1
|
Ogunro OB. An updated and comprehensive review of the health benefits and pharmacological activities of hesperidin. Biochem Biophys Res Commun 2025; 772:151974. [PMID: 40414011 DOI: 10.1016/j.bbrc.2025.151974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVES This review aims to comprehensively assess the health benefits and pharmacological activities of hesperidin, a flavonoid commonly found in citrus fruits. It consolidates recent research findings to provide insights into hesperidin's diverse health-promoting effects. KEY FINDINGS Hesperidin has gained significant attention recently for its notable pharmacological activities and potential health benefits. Studies reveal its antioxidant properties, protecting cells from oxidative damage, and its anti-inflammatory effects, inhibiting pro-inflammatory cytokines and enzymes. Also, hesperidin shows promise in cardiovascular health by reducing blood pressure and cholesterol levels and enhancing endothelial function. It also exhibits anticancer potential by hindering cell proliferation, inducing apoptosis, and suppressing tumour growth. Moreover, hesperidin demonstrates neuroprotective effects, potentially mitigating neuroinflammation and oxidative stress associated with neurodegenerative diseases. Furthermore, it displays beneficial effects in metabolic disorders such as diabetes, obesity, and fatty liver disease by influencing glucose metabolism, lipid profile, and insulin sensitivity. SUMMARY Hesperidin exhibits a wide range of health benefits and pharmacological activities, making it a promising candidate for therapeutic interventions in various diseases. Its antioxidant, anti-inflammatory, cardiovascular, anticancer, neuroprotective, and metabolic effects underscore its potential as a valuable natural compound for promoting health and preventing chronic diseases.
Collapse
Affiliation(s)
- Olalekan Bukunmi Ogunro
- Drug Discovery, Toxicology, and Pharmacology Research Laboratory, Department of Biological Sciences, KolaDaisi University, Ibadan, Nigeria.
| |
Collapse
|
2
|
Gomez-Pinilla F, Thapak P. Exercise epigenetics is fueled by cell bioenergetics: Supporting role on brain plasticity and cognition. Free Radic Biol Med 2024; 220:43-55. [PMID: 38677488 PMCID: PMC11144461 DOI: 10.1016/j.freeradbiomed.2024.04.237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/04/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Exercise has the unique aptitude to benefit overall health of body and brain. Evidence indicates that the effects of exercise can be saved in the epigenome for considerable time to elevate the threshold for various diseases. The action of exercise on epigenetic regulation seems central to building an "epigenetic memory" to influence long-term brain function and behavior. As an intrinsic bioenergetic process, exercise engages the function of the mitochondria and redox pathways to impinge upon molecular mechanisms that regulate synaptic plasticity and learning and memory. We discuss how the action of exercise uses mechanisms of bioenergetics to support a "epigenetic memory" with long-term implications for neural and behavioral plasticity. This information is crucial for directing the power of exercise to reduce the burden of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Pavan Thapak
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
3
|
Jiang SZ, Shahoha M, Zhang HY, Brancaleone W, Elkahloun A, Tejeda HA, Ashery U, Eiden LE. The guanine nucleotide exchange factor RapGEF2 is required for ERK-dependent immediate-early gene (Egr1) activation during fear memory formation. Cell Mol Life Sci 2024; 81:48. [PMID: 38236296 PMCID: PMC11071968 DOI: 10.1007/s00018-023-04999-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/16/2023] [Accepted: 10/04/2023] [Indexed: 01/19/2024]
Abstract
The MAP kinase ERK is important for neuronal plasticity underlying associative learning, yet specific molecular pathways for neuronal ERK activation are undetermined. RapGEF2 is a neuron-specific cAMP sensor that mediates ERK activation. We investigated whether it is required for cAMP-dependent ERK activation leading to other downstream neuronal signaling events occurring during associative learning, and if RapGEF2-dependent signaling impairments affect learned behavior. Camk2α-cre+/-::RapGEF2fl/fl mice with depletion of RapGEF2 in hippocampus and amygdala exhibit impairments in context- and cue-dependent fear conditioning linked to corresponding impairment in Egr1 induction in these two brain regions. Camk2α-cre+/-::RapGEF2fl/fl mice show decreased RapGEF2 expression in CA1 and dentate gyrus associated with abolition of pERK and Egr1, but not of c-Fos induction, following fear conditioning, impaired freezing to context after fear conditioning, and impaired cAMP-dependent long-term potentiation at perforant pathway and Schaffer collateral synapses in hippocampal slices ex vivo. RapGEF2 expression is largely eliminated in basolateral amygdala, also involved in fear memory, in Camk2α-cre+/-::RapGEF2fl/fl mice. Neither Egr1 nor c-fos induction in BLA after fear conditioning, nor cue-dependent fear learning, are affected by ablation of RapGEF2 in BLA. However, Egr1 induction (but not that of c-fos) in BLA is reduced after restraint stress-augmented fear conditioning, as is freezing to cue after restraint stress-augmented fear conditioning, in Camk2α-cre+/-::RapGEF2fl/fl mice. Cyclic AMP-dependent GEFs have been genetically associated as risk factors for schizophrenia, a disorder associated with cognitive deficits. Here we show a functional link between one of them, RapGEF2, and cognitive processes involved in associative learning in amygdala and hippocampus.
Collapse
Affiliation(s)
- Sunny Zhihong Jiang
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA
| | - Meishar Shahoha
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, and Sagol School of Neuroscience, Tel Aviv University, Sherman Building Rm 719, Ramat Aviv, 69978, Tel Aviv, Israel
| | - Hai-Ying Zhang
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA
| | - William Brancaleone
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA
| | | | - Hugo A Tejeda
- Unit on Neuromodulation and Synaptic Integration, NIMH-IRP, Bethesda, MD, USA
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, and Sagol School of Neuroscience, Tel Aviv University, Sherman Building Rm 719, Ramat Aviv, 69978, Tel Aviv, Israel.
| | - Lee E Eiden
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA.
| |
Collapse
|
4
|
Latchney SE, Cadney MD, Hopkins A, Garland T. Maternal upbringing and selective breeding for voluntary exercise behavior modify patterns of DNA methylation and expression of genes in the mouse brain. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12858. [PMID: 37519068 PMCID: PMC10733581 DOI: 10.1111/gbb.12858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/26/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023]
Abstract
Selective breeding has been utilized to study the genetic basis of exercise behavior, but research suggests that epigenetic mechanisms, such as DNA methylation, also contribute to this behavior. In a previous study, we demonstrated that the brains of mice from a genetically selected high runner (HR) line have sex-specific changes in DNA methylation patterns in genes known to be genomically imprinted compared to those from a non-selected control (C) line. Through cross-fostering, we also found that maternal upbringing can modify the DNA methylation patterns of additional genes. Here, we identify an additional set of genes in which DNA methylation patterns and gene expression may be altered by selection for increased wheel-running activity and maternal upbringing. We performed bisulfite sequencing and gene expression assays of 14 genes in the brain and found alterations in DNA methylation and gene expression for Bdnf, Pde4d and Grin2b. Decreases in Bdnf methylation correlated with significant increases in Bdnf gene expression in the hippocampus of HR compared to C mice. Cross-fostering also influenced the DNA methylation patterns for Pde4d in the cortex and Grin2b in the hippocampus, with associated changes in gene expression. We also found that the DNA methylation patterns for Atrx and Oxtr in the cortex and Atrx and Bdnf in the hippocampus were further modified by sex. Together with our previous study, these results suggest that DNA methylation and the resulting change in gene expression may interact with early-life influences to shape adult exercise behavior.
