID: 27494324 PMCID: PMC4975466 DOI: 10.1371/journal.pone.0160585] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/21/2016] [Indexed: 12/11/2022] Open
Abstract
Prenatal cocaine exposure causes profound changes in neurobehavior as well as synaptic function and structure with compromised glutamatergic transmission. Since synaptic health and glutamatergic activity are tightly regulated by brain-derived neurotrophic factor (BDNF) signaling through its cognate tyrosine receptor kinase B (TrkB), we hypothesized that prenatal cocaine exposure alters BDNF-TrkB signaling during brain development. Here we show prenatal cocaine exposure enhances BDNF-TrkB signaling in hippocampus and prefrontal cortex (PFCX) of 21-day-old rats without affecting the expression levels of TrkB, P75NTR, signaling molecules, NMDA receptor—NR1 subunit as well as proBDNF and BDNF. Prenatal cocaine exposure reduces activity-dependent proBDNF and BDNF release and elevates BDNF affinity for TrkB leading to increased tyrosine-phosphorylated TrkB, heightened Phospholipase C-γ1 and N-Shc/Shc recruitment and higher downstream PI3K and ERK activation in response to ex vivo BDNF. The augmented BDNF-TrkB signaling is accompanied by increases in association between activated TrkB and NMDARs. These data suggest that cocaine exposure during gestation upregulates BDNF-TrkB signaling and its interaction with NMDARs by increasing BDNF affinity, perhaps in an attempt to restore the diminished excitatory neurotransmission.
Collapse
Affiliation(s)
- Andres Stucky
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
- Department of Biology, Neuroscience Program, Graduate School of The City University of New York, New York, New York, 10061, United States of America
| | - Kalindi P. Bakshi
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
| | - Eitan Friedman
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
| | - Hoau-Yan Wang
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
- * E-mail:
| |
Collapse
|
43
|
Becker-Krail D, Farrand AQ, Boger HA, Lavin A. Effects of fingolimod administration in a genetic model of cognitive deficits. J Neurosci Res 2016; 95:1174-1181. [PMID: 27439747 DOI: 10.1002/jnr.23799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/23/2016] [Accepted: 05/30/2016] [Indexed: 11/09/2022]
Abstract
Notwithstanding recent advances, cognitive impairments are among the most difficult-to-treat symptoms in neuropsychiatric disorders. Deficits in information processing contributing to memory and sociability impairments are found across neuropsychiatric-related disorders. Previously, we have shown that mutations in the DTNBP1 gene (encoding dystrobrevin-binding protein 1 [dysbindin-1]) lead to abnormalities in synaptic glutamate release in the prefrontal cortex (PFC) and hippocampus and to cognitive deficits; glutamatergic transmission is important for cortical recurrent excitation that allows information processing in the PFC. To investigate possible means of restoring glutamate release and improving cognitive impairments, we assess the effects of increasing endogenous levels of brain-derived neurotrophic factor (BDNF) in a dysbindin-1-deficient mouse model. Increasing endogenous levels of BDNF may aid in remediating cognitive deficits, given the roles of BDNF in synaptic transmission, plasticity, and neuroprotection. To increase BDNF, we use a novel strategy, repeated intraperitoneal injections of fingolimod (Gilenya). Sphingolipids have recently been shown to have therapeutic value in several neurology-related disorders. Both wild-type (WT) and mutant (MUT) genotypes were tested for sociability and recognition memory, followed by measuring endogenous BDNF levels and presynaptic [Ca2+ ]i within the PFC. Both genotypes were treated for 1 week with either saline or fingolimod. Relative to WT mice, MUT mice demonstrated impairments in sociability and recognition memory and lower presynaptic calcium. After fingolimod treatment, MUT mice exhibited significant improvements in sociability and recognition memory and increases in presynaptic calcium and endogenous concentrations of BDNF. These results show promise for counteracting the cognitive impairments seen in neuropsychiatric disorders and may shed light on the role of dysbindin-1. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - A Q Farrand
- Deptartment of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - H A Boger
- Deptartment of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - A Lavin
- Deptartment of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
44
|
Itoh N, Enomoto A, Nagai T, Takahashi M, Yamada K. Molecular mechanism linking BDNF/TrkB signaling with the NMDA receptor in memory: the role of Girdin in the CNS. Rev Neurosci 2016; 27:481-90. [DOI: 10.1515/revneuro-2015-0072] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/14/2016] [Indexed: 12/21/2022]
Abstract