Collapse
Affiliation(s)
- Sarah E. Latchney
- Department of BiologySt. Mary's College of MarylandSt. Mary's CityMarylandUSA
| | - Marcell D. Cadney
- Department of Evolution, Ecology, and Organismal BiologyUniversity of CaliforniaRiversideCaliforniaUSA
- Neuroscience Research Institute, University of CaliforniaSanta BarbaraCaliforniaUSA
| | | | - Theodore Garland
- Department of Evolution, Ecology, and Organismal BiologyUniversity of CaliforniaRiversideCaliforniaUSA
| |
Collapse
|
5
|
Mucignat-Caretta C, Caretta A. Estimation of cAMP binding in hippocampus CA1 field by a fluorescent probe. Front Cell Dev Biol 2023; 11:1267956. [PMID: 37842083 PMCID: PMC10570460 DOI: 10.3389/fcell.2023.1267956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023] Open
Abstract
The hippocampus is an allocortex structure involved in many complex processes, from memory formation to spatial navigation. It starts developing during prenatal life but acquires its adult functional properties around the peripubertal age, in both humans and mice. Such prolonged maturation is accompanied by structural changes in microcircuitry and functional changes involving biochemical and electrophysiological events. Moreover, hippocampus undergoes plasticity phenomena throughout life. In murine rodents, the most relevant maturation steps in Cornu Ammonis 1 (CA1) hippocampal subfield occur during the third-fourth weeks of life. During this period, also the expression and localization of cAMP-dependent protein kinases (PKA) refines: many regulatory (R1A) PKA clusters appear, bound to the cytoskeleton. Here the binding characteristics of R1A are determined in CA1 by using confocal microscopy. Apparently, two binding sites are present with no evidence of cooperativity. Equilibrium dissociation constant is estimated around 22.9 nM. This value is lower from that estimated for R1A in soluble form, suggesting a different binding site conformation or accessibility in the tissue. The method described here may be useful to track the developmental changes in binding activity, which affects cAMP availability at selected intracellular microzones. Possible relations with functional consequences are discussed.
Collapse
Affiliation(s)
| | - Antonio Caretta
- Department of Food and Drug Science, University of Parma, Parma, Italy
| |
Collapse
|
6
|
Kumar H, Chakrabarti A, Sarma P, Modi M, Banerjee D, Radotra BD, Bhatia A, Medhi B. Novel therapeutic mechanism of action of metformin and its nanoformulation in Alzheimer's disease and role of AKT/ERK/GSK pathway. Eur J Pharm Sci 2023; 181:106348. [PMID: 36496166 DOI: 10.1016/j.ejps.2022.106348] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Brain Insulin-resistance plays a critical role in pathogenesis of Alzheimer's disease (AD). Current study explored the therapeutic mechanism of metformin (insulin sensitizer) and its solid-lipid nanoformulation (SLN) in rat-model of AD. In our study, SLN was prepared using microemulsion method. AD was induced with ICV-Aβ whereas the control-group (sham) received ICV-NS. Treatment arms included, disease-control (no treatment), Metformin (50 mg/kg, 100 mg/kg and 150 mg/kg), SLN-metformin 50 mg/kg and memantine 1.8 mg/kg (positive-control). Animals were tested for cognitive performance (EPM, MWM) after 21 days of therapy and sacrificed. Aβ (1-42), hyperphosphorylated tau, pAKTser473, GSK-3β, p-ERK (ELISA), metformin level(HPLC), neuronal injury score(H&E), Bcl2 and Bax(IHC) was evaluated in isolated brain. In our study, metformin-SLN were of spherical shape (size<200 nm) with 94.08% entrapment efficiency. Metformin was detectable in brain. Compared to sham, the disease-control group showed significantly higher (p ≤ 0.05) memory impairment(MWM and EPM), hyperphosphorylated tau, Aβ(1-42), neuronal-injury, Bax and lower Bcl-2 expression. Treatment with metformin and nanoformulation significantly reverse these parameters. AKT-ERK-GSK3β-Hyperphosphorylated tau pathway was found to be involved in the protective efficacy of metformin. To conclude, both metformin and its SLN were found to be effective as therapeutic agents in AD which act through the AKT-ERK-GSK3β-Hyperphosphorylated tau pathway. We need population based studies to confirm the same.
Collapse
Affiliation(s)
| | | | | | | | | | - B D Radotra
- Department of Histopathology, PGIMER, Chandigarh
| | - Alka Bhatia
- Department of Experimental Medicine & Biotechnology, PGIMER, Chandigarh.
| | | |
Collapse
|
7
|
Zha C, Sossin WS. The molecular diversity of plasticity mechanisms underlying memory: An evolutionary perspective. J Neurochem 2022; 163:444-460. [PMID: 36326567 DOI: 10.1111/jnc.15717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/29/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Experience triggers molecular cascades in organisms (learning) that lead to alterations (memory) to allow the organism to change its behavior based on experience. Understanding the molecular mechanisms underlying memory, particularly in the nervous system of animals, has been an exciting scientific challenge for neuroscience. We review what is known about forms of neuronal plasticity that underlie memory highlighting important issues in the field: (1) the importance of being able to measure how neurons are activated during learning to identify the form of plasticity that underlies memory, (2) the many distinct forms of plasticity important for memories that naturally decay both within and between organisms, and (3) unifying principles underlying the formation and maintenance of long-term memories. Overall, the diversity of molecular mechanisms underlying memories that naturally decay contrasts with more unified molecular mechanisms implicated in long-lasting changes. Despite many advances, important questions remain as to which mechanisms of neuronal plasticity underlie memory.
Collapse
Affiliation(s)
- Congyao Zha
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Wayne S Sossin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Parent MB. Using Postmeal Measures and Manipulations to Investigate Hippocampal Mnemonic Control of Eating Behavior. Neuroscience 2022; 497:228-238. [PMID: 34998891 PMCID: PMC9256844 DOI: 10.1016/j.neuroscience.2021.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022]
Abstract
Episodic meal-related memories provide the brain with a powerful mechanism for tracking and controlling eating behavior because they contain a detailed record of recent energy intake that likely outlasts the physiological signals generated by feeding bouts. This review briefly summarizes evidence from human participants showing that episodic meal-related memory limits later eating behavior and then describes our research aimed at investigating whether hippocampal neurons mediate the inhibitory effects of meal-related memory on subsequent feeding. Our approach has been inspired by pioneering work conducted by Ivan Izquierdo and others who used posttraining manipulations to investigate memory consolidation. This review describes the rationale and value of posttraining manipulations, how Izquierdo used them to demonstrate that dorsal hippocampal (dHC) neurons are critical for memory consolidation, and how we have adapted this strategy to investigate whether dHC neurons are necessary for mnemonic control of energy intake. I describe our evidence showing that ingestion activates the molecular processes necessary for synaptic plasticity and memory during the early postprandial period, when the memory of the meal would be undergoing consolidation, and then summarize our findings showing that neural activity in dHC neurons is critical during the early postprandial period for limiting future intake. Collectively, our evidence supports the hypothesis that dHC neurons mediate the inhibitory effects of ingestion-related memory on future intake and demonstrates that post-experience memory modulation is not confined to artificial laboratory memory tasks.
Collapse
Affiliation(s)
- M B Parent
- Neuroscience Institute & Department of Psychology, Georgia State University, PO Box 5030, Atlanta, GA 30303, USA.
| |
Collapse
|
9
|
Shim Y, Han HJ, Park KW, Kim BC, Park KH, Park MY, Kim HJ, Moon SY, Choi SH, Park KW, Yang DW, Yoon SJ, Kim SY, Youn YC, Choi HJ, Yoon KE, Cho HJ, Han SH. A Multicenter, Randomized, Double-Blind, Placebo-Controlled, Phase IIb Clinical Study to Evaluate the Safety and Efficacy of DHP1401 in Patients with Mild to Moderate Alzheimer's Disease Treated with Donepezil: DHP1401 Randomized Trial in Mild to Moderate Alzheimer's Disease (DRAMA). J Alzheimers Dis 2022; 87:391-403. [PMID: 35275529 DOI: 10.3233/jad-215277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Preclinical studies in transgenic models of Alzheimer's disease (AD) suggest that DHP1401 has neuroprotective and memory-enhancing effects. OBJECTIVE To evaluate the efficacy and safety of DHP1401 in AD patients treated with donepezilMethods:Methods: In a double-blind study, patients with mild-to-moderate AD were randomized (1:1:1) to receive a twice daily total dose of 500 mg or 1000 mg DHP1401 or placebo for 24 weeks. Tolerability and safety were monitored at baseline and weeks 12 and 24. RESULTS total of 180 patients were randomized to Active 1 (500 mg: n = 62), Active 2 (1000 mg: n = 53), and control groups (n = 65) in 16 sites in Korea. There was no significant difference in the Alzheimer's Disease Assessment Scale (ADAS-cog) score, the primary efficacy endpoint, from baseline. However, in the subgroup with mild AD patients (MMSE, 20-26) who received the high dose of DHP1401 and the group that received donepezil 5 mg, the ADAS-cog scores improved. MMSE and K-TMT-e type B were significant in both active groups at week 24. The most frequently observed symptom was dizziness (2.78%), and the most commonly observed reactions were related to metabolism and nutrition disorders (5.00%). No remarkable adverse events were observed for 24 weeks. CONCLUSION Although the effectiveness of DHP1401 was not proved to be superior as the primary efficacy endpoint, the secondary endpoints, MMSE and K-TMT-e type B, showed significant beneficial effects. Also, the subgroups showed that ADAS-cog scores significantly were improved. DHP1401 could be proven beneficial for the AD treatment by further clinical trials.
Collapse
Affiliation(s)
- YongSoo Shim
- Department of Neurology, The Catholic University of Korea Eunpyeong St. Mary's Hospital, Seoul, Republic of Korea
| | - Hyun Jeong Han
- Department of Neurology, Myongji Hospital, Hanyang University College of Medicine, Goyang, Republic of Korea
| | - Kyung Won Park
- Department of Neurology, Dong-A University College of Medicine and Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Kee Hyung Park
- Department of Neurology, College of Medicine, Gachon University, Gil Medical Center, Incheon, Republic of Korea
| | - Mee Young Park
- Department of Neurology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Hee-Jin Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - So Young Moon
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Kun Woo Park
- Department of Neurology, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Dong Won Yang
- Department of Neurology, The Catholic University of Korea, Seoul St. Mary's hospital, Seoul, Republic of Korea
| | - Soo Jin Yoon
- Department of Neurology, Daejeon Eulji Medical Center, Eulji University, Daejeon, Republic of Korea
| | - Sang Yun Kim
- Department of Neurology, Seoul National University Bundang Hospital & Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University, College of Medicine, Seoul, Republic of Korea
| | - Ho Jin Choi
- Department of Neurology, Hanyang University College of Medicine, Hanyang University Guri Hospital, Guri, Republic of Korea
| | - Koung Eun Yoon
- Clinical Trial Team, Daehwa Pharmaceutical Co., Ltd, Seoul, Republic of Korea
| | - Hyun Ju Cho
- Clinical Trial Team, Daehwa Pharmaceutical Co., Ltd, Seoul, Republic of Korea
| | - Seol-Heui Han
- Department of Neurology, Konkuk University Medical Center, Seoul, Republic of Korea
| |
Collapse
|
10
|
Calcium-/Calmodulin-Dependent Protein Kinase II (CaMKII) Inhibition Induces Learning and Memory Impairment and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:4635054. [PMID: 34976299 PMCID: PMC8718318 DOI: 10.1155/2021/4635054] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022]
Abstract
Objectives Inhibition of calcium-/calmodulin- (CaM-) dependent kinase II (CaMKII) is correlated with epilepsy. However, the specific mechanism that underlies learning and memory impairment and neuronal death by CaMKII inhibition remains unclear. Materials and Methods In this study, KN93, a CaMKII inhibitor, was used to investigate the role of CaMKII during epileptogenesis. We first identified differentially expressed genes (DEGs) in primary cultured hippocampal neurons with or without KN93 treatment using RNA-sequencing. Then, the impairment of learning and memory by KN93-induced CaMKII inhibition was assessed using the Morris water maze test. In addition, Western blotting, immunohistochemistry, and TUNEL staining were performed to determine neuronal death, apoptosis, and the relative signaling pathway. Results KN93-induced CaMKII inhibition decreased cAMP response element-binding (CREB) protein activity and impaired learning and memory in Wistar and tremor (TRM) rats, an animal model of genetic epilepsy. CaMKII inhibition also induced neuronal death and reactive astrocyte activation in both the Wistar and TRM hippocampi, deregulating mitogen-activated protein kinases. Meanwhile, neuronal death and neuron apoptosis were observed in PC12 and primary cultured hippocampal neurons after exposure to KN93, which was reversed by SP600125, an inhibitor of c-Jun N-terminal kinase (JNK). Conclusions CaMKII inhibition caused learning and memory impairment and apoptosis, which might be related to dysregulated JNK signaling.
Collapse
|
11
|
Parent MB, Higgs S, Cheke LG, Kanoski SE. Memory and eating: A bidirectional relationship implicated in obesity. Neurosci Biobehav Rev 2022; 132:110-129. [PMID: 34813827 PMCID: PMC8816841 DOI: 10.1016/j.neubiorev.2021.10.051] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/17/2021] [Accepted: 10/28/2021] [Indexed: 01/03/2023]
Abstract
This paper reviews evidence demonstrating a bidirectional relationship between memory and eating in humans and rodents. In humans, amnesia is associated with impaired processing of hunger and satiety cues, disrupted memory of recent meals, and overconsumption. In healthy participants, meal-related memory limits subsequent ingestive behavior and obesity is associated with impaired memory and disturbances in the hippocampus. Evidence from rodents suggests that dorsal hippocampal neural activity contributes to the ability of meal-related memory to control future intake, that endocrine and neuropeptide systems act in the ventral hippocampus to provide cues regarding energy status and regulate learned aspects of eating, and that consumption of hypercaloric diets and obesity disrupt these processes. Collectively, this evidence indicates that diet-induced obesity may be caused and/or maintained, at least in part, by a vicious cycle wherein excess intake disrupts hippocampal functioning, which further increases intake. This perspective may advance our understanding of how the brain controls eating, the neural mechanisms that contribute to eating-related disorders, and identify how to treat diet-induced obesity.
Collapse
Affiliation(s)
- Marise B Parent
- Neuroscience Institute & Department of Psychology, Georgia State University, Box 5030, Atlanta, GA 30303-5030, United States.
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, BI5 2TT, United Kingdom.
| | - Lucy G Cheke
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB, United Kingdom.
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, 90089-0371, United States.
| |
Collapse
|
12
|
van der Westhuizen ET, Choy KHC, Valant C, McKenzie-Nickson S, Bradley SJ, Tobin AB, Sexton PM, Christopoulos A. Fine Tuning Muscarinic Acetylcholine Receptor Signaling Through Allostery and Bias. Front Pharmacol 2021; 11:606656. [PMID: 33584282 PMCID: PMC7878563 DOI: 10.3389/fphar.2020.606656] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
The M1 and M4 muscarinic acetylcholine receptors (mAChRs) are highly pursued drug targets for neurological diseases, in particular for Alzheimer's disease and schizophrenia. Due to high sequence homology, selective targeting of any of the M1-M5 mAChRs through the endogenous ligand binding site has been notoriously difficult to achieve. With the discovery of highly subtype selective mAChR positive allosteric modulators in the new millennium, selectivity through targeting an allosteric binding site has opened new avenues for drug discovery programs. However, some hurdles remain to be overcome for these promising new drug candidates to progress into the clinic. One challenge is the potential for on-target side effects, such as for the M1 mAChR where over-activation of the receptor by orthosteric or allosteric ligands can be detrimental. Therefore, in addition to receptor subtype selectivity, a drug candidate may need to exhibit a biased signaling profile to avoid such on-target adverse effects. Indeed, recent studies in mice suggest that allosteric modulators for the M1 mAChR that bias signaling toward specific pathways may be therapeutically important. This review brings together details on the signaling pathways activated by the M1 and M4 mAChRs, evidence of biased agonism at these receptors, and highlights pathways that may be important for developing new subtype selective allosteric ligands to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Emma T. van der Westhuizen
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - K. H. Christopher Choy
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Simon McKenzie-Nickson
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Sophie J. Bradley
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Andrew B. Tobin
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Patrick M. Sexton
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| |
Collapse
|
13
|
Sleep Deprivation and Neurological Disorders. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5764017. [PMID: 33381558 PMCID: PMC7755475 DOI: 10.1155/2020/5764017] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
Sleep plays an important role in maintaining neuronal circuitry, signalling and helps maintain overall health and wellbeing. Sleep deprivation (SD) disturbs the circadian physiology and exerts a negative impact on brain and behavioural functions. SD impairs the cellular clearance of misfolded neurotoxin proteins like α-synuclein, amyloid-β, and tau which are involved in major neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. In addition, SD is also shown to affect the glymphatic system, a glial-dependent metabolic waste clearance pathway, causing accumulation of misfolded faulty proteins in synaptic compartments resulting in cognitive decline. Also, SD affects the immunological and redox system resulting in neuroinflammation and oxidative stress. Hence, it is important to understand the molecular and biochemical alterations that are the causative factors leading to these pathophysiological effects on the neuronal system. This review is an attempt in this direction. It provides up-to-date information on the alterations in the key processes, pathways, and proteins that are negatively affected by SD and become reasons for neurological disorders over a prolonged period of time, if left unattended.