AbstractIt is well known that synaptic plasticity is the cellular mechanism underlying learning and memory. Activity-dependent synaptic changes in electrical properties and morphology, including synaptogenesis, lead to alterations of synaptic strength, which is associated with long-term potentiation (LTP). Brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) signaling is involved in learning and memory formation by regulating synaptic plasticity. The phosphatidylinositol 3-kinase (PI3-K)/Akt pathway is one of the key signaling cascades downstream BDNF/TrkB and is believed to modulate N-methyl-d-aspartate (NMDA) receptor-mediated synaptic plasticity. However, the molecular mechanism underlying the connection between these two key players in synaptic plasticity remains largely unknown. Girders of actin filament (Girdin), an Akt substrate that directly binds to actin filaments, has been shown to play a role in neuronal migration and neuronal development. Recently, we identified Girdin as a key molecule involved in regulating long-term memory. It was demonstrated that phosphorylation of Girdin by Akt contributed to the maintenance of LTP by linking the BDNF/TrkB signaling pathway with NMDA receptor activity. These findings indicate that Girdin plays a pivotal role in a variety of processes in the CNS. Here, we review recent advances in our understanding about the roles of Girdin in the CNS and focus particularly on neuronal migration and memory.
Collapse
Affiliation(s)
| | | | - Taku Nagai
- 1Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8560, Japan
| | - Masahide Takahashi
- 2Department of Pathology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8550, Japan
| | - Kiyofumi Yamada
- 1Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8560, Japan
| |
Collapse
|
45
|
Ebajemito JK, Furlan L, Nissen C, Sterr A. Application of Transcranial Direct Current Stimulation in Neurorehabilitation: The Modulatory Effect of Sleep. Front Neurol 2016; 7:54. [PMID: 27092103 PMCID: PMC4822081 DOI: 10.3389/fneur.2016.00054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 03/24/2016] [Indexed: 12/20/2022] Open
Abstract
The relationship between sleep disorders and neurological disorders is often reciprocal, such that sleep disorders are worsened by neurological symptoms and that neurological disorders are aggravated by poor sleep. Animal and human studies further suggest that sleep disruption not only worsens single neurological symptoms but may also lead to long-term negative outcomes. This suggests that sleep may play a fundamental role in neurorehabilitation and recovery. We further propose that sleep may not only alter the efficacy of behavioral treatments but also plasticity-enhancing adjunctive neurostimulation methods, such as transcranial direct current stimulation (tDCS). At present, sleep receives little attention in the fields of neurorehabilitation and neurostimulation. In this review, we draw together the strands of evidence from both fields of research to highlight the proposition that sleep is an important parameter to consider in the application of tDCS as a primary or adjunct rehabilitation intervention.
Collapse
Affiliation(s)
- James K Ebajemito
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey , Guildford , UK
| | - Leonardo Furlan
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey , Guildford , UK
| | - Christoph Nissen
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical Center , Freiburg , Germany
| | - Annette Sterr
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; Department of Neurology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
46
|
Parikh V, Naughton SX, Yegla B, Guzman DM. Impact of partial dopamine depletion on cognitive flexibility in BDNF heterozygous mice. Psychopharmacology (Berl) 2016; 233:1361-75. [PMID: 26861892 PMCID: PMC4814303 DOI: 10.1007/s00213-016-4229-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/30/2016] [Indexed: 12/17/2022]
Abstract
RATIONALE Cognitive flexibility is a key component of executive function and is disrupted in major psychiatric disorders. Brain-derived neurotrophic factor (BDNF) exerts neuromodulatory effects on synaptic transmission and cognitive/affective behaviors. However, the causal mechanisms linking BDNF hypofunction with executive deficits are not well understood. OBJECTIVES Here, we assessed the consequences of BDNF hemizygosity on cognitive flexibility in mice performing an operant conditioning task. As dopaminergic-glutamatergic interaction in the striatum is important for cognitive processing, and BDNF heterozygous (BDNF(+/-)) mice display a higher dopamine tone in the dorsal striatum, we also assessed the effects of partial striatal dopamine depletion on task performance and glutamate release. RESULTS BDNF(+/-) mice acquired discrimination learning as well as new rule learning during set-shifting as efficiently as wild-type mice. However, partial removal of striatal dopaminergic inputs with 6-hydroxydopamine (6-OHDA) impaired these cognitive processes by impeding the maintenance of a new learning strategy in both genotypes. BDNF mutants exhibited performance impairments during reversal learning, and these deficits were associated with increased perseveration to the previously acquired strategy. Partial dopamine depletion of the striatum reversed these cognitive impairments. Additionally, reduction in depolarization-evoked glutamate release noted in the dorsal striatum of BDNF(+/-) mice was not observed in 6-OHDA-infused BDNF mutants indicating normalization of glutamatergic transmission in these animals. CONCLUSIONS Our data illustrate that BDNF signaling regulates cognitive control processes presumably by maintaining striatal dopamine-glutamate balance. Moreover, aberrations in BDNF signaling may act as a common neurobiological substrate that accounts for executive dysfunction observed in multiple psychiatric conditions.