Collapse
|
14
|
Swenson S, Blum K, McLaughlin T, Gold MS, Thanos PK. The therapeutic potential of exercise for neuropsychiatric diseases: A review. J Neurol Sci 2020; 412:116763. [PMID: 32305746 DOI: 10.1016/j.jns.2020.116763] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/14/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023]
Abstract
Exercise is known to have a myriad of health benefits. There is much to be learned from the effects of exercise and its potential for prevention, attenuation and treatment of multiple neuropsychiatric diseases and behavioral disorders. Furthermore, recent data and research on exercise benefits with respect to major health crises, such as, that of opioid and general substance use disorders, make it very important to better understand and review the mechanisms of exercise and how it could be utilized for effective treatments or adjunct treatments for these diseases. In addition, mechanisms, epigenetics and sex differences are examined and discussed in terms of future research implications.
Collapse
Affiliation(s)
- Sabrina Swenson
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Kenneth Blum
- Western Univesity Health Sciences, Graduate College, Pomona, CA, USA
| | | | - Mark S Gold
- Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY, USA; Department of Psychology, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
15
|
Kir6.1 Heterozygous Mice Exhibit Aberrant Amygdala-Dependent Cued Fear Memory. Mol Neurobiol 2019; 57:1622-1635. [PMID: 31808063 DOI: 10.1007/s12035-019-01840-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 11/18/2019] [Indexed: 10/25/2022]
Abstract
ATP-sensitive K+ (KATP) channels are predominantly expressed in the brain and consist of four identical inward-rectifier potassium ion channel subunits (Kir6.1 or Kir6.2) and four identical high-affinity sulfonylurea receptor subunits (SUR1, SUR2A, or SUR2B). We previously observed that chronic corticosterone-treated (CORT) mice exhibited enhanced anxiety-like behaviors and cued fear memory. In the present study, the protein and mRNA expression levels of Kir6.1, but not Kir6.2, were decreased in the lateral amygdala (LA) of CORT mice. Heterozygous Kir6.1-null (Kir6.1+/-) mice also showed enhanced tone (cued) fear memory and long-term potentiation (LTP) in the cortico-LA pathway compared to those in wild-type mice. However, LTP was not enhanced in the hippocampal CA1 regions of Kir6.1+/- mice. Consistent with increased cued fear memory, both Ca2+/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) activities were significantly elevated in the LAs of Kir6.1+/- mice after tone stimulation. Our results indicate that increased CaMKII and ERK activities may induce LTP in the LA in Kir6.1+/- mice, leading to aberrant cued fear memory. The changes in neural plasticity in the LA of Kir6.1+/- mice were associated with anxiety-like behaviors and may be related to the pathogenic mechanisms of anxiety disorders in human patients.
Collapse
|
16
|
Evans PD. Rapid signalling responses via the G protein-coupled estrogen receptor, GPER, in a hippocampal cell line. Steroids 2019; 152:108487. [PMID: 31499073 DOI: 10.1016/j.steroids.2019.108487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 01/14/2023]
Abstract
The rapid non-genomic actions of 17β-estradiol in multiple tissues, including the nervous system, may involve the activation of the G-protein-coupled receptor, GPER. Different signalling pathways have been suggested to be activated by GPER in different cell lines and tissues. Controversially, GPER has also been suggested to be activated by the mineralocorticoid aldosterone, and by the non-steroidal diphenylacrylamide compound, STX, in some preparations. Evidence for the ability of the GPER agonist, G-1, and for aldosterone in the presence of the mineralocorticoid receptor antagonist, eplerenone, to potentiate forskolin-stimulated cyclic AMP levels in the hippocampal clonal cell line, mHippoE-18 is reviewed. The effects of both agents are blocked by the GPER antagonist G36, by PTX, (suggesting the involvement of Gi/o G proteins), by BAPTA-AM, (suggesting they are calcium sensitive), by wortmannin (suggesting an involvement of PI3Kinase) and by soluble amyloid-β peptides. STX also stimulates cyclic AMP levels in mHippoE-18 cells and these effects are blocked by G36 and PTX, as well as by amyloid-β peptides. This suggests that both aldosterone and STX may be capable of activating GPER in mHippoE-18 cells. Possible molecular mechanisms that may underlie these effects are discussed, together with possible forward directions for research on rapid non-genomic signalling by GPER, emphasising the importance of understanding the spatio-temporal aspects of its signalling in various tissues.
Collapse
Affiliation(s)
- Peter D Evans
- The Signalling Laboratory, The Babraham Institute, The Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
17
|
Evans PD. Aldosterone, STX and amyloid-β 1-42 peptides modulate GPER (GPR30) signalling in an embryonic mouse hippocampal cell line (mHippoE-18). Mol Cell Endocrinol 2019; 496:110537. [PMID: 31404576 DOI: 10.1016/j.mce.2019.110537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 01/02/2023]
Abstract
The GPCR, GPER, mediates many of the rapid, non-genomic actions of 17β-estradiol in multiple tissues, including the nervous system. Controversially, it has also been suggested to be activated by aldosterone, and by the non-steroidal diphenylacrylamide compound, STX, in some preparations. Here, the ability of the GPER agonist, G-1, and aldosterone in the presence of the mineralocorticoid receptor antagonist, eplerenone, to potentiate forskolin-stimulated cyclic AMP levels in the hippocampal clonal cell line, mHippoE-18, are compared. Both stimulatory effects are blocked by the GPER antagonist G36, by PTX, (suggesting the involvement of Gi/o G proteins), by BAPTA-AM, (suggesting they are calcium sensitive), by wortmannin (suggesting an involvement of PI3Kinase) and by soluble amyloid-β peptides. STX also stimulates cyclic AMP levels in mHippoE-18 cells and these effects are blocked by G36 and PTX, as well as by amyloid-β peptides. This suggests that both aldosterone and STX may modulate GPER signalling in mHippoE-18 cells.
Collapse
Affiliation(s)
- Peter D Evans
- The Signalling Laboratory, The Babraham Institute, The Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
18
|
Duda P, Wójcicka O, Wiśniewski JR, Rakus D. Global quantitative TPA-based proteomics of mouse brain structures reveals significant alterations in expression of proteins involved in neuronal plasticity during aging. Aging (Albany NY) 2019; 10:1682-1697. [PMID: 30026405 PMCID: PMC6075443 DOI: 10.18632/aging.101501] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/15/2018] [Indexed: 01/17/2023]
Abstract
Aging is believed to be the result of alterations of protein expression and accumulation of changes in biomolecules. Although there are numerous reports demonstrating changes in protein expression in brain during aging, only few of them describe global changes at the protein level. Here, we present the deepest quantitative proteomic analysis of three brain regions, hippocampus, cortex and cerebellum, in mice aged 1 or 12 months, using the total protein approach technique. In all the brain regions, both in young and middle-aged animals, we quantitatively measured over 5,200 proteins. We found that although the total protein expression in middle-aged brain structures is practically unaffected by aging, there are significant differences between young and middle-aged mice in the expression of some receptors and signaling cascade proteins proven to be significant for learning and memory formation. Our analysis demonstrates that the hippocampus is the most variable structure during natural aging and that the first symptoms of weakening of neuronal plasticity may be observed on protein level in middle-aged animals.
Collapse
Affiliation(s)
- Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw 50-137, Poland
| | - Olga Wójcicka
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw 50-137, Poland
| | - Jacek R Wiśniewski
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried 82152, Germany
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw 50-137, Poland
| |
Collapse
|
19
|
Tyebji S, Seizova S, Garnham AL, Hannan AJ, Tonkin CJ. Impaired social behaviour and molecular mediators of associated neural circuits during chronic Toxoplasma gondii infection in female mice. Brain Behav Immun 2019; 80:88-108. [PMID: 30807837 DOI: 10.1016/j.bbi.2019.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 02/22/2019] [Indexed: 12/24/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a neurotropic parasite that is associated with various neuropsychiatric disorders. Rodents infected with T. gondii display a plethora of behavioural alterations, and Toxoplasma infection in humans has been strongly associated with disorders such as schizophrenia, in which impaired social behaviour is an important feature. Elucidating changes at the cellular level relevant to neuropsychiatric conditions can lead to effective therapies. Here, we compare changes in behaviour during an acute and chronic T. gondii infection in female mice. Further, we notice that during chronic phase of infection, mice display impaired sociability when exposed to a novel conspecific. Also, we show that T. gondii infected mice display impaired short-term social recognition memory. However, object recognition memory remains intact. Using c-Fos as a marker of neuronal activity, we show that infection leads to an impairment in neuronal activation in the medial prefrontal cortex, hippocampus as well as the amygdala when mice are exposed to a social environment and a change in functional connectivity between these regions. We found changes in synaptic proteins that play a role in the process of neuronal activation such as synaptophysin, PSD-95 and changes in downstream substrates of cell activity such as cyclic AMP, phospho-CREB and BDNF. Our results point towards an imbalance in neuronal activity that can lead to a wider range of neuropsychiatric problems upon T. gondii infection.
Collapse
Affiliation(s)
- Shiraz Tyebji
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria, Australia.
| | - Simona Seizova
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia.
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville 3052, Victoria, Australia.
| | - Christopher J Tonkin
- The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne 3052, Australia.
| |
Collapse
|
20
|
Naewla S, Sirichoat A, Pannangrong W, Chaisawang P, Wigmore P, Welbat JU. Hesperidin Alleviates Methotrexate-Induced Memory Deficits via Hippocampal Neurogenesis in Adult Rats. Nutrients 2019; 11:nu11040936. [PMID: 31027240 PMCID: PMC6521088 DOI: 10.3390/nu11040936] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 01/25/2023] Open
Abstract
Methotrexate (MTX), a folic acid antagonist, is widely used in cancer treatment. However, treatment with MTX reduces hippocampal neurogenesis, leading to memory deficits. Hesperidin (Hsd) is a flavonoid glycoside that promotes anti-inflammation, acts as an antioxidant, and has neuroprotective properties. Consumption of Hsd enhances learning and memory. In the present study, we investigated the protective effects of Hsd against MTX-induced impairments of memory and neurogenesis; male Sprague Dawley rats were administered with a single dose of MTX (75 mg/kg) by intravenous (i.v.) injection on days 8 and 15 or Hsd (100 mg/kg) by oral gavage for 21 days. Memory was tested using novel object location (NOL) and novel object recognition (NOR) tasks. Immunofluorescence staining of Ki-67, bromodeoxyuridine (BrdU), and doublecortin (DCX) was performed to assess cell proliferation, survival, and immature neurons. The data showed that Hsd and MTX did not disable locomotor ability. The MTX animals exhibited memory deficits in both memory tests. There were significant decreases in the numbers of cell proliferation, survival, and immature neurons in the MTX animals. However, co-administration with MTX and Hsd alleviated memory loss and neurogenesis decline. These results revealed that Hsd could protect against MTX side effects in the animals in this study.
Collapse
Affiliation(s)
- Salinee Naewla
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Apiwat Sirichoat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Wanassanan Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Pornthip Chaisawang
- Faculty of Medical Science, Nakhonratchasima College, Nakhon Ratchasima 30000, Thailand.
| | - Peter Wigmore
- School of Life Sciences, Medical School, Queen's Medical Centre, Nottingham University, Nottingham NG7 2RD, UK.
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
- Neuroscience Research and Development Group, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
21
|
Ross A, Barnett N, Faulkner A, Hannapel R, Parent MB. Sucrose ingestion induces glutamate AMPA receptor phosphorylation in dorsal hippocampal neurons: Increased sucrose experience prevents this effect. Behav Brain Res 2019; 359:792-798. [PMID: 30076854 PMCID: PMC6594687 DOI: 10.1016/j.bbr.2018.07.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/19/2018] [Accepted: 07/31/2018] [Indexed: 11/26/2022]
Abstract
Evidence suggests that meal-related memory influences later eating behavior. Memory can serve as a powerful mechanism for controlling eating behavior because it provides a record of recent intake that likely outlasts most physiological signals generated by ingestion. Dorsal (dHC) and ventral hippocampal (vHC) neurons are critical for memory, and we demonstrated previously that they limit energy intake during the postprandial period. If dHC or vHC neurons control intake through a process that requires memory, then ingestion should increase events necessary for synaptic plasticity in dHC and vHC during the postprandial period. To test this, we determined whether ingesting a sucrose solution induced posttranslational events critical for hippocampal synaptic plasticity: phosphorylation of AMPAR GluA1 subunits at 1) serine 831 (pSer831) and 2) serine 845 (pSer845). We also examined whether increasing the amount of previous experience with the sucrose solution, which would be expected to decrease the mnemonic demand involved in an ingestion bout, would also attenuate sucrose-induced phosphorylation. Quantitative immunoblotting of dHC and vHC membrane fractions demonstrated that sucrose ingestion increased postprandial pSer831 in dHC but not vHC. Increased previous sucrose experience prevented sucrose-induced dHC pSer831. Sucrose ingestion did not affect pSer845 in either dHC or vHC. Thus, the present findings show that ingestion activates a postranslational event necessary for synaptic plasticity in an experience-dependent manner, which is consistent with the hypothesis that dHC neurons form a memory of a meal during the postprandial period.
Collapse
Affiliation(s)
- Amy Ross
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA, 30302, United States
| | - Nicolette Barnett
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA, 30302, United States
| | - Alexa Faulkner
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA, 30302, United States
| | - Reilly Hannapel
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA, 30302, United States
| | - Marise B Parent
- Neuroscience Institute, Georgia State University, P.O. Box 5030, Atlanta, GA, 30302, United States; Department of Psychology, Georgia State University, P.O. Box 5030, Atlanta, GA, 30302, United States.
| |
Collapse
|
22
|
Effect of Exercise and Aβ Protein Infusion on Long-Term Memory-Related Signaling Molecules in Hippocampal Areas. Mol Neurobiol 2018; 56:4980-4987. [DOI: 10.1007/s12035-018-1425-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022]
|
23
|
Lee JH, Jahrling JB, Denner L, Dineley KT. Targeting Insulin for Alzheimer’s Disease: Mechanisms, Status and Potential Directions. J Alzheimers Dis 2018; 64:S427-S453. [DOI: 10.3233/jad-179923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jung Hyun Lee
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jordan B. Jahrling
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Larry Denner
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Kelly T. Dineley
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
24
|
Circadian Regulation of Hippocampal-Dependent Memory: Circuits, Synapses, and Molecular Mechanisms. Neural Plast 2018; 2018:7292540. [PMID: 29593785 PMCID: PMC5822921 DOI: 10.1155/2018/7292540] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Circadian modulation of learning and memory efficiency is an evolutionarily conserved phenomenon, occurring in organisms ranging from invertebrates to higher mammalian species, including humans. While the suprachiasmatic nucleus (SCN) of the hypothalamus functions as the master mammalian pacemaker, recent evidence suggests that forebrain regions, including the hippocampus, exhibit oscillatory capacity. This finding, as well as work on the cellular signaling events that underlie learning and memory, has opened promising new avenues of investigation into the precise cellular, molecular, and circuit-based mechanisms by which clock timing impacts plasticity and cognition. In this review, we examine the complex molecular relationship between clock timing and memory, with a focus on hippocampal-dependent tasks. We evaluate how the dysregulation of circadian timing, both at the level of the SCN and at the level of ancillary forebrain clocks, affects learning and memory. Further, we discuss experimentally validated intracellular signaling pathways (e.g., ERK/MAPK and GSK3β) and potential cellular signaling mechanisms by which the clock affects learning and memory formation. Finally, we examine how long-term potentiation (LTP), a synaptic process critical to the establishment of several forms of memory, is regulated by clock-gated processes.