Collapse
Affiliation(s)
- Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA.
| | | | | | | |
Collapse
|
47
|
Scholz-Starke J, Cesca F. Stepping Out of the Shade: Control of Neuronal Activity by the Scaffold Protein Kidins220/ARMS. Front Cell Neurosci 2016; 10:68. [PMID: 27013979 PMCID: PMC4789535 DOI: 10.3389/fncel.2016.00068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/01/2016] [Indexed: 12/31/2022] Open
Abstract
The correct functioning of the nervous system depends on the exquisitely fine control of neuronal excitability and synaptic plasticity, which relies on an intricate network of protein-protein interactions and signaling that shapes neuronal homeostasis during development and in adulthood. In this complex scenario, Kinase D interacting substrate of 220 kDa/ankyrin repeat-rich membrane spanning (Kidins220/ARMS) acts as a multi-functional scaffold protein with preferential expression in the nervous system. Engaged in a plethora of interactions with membrane receptors, cytosolic signaling components and cytoskeletal proteins, Kidins220/ARMS is implicated in numerous cellular functions including neuronal survival, neurite outgrowth and maturation and neuronal activity, often in the context of neurotrophin (NT) signaling pathways. Recent studies have highlighted a number of cell- and context-specific roles for this protein in the control of synaptic transmission and neuronal excitability, which are at present far from being completely understood. In addition, some evidence has began to emerge, linking alterations of Kidins220 expression to the onset of various neurodegenerative diseases and neuropsychiatric disorders. In this review, we present a concise summary of our fragmentary knowledge of Kidins220/ARMS biological functions, focusing on the mechanism(s) by which it controls various aspects of neuronal activity. We have tried, where possible, to discuss the available evidence in the wider context of NT-mediated regulation, and to outline emerging roles of Kidins220/ARMS in human pathologies.
Collapse
Affiliation(s)
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia Genova, Italy
| |
Collapse
|
48
|
Aarse J, Herlitze S, Manahan-Vaughan D. The requirement of BDNF for hippocampal synaptic plasticity is experience-dependent. Hippocampus 2016; 26:739-51. [PMID: 26662461 PMCID: PMC5066736 DOI: 10.1002/hipo.22555] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2015] [Indexed: 12/05/2022]
Abstract
Brain‐derived neurotrophic factor (BDNF) supports neuronal survival, growth, and differentiation and has been implicated in forms of hippocampus‐dependent learning. In vitro, a specific role in hippocampal synaptic plasticity has been described, although not all experience‐dependent forms of synaptic plasticity critically depend on BDNF. Synaptic plasticity is likely to enable long‐term synaptic information storage and memory, and the induction of persistent (>24 h) forms, such as long‐term potentiation (LTP) and long‐term depression (LTD) is tightly associated with learning specific aspects of a spatial representation. Whether BDNF is required for persistent (>24 h) forms of LTP and LTD, and how it contributes to synaptic plasticity in the freely behaving rodent has never been explored. We examined LTP, LTD, and related forms of learning in the CA1 region of freely dependent mice that have a partial knockdown of BDNF (BDNF+/−). We show that whereas early‐LTD (<90min) requires BDNF, short‐term depression (<45 min) does not. Furthermore, BDNF is required for LTP that is induced by mild, but not strong short afferent stimulation protocols. Object‐place learning triggers LTD in the CA1 region of mice. We observed that object‐place memory was impaired and the object‐place exploration failed to induce LTD in BDNF+/− mice. Furthermore, spatial reference memory, that is believed to be enabled by LTP, was also impaired. Taken together, these data indicate that BDNF is required for specific, but not all, forms of hippocampal‐dependent information storage and memory. Thus, very robust forms of synaptic plasticity may circumvent the need for BDNF, rather it may play a specific role in the optimization of weaker forms of plasticity. The finding that both learning‐facilitated LTD and spatial reference memory are both impaired in BDNF+/− mice, suggests moreover, that it is critically required for the physiological encoding of hippocampus‐dependent memory. © 2015 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Janna Aarse