Collapse
|
25
|
Zhang MQ, Li R, Wang YQ, Huang ZL. Neural Plasticity Is Involved in Physiological Sleep, Depressive Sleep Disturbances, and Antidepressant Treatments. Neural Plast 2017; 2017:5870735. [PMID: 29181202 PMCID: PMC5664320 DOI: 10.1155/2017/5870735] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/27/2017] [Accepted: 07/13/2017] [Indexed: 12/28/2022] Open
Abstract
Depression, which is characterized by a pervasive and persistent low mood and anhedonia, greatly impacts patients, their families, and society. The associated and recurring sleep disturbances further reduce patient's quality of life. However, therapeutic sleep deprivation has been regarded as a rapid and robust antidepressant treatment for several decades, which suggests a complicated role of sleep in development of depression. Changes in neural plasticity are observed during physiological sleep, therapeutic sleep deprivation, and depression. This correlation might help us to understand better the mechanism underlying development of depression and the role of sleep. In this review, we first introduce the structure of sleep and the facilitated neural plasticity caused by physiological sleep. Then, we introduce sleep disturbances and changes in plasticity in patients with depression. Finally, the effects and mechanisms of antidepressants and therapeutic sleep deprivation on neural plasticity are discussed.
Collapse
Affiliation(s)
- Meng-Qi Zhang
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Rui Li
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yi-Qun Wang
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Zhi-Li Huang
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
26
|
Fernandes J, Arida RM, Gomez-Pinilla F. Physical exercise as an epigenetic modulator of brain plasticity and cognition. Neurosci Biobehav Rev 2017; 80:443-456. [PMID: 28666827 DOI: 10.1016/j.neubiorev.2017.06.012] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/18/2017] [Accepted: 06/26/2017] [Indexed: 02/08/2023]
Abstract
A large amount of evidence has demonstrated the power of exercise to support cognitive function, the effects of which can last for considerable time. An emerging line of scientific evidence indicates that the effects of exercise are longer lasting than previously thought up to the point to affect future generations. The action of exercise on epigenetic regulation of gene expression seem central to building an "epigenetic memory" to influence long-term brain function and behavior. In this review article, we discuss new developments in the epigenetic field connecting exercise with changes in cognitive function, including DNA methylation, histone modifications and microRNAs (miRNAs). The understanding of how exercise promotes long-term cognitive effects is crucial for directing the power of exercise to reduce the burden of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Jansen Fernandes
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Department of Physiology-Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Ricardo Mario Arida
- Department of Physiology-Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
27
|
Emery AC, Xu W, Eiden MV, Eiden LE. Guanine nucleotide exchange factor Epac2-dependent activation of the GTP-binding protein Rap2A mediates cAMP-dependent growth arrest in neuroendocrine cells. J Biol Chem 2017; 292:12220-12231. [PMID: 28546426 DOI: 10.1074/jbc.m117.790329] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/23/2017] [Indexed: 11/06/2022] Open
Abstract
First messenger-dependent activation of MAP kinases in neuronal and endocrine cells is critical for cell differentiation and function and requires guanine nucleotide exchange factor (GEF)-mediated activation of downstream Ras family small GTPases, which ultimately lead to ERK, JNK, and p38 phosphorylation. Because there are numerous GEFs and also a host of Ras family small GTPases, it is important to know which specific GEF-small GTPase dyad functions in a given cellular process. Here we investigated the upstream activators and downstream effectors of signaling via the GEF Epac2 in the neuroendocrine NS-1 cell line. Three cAMP sensors, Epac2, PKA, and neuritogenic cAMP sensor-Rapgef2, mediate distinct cellular outputs: p38-dependent growth arrest, cAMP response element-binding protein-dependent cell survival, and ERK-dependent neuritogenesis, respectively, in these cells. Previously, we found that cAMP-induced growth arrest of PC12 and NS-1 cells requires Epac2-dependent activation of p38 MAP kinase, which posed the important question of how Epac2 engages p38 without simultaneously activating other MAP kinases in neuronal and endocrine cells. We now show that the small GTP-binding protein Rap2A is the obligate effector for, and GEF substrate of, Epac2 in mediating growth arrest through p38 activation in NS-1 cells. This new pathway is distinctly parcellated from the G protein-coupled receptor → Gs → adenylate cyclase → cAMP → PKA → cAMP response element-binding protein pathway mediating cell survival and the G protein-coupled receptor → Gs → adenylate cyclase → cAMP → neuritogenic cAMP sensor-Rapgef2 → B-Raf → MEK → ERK pathway mediating neuritogenesis in NS-1 cells.
Collapse
Affiliation(s)
- Andrew C Emery
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892
| | - Wenqin Xu
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892
| | - Maribeth V Eiden
- Office of the Scientific Director, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland 20892.
| |
Collapse
|
28
|
Dessauer CW, Watts VJ, Ostrom RS, Conti M, Dove S, Seifert R. International Union of Basic and Clinical Pharmacology. CI. Structures and Small Molecule Modulators of Mammalian Adenylyl Cyclases. Pharmacol Rev 2017; 69:93-139. [PMID: 28255005 PMCID: PMC5394921 DOI: 10.1124/pr.116.013078] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adenylyl cyclases (ACs) generate the second messenger cAMP from ATP. Mammalian cells express nine transmembrane AC (mAC) isoforms (AC1-9) and a soluble AC (sAC, also referred to as AC10). This review will largely focus on mACs. mACs are activated by the G-protein Gαs and regulated by multiple mechanisms. mACs are differentially expressed in tissues and regulate numerous and diverse cell functions. mACs localize in distinct membrane compartments and form signaling complexes. sAC is activated by bicarbonate with physiologic roles first described in testis. Crystal structures of the catalytic core of a hybrid mAC and sAC are available. These structures provide detailed insights into the catalytic mechanism and constitute the basis for the development of isoform-selective activators and inhibitors. Although potent competitive and noncompetitive mAC inhibitors are available, it is challenging to obtain compounds with high isoform selectivity due to the conservation of the catalytic core. Accordingly, caution must be exerted with the interpretation of intact-cell studies. The development of isoform-selective activators, the plant diterpene forskolin being the starting compound, has been equally challenging. There is no known endogenous ligand for the forskolin binding site. Recently, development of selective sAC inhibitors was reported. An emerging field is the association of AC gene polymorphisms with human diseases. For example, mutations in the AC5 gene (ADCY5) cause hyperkinetic extrapyramidal motor disorders. Overall, in contrast to the guanylyl cyclase field, our understanding of the (patho)physiology of AC isoforms and the development of clinically useful drugs targeting ACs is still in its infancy.
Collapse
Affiliation(s)
- Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Val J Watts
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Rennolds S Ostrom
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Marco Conti
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Stefan Dove
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Roland Seifert
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| |
Collapse
|
29
|
Jo SY, Jung IH, Yi JH, Choi TJ, Lee S, Jung JW, Yun J, Lee YC, Ryu JH, Kim DH. Ethanol extract of the seed of Zizyphus jujuba var. spinosa potentiates hippocampal synaptic transmission through mitogen-activated protein kinase, adenylyl cyclase, and protein kinase A pathways. JOURNAL OF ETHNOPHARMACOLOGY 2017; 200:16-21. [PMID: 28167293 DOI: 10.1016/j.jep.2017.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 01/20/2017] [Accepted: 02/03/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As the seed of Zizyphus jujuba var. spinosa (Bunge) Hu ex H.F. Chow (Rhamnaceae) has been used to sleep disturbances in traditional Chinese and Korean medicine, many previous studies have focused on its sedative effect. AIM OF THE STUDY Recently, we reported the neuroprotective effect of the effect of Z. jujuba var. spinosa. However, its effects on synaptic function have not yet been studied. In this project, we examined the action of ethanol extract of the seed of Z. jujuba var. spinosa (DHP1401) on synaptic transmission in the hippocampus. MATERIALS AND METHODS To investigate the effects of DHP1401, field recordings were conducted using hippocampal slices (400µm). Object recognition test was introduced to examine whether DHP1401 affect normal recognition memory. RESULTS DHP1401 (50μg/ml) induced a significant increase in synaptic activity in Shaffer collateral pathway in a concentration-dependent manner. This increase of synaptic responses was blocked by NBQX, a broad spectrum α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor antagonist, but not IEM-1460, a Ca2+-permeable AMPAR blocker. Moreover, U0126, a mitogen-activated protein kinase inhibitor, SQ22536, an adenylyl cyclase inhibitor, and PKI, a protein kinase A inhibitor, blocked DHP1401-induced increase in synaptic transmission. Finally, DHP1401 facilitated object recognition memory. CONCLUSIONS These results suggest that DHP1401 increase synaptic transmission through increase of synaptic AMPAR transmission via MAPK, AC and PAK.
Collapse
Affiliation(s)
- So Yeon Jo
- Department of Biotechnology, College of Natural Resources and Life Science, Dong-A University, Busan 49315, Republic of Korea.
| | - In Ho Jung
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoeki-dong, Dongdaemoon-Ku, Seoul 02447, Republic of Korea; Daehwa Pharmaceutical Co., Ltd., Seongnam 13488, Republic of Korea.
| | - Jee Hyun Yi
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, UK.
| | - Tae Joon Choi
- Daehwa Pharmaceutical Co., Ltd., Seongnam 13488, Republic of Korea.
| | - Seungheon Lee
- Department of Aquatic Biomedical Sciences, School of Marine Biomedical Science, College of Ocean Science, Jeju National University, Jeju 63243, Republic of Korea.
| | - Ji Wook Jung
- Department of Herbal Medicinal Pharmacology, College of Herbal Bio-industry, Daegu Haany University, Kyungsan 38610, Republic of Korea.
| | - Jeanho Yun
- Department of Biochemistry, College of Medicine, Dong-A University, Busan 607-714, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| | - Young Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoeki-dong, Dongdaemoon-Ku, Seoul 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| |
Collapse
|
30
|
Kreutzmann JC, Tudor JC, Angelakos CC, Abel T. The Impact of Sleep Deprivation on Molecular Mechanisms of Memory Consolidation in Rodents. COGNITIVE NEUROSCIENCE OF MEMORY CONSOLIDATION 2017. [DOI: 10.1007/978-3-319-45066-7_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
31
|
Different effects of prenatal stress on ERK2/CREB/Bcl-2 expression in the hippocampus and the prefrontal cortex of adult offspring rats. Neuroreport 2016; 27:600-4. [DOI: 10.1097/wnr.0000000000000581] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
32
|
Létondor A, Buaud B, Vaysse C, Richard E, Layé S, Pallet V, Alfos S. EPA/DHA and Vitamin A Supplementation Improves Spatial Memory and Alleviates the Age-related Decrease in Hippocampal RXRγ and Kinase Expression in Rats. Front Aging Neurosci 2016; 8:103. [PMID: 27242514 PMCID: PMC4860397 DOI: 10.3389/fnagi.2016.00103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/22/2016] [Indexed: 12/16/2022] Open
Abstract
Studies suggest that eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and vitamin A are critical to delay aged-related cognitive decline. These nutrients regulate gene expression in the brain by binding to nuclear receptors such as the retinoid X receptors (RXRs) and the retinoic acid receptors (RARs). Moreover, EPA/DHA and retinoids activate notably kinase signaling pathways such as AKT or MAPK, which includes ERK1/2. This suggests that these nutrients may modulate brain function in a similar way. Therefore, we investigated in middle-aged rats the behavioral and molecular effects of supplementations with EPA/DHA and vitamin A alone or combined. 18-month-old rats exhibited reference and working memory deficits in the Morris water maze, associated with a decrease in serum vitamin A and hippocampal EPA/DHA contents. RARα, RXRβ, and RXRγ mRNA expression and CAMKII, AKT, ERK1/2 expression were decreased in the hippocampus of middle-aged rats. A combined EPA/DHA and vitamin A supplementation had a beneficial additive effect on reference memory but not in working memory in middle-aged rats, associated with an alleviation of the age-related decrease in RXRγ, CAMKII, AKT, and ERK1 expression in the hippocampus. This study provides a new combined nutritional strategy to delay brain aging.
Collapse
Affiliation(s)
- Anne Létondor
- Université de Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; INRA, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; Bordeaux INP, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; ITERG, Institut des Corps GrasPessac, France
| | | | | | - Emmanuel Richard
- INSERM, Biothérapie des Maladies Génétiques Inflammatoires et Cancers, U1035 Bordeaux, France
| | - Sophie Layé
- Université de Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; INRA, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France
| | - Véronique Pallet
- Université de Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; INRA, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; Bordeaux INP, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France
| | - Serge Alfos
- Université de Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; INRA, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France; Bordeaux INP, Nutrition et Neurobiologie Intégrée, UMR 1286Bordeaux, France
| |
Collapse
|
33
|
Horiguchi M, Miyauchi M, Neugebauer NM, Oyamada Y, Meltzer HY. Prolonged reversal of the phencyclidine-induced impairment in novel object recognition by a serotonin (5-HT)1A-dependent mechanism. Behav Brain Res 2016; 301:132-41. [DOI: 10.1016/j.bbr.2015.08.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/27/2015] [Accepted: 08/30/2015] [Indexed: 02/08/2023]
|
34
|
Li Y, Shang J, Jiang Z, Zhang L, Su X. Regulation mechanism of peptides derived from sea cucumber ( Apostichopus japonicas) for modulation of learning and memory. Food Sci Biotechnol 2016; 25:241-246. [PMID: 30263263 DOI: 10.1007/s10068-016-0035-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 06/08/2015] [Accepted: 09/14/2015] [Indexed: 10/22/2022] Open
Abstract
Peptides derived from the sea cucumber Apostichopus japonicas have immunomodulatory and antioxidant activities. Three dosage levels of peptide hydrolysates were fed to Institute of Cancer Research (ICR) mice. Standardized Y and Morris water mazes were used to determine the effects of A. japonicas peptides on learning and memory. All 3 mouse test groups showed reductions in the latency period (LP), the number of times that a mouse swam over the location of a platform (NCP), the ratio of the distance from a target quadrant to the total swimming distance (DTQ), and the time spent in the target quadrant (TTQ). Microarray analysis was then carried out and 308 genes were differentially expressed between groups, of which 235 were up and 73 were down regulated in the brains of test group animals, compared with control group animals. Expressions of genes identified in microarrays were then analyzed using qRT-PCR.