- Department of Neurophysiology, Medical Faculty.,International Graduate School of Neuroscience
| | - Stefan Herlitze
- Faculty of Biology and Biotechnology, Department of Zoology and Neurobiology Ruhr University, Bochum, 44780 Bochum, Germany
| | - Denise Manahan-Vaughan
- Department of Neurophysiology, Medical Faculty.,International Graduate School of Neuroscience
| |
Collapse
|
49
|
Luo HB, Li Y, Liu ZJ, Cao L, Zhang ZQ, Wang Y, Zhang XY, Liu Z, Shi XQ. Protective effect of tetrahydroxy stilbene glucoside on learning and memory by regulating synaptic plasticity. Neural Regen Res 2016; 11:1480-1486. [PMID: 27857754 PMCID: PMC5090853 DOI: 10.4103/1673-5374.191223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Damage to synaptic plasticity induced by neurotoxicity of amyloid-beta is regarded to be one of the pathological mechanisms of learning and memory disabilities in Alzheimer's disease patients. This study assumed that the damage of amyloid-beta to learning and memory abilities was strongly associated with the changes in the Fyn/N-methyl-D-aspartate receptor 2B (NR2B) expression. An APP695V7171 transgenic mouse model of Alzheimer's disease was used and treatment with tetrahydroxy-stilbene glucoside was administered intragastrically. Results showed that intragastric administration of tetrahydroxy-stilbene glucoside improved the learning and memory abilities of the transgenic mice through increasing NR2B receptors and Fyn expression. It also reversed parameters for synaptic interface structure of gray type I. These findings indicate that tetrahydroxy stilbene glucoside has protective effects on the brain, and has prospects for its clinical application to improve the learning and memory abilities and treat Alzheimer's disease.
Collapse
Affiliation(s)
- Hong-Bo Luo
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Yun Li
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Zun-Jing Liu
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Li Cao
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Zhi-Qiang Zhang
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Yong Wang
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Xiao-Yan Zhang
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Zhao Liu
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| | - Xiang-Qun Shi
- Department of Neurology, Lanzhou General Hospital, Lanzhou Military Area Command, Lanzhou, Gansu Province, China
| |
Collapse
|
50
|
Ju YY, Long JD, Liu Y, Liu JG. Formation of aversive memories associated with conditioned drug withdrawal requires BDNF expression in the amygdala in acute morphine-dependent rats. Acta Pharmacol Sin 2015; 36:1437-43. [PMID: 26567727 DOI: 10.1038/aps.2015.94] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/14/2015] [Indexed: 01/14/2023] Open
Abstract
AIM Brain-derived neurotrophic factor (BDNF) plays an important role in learning and memory in multiple brain areas. In the present study, we investigated the roles of BDNF in aversive memories associated with conditioned drug withdrawal in acute morphine-dependent rats. METHODS Conditioned place aversion (CPA) was induced in male SD rats exposed to a single dose of morphine (10 mg/kg, sc) followed by naloxone (0.3 mg/kg, sc). In some rats, BDNF receptor antagonist K252a (8.5 ng per side) or BDNF scavenger TrkB-FC (0.65 μg per side) was bilaterally microinjected into amygdala before naloxone injection. BDNF mRNA and protein expression levels in amygdala were detected after the behavior testing. RESULTS CPA behavior was induced in rats by the naloxone-precipitated morphine withdrawal, which was accompanied by significantly increased levels of BDNF mRNA and protein in the amygdala. Bilateral microinjection of TrkB-FC or K252a into the amygdala completely blocked CPA behavior in the rats. CONCLUSION Formation of aversive memories associated with conditioned drug withdrawal in acute morphine-dependent rats requires BDNF expression in the amygdala.
Collapse
|