Collapse
Affiliation(s)
- Yanyan Li
- 1Jiangsu Center for Drug Screening, Jiangsu Center for Pharmacodynamics Research and Evaluation, Key Laboratory of Drug Quality Control and Pharmacovigilance of the Ministry of Education, China Pharmaceutical University, Nanjing, 210009 China
| | - Jing Shang
- 1Jiangsu Center for Drug Screening, Jiangsu Center for Pharmacodynamics Research and Evaluation, Key Laboratory of Drug Quality Control and Pharmacovigilance of the Ministry of Education, China Pharmaceutical University, Nanjing, 210009 China
| | - Zhenzhou Jiang
- 1Jiangsu Center for Drug Screening, Jiangsu Center for Pharmacodynamics Research and Evaluation, Key Laboratory of Drug Quality Control and Pharmacovigilance of the Ministry of Education, China Pharmaceutical University, Nanjing, 210009 China
| | - Luyong Zhang
- 1Jiangsu Center for Drug Screening, Jiangsu Center for Pharmacodynamics Research and Evaluation, Key Laboratory of Drug Quality Control and Pharmacovigilance of the Ministry of Education, China Pharmaceutical University, Nanjing, 210009 China
| | - Xiurong Su
- 2School of Marine Science, Ningbo University, Ningbo, 315211 China
| |
Collapse
|
35
|
Liu Y, Wang Y, Zhu G, Sun J, Bi X, Baudry M. A calpain-2 selective inhibitor enhances learning & memory by prolonging ERK activation. Neuropharmacology 2016; 105:471-477. [PMID: 26907807 DOI: 10.1016/j.neuropharm.2016.02.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/18/2016] [Accepted: 02/15/2016] [Indexed: 02/04/2023]
Abstract
While calpain-1 activation is required for LTP induction by theta burst stimulation (TBS), calpain-2 activation limits its magnitude during the consolidation period. A selective calpain-2 inhibitor applied either before or shortly after TBS enhanced the degree of potentiation. In the present study, we tested whether the selective calpain-2 inhibitor, Z-Leu-Abu-CONH-CH2-C6H3 (3, 5-(OMe)2 (C2I), could enhance learning and memory in wild-type (WT) and calpain-1 knock-out (C1KO) mice. We first showed that C2I could reestablish TBS-LTP in hippocampal slices from C1KO mice, and this effect was blocked by PD98059, an inhibitor of ERK. TBS resulted in PTEN degradation in hippocampal slices from both WT and C1KO mice, and C2I treatment blocked this effect in both mouse genotypes. Systemic injection of C2I 30 min before training in the fear-conditioning paradigm resulted in a biphasic dose-response curve, with low doses enhancing and high doses inhibiting freezing behavior. The difference between the doses needed to enhance and inhibit learning matches the difference in concentrations producing inhibition of calpain-2 and calpain-1. A low dose of C2I also restored normal learning in a novel object recognition task in C1KO mice. Levels of SCOP, a ERK phosphatase known to be cleaved by calpain-1, were decreased in dorsal hippocampus early but not late following training in WT mice; C2I treatment did not affect the early decrease in SCOP levels but prevented its recovery at the later time-point and prolonged ERK activation. The results indicate that calpain-2 activation limits the extent of learning, an effect possibly due to temporal limitation of ERK activation, as a result of SCOP synthesis induced by calpain-2-mediated PTEN degradation.
Collapse
Affiliation(s)
- Yan Liu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Yubin Wang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Guoqi Zhu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA.
| |
Collapse
|
36
|
Adult Hippocampal Neurogenesis, Fear Generalization, and Stress. Neuropsychopharmacology 2016; 41:24-44. [PMID: 26068726 PMCID: PMC4677119 DOI: 10.1038/npp.2015.167] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/29/2015] [Accepted: 06/05/2015] [Indexed: 12/21/2022]
Abstract
The generalization of fear is an adaptive, behavioral, and physiological response to the likelihood of threat in the environment. In contrast, the overgeneralization of fear, a cardinal feature of posttraumatic stress disorder (PTSD), manifests as inappropriate, uncontrollable expression of fear in neutral and safe environments. Overgeneralization of fear stems from impaired discrimination of safe from aversive environments or discernment of unlikely threats from those that are highly probable. In addition, the time-dependent erosion of episodic details of traumatic memories might contribute to their generalization. Understanding the neural mechanisms underlying the overgeneralization of fear will guide development of novel therapeutic strategies to combat PTSD. Here, we conceptualize generalization of fear in terms of resolution of interference between similar memories. We propose a role for a fundamental encoding mechanism, pattern separation, in the dentate gyrus (DG)-CA3 circuit in resolving interference between ambiguous or uncertain threats and in preserving episodic content of remote aversive memories in hippocampal-cortical networks. We invoke cellular-, circuit-, and systems-based mechanisms by which adult-born dentate granule cells (DGCs) modulate pattern separation to influence resolution of interference and maintain precision of remote aversive memories. We discuss evidence for how these mechanisms are affected by stress, a risk factor for PTSD, to increase memory interference and decrease precision. Using this scaffold we ideate strategies to curb overgeneralization of fear in PTSD.
Collapse
|
37
|
Ikoma-Seki K, Nakamura K, Morishita S, Ono T, Sugiyama K, Nishino H, Hirano H, Murakoshi M. Role of LRP1 and ERK and cAMP Signaling Pathways in Lactoferrin-Induced Lipolysis in Mature Rat Adipocytes. PLoS One 2015; 10:e0141378. [PMID: 26506094 PMCID: PMC4623961 DOI: 10.1371/journal.pone.0141378] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 10/06/2015] [Indexed: 01/14/2023] Open
Abstract
Lactoferrin (LF) is a multifunctional glycoprotein present in milk. A clinical study showed that enteric-coated bovine LF tablets decrease visceral fat accumulation. Furthermore, animal studies revealed that ingested LF is partially delivered to mesenteric fat, and in vitro studies showed that LF promotes lipolysis in mature adipocytes. The aim of the present study was to determine the mechanism underlying the induction of lipolysis in mature adipocytes that is induced by LF. To address this question, we used proteomics techniques to analyze protein expression profiles. Mature adipocytes from primary cultures of rat mesenteric fat were collected at various times after exposure to LF. Proteomic analysis revealed that the expression levels of hormone-sensitive lipase (HSL), which catalyzes the rate-limiting step of lipolysis, were upregulated and that HSL was activated by protein kinase A within 15 min after the cells were treated with LF. We previously reported that LF increases the intracellular concentration of cyclic adenosine monophosphate (cAMP), suggesting that LF activates the cAMP signaling pathway. In this study, we show that the expression level and the activity of the components of the extracellular signal-regulated kinase (ERK) signaling pathway were upregulated. Moreover, LF increased the activity of the transcription factor cAMP response element binding protein (CREB), which acts downstream in the cAMP and ERK signaling pathways and regulates the expression levels of adenylyl cyclase and HSL. Moreover, silencing of the putative LF receptor low-density lipoprotein receptor-related protein 1 (LRP1) attenuated lipolysis in LF-treated adipocytes. These results suggest that LF promoted lipolysis in mature adipocytes by regulating the expression levels of proteins involved in lipolysis through controlling the activity of cAMP/ERK signaling pathways via LRP1.
Collapse
Affiliation(s)
- Keiko Ikoma-Seki
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
- * E-mail:
| | - Kanae Nakamura
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
| | - Satoru Morishita
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
| | - Tomoji Ono
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Keikichi Sugiyama
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Ritsumeikan University, Shiga, Japan
| | - Hoyoku Nishino
- Kyoto Prefectural University of Medicine, Kyoto, Japan
- Ritsumeikan University, Shiga, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Michiaki Murakoshi
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
- Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
38
|
Chen Y, Zhou S, Hua K, Xiao H, Li Z, Liu M, Luo R, Bi D, Zhou R, Jin H. Haemophilus parasuis infection activates chemokine RANTES in PK-15 cells. Mol Immunol 2015. [PMID: 26198698 DOI: 10.1016/j.molimm.2015.06.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
RANTES is a member of the CC chemokine involved in inflammation and immune response during pathogen infection. In our previous study, Haemophilus parasuis (H. parasuis), which is responsible for the great economic losses in the pig industry worldwide, has been shown to enhance RANTES expression in PK-15 cells. However, the mechanisms behind this biological phenomenon have remained unclear. In this study, we showed that H. parasuis infection significantly upregulated RANTES gene transcription in a time- and dose-dependent manner. Promoter analysis by site-directed mutagenesis indicated that the nuclear factor NF-κB binding site was the most important cis-regulatory element controlling H. parasuis-induced RANTES transcription. Inhibition of NF-κB and JNK activity also significantly reduced H. parasuis-induced RANTES production. In addition, TLRs signaling pathway was found to be involved in H. parasuis induced-RANTES expression. These results represent an important molecular mechanism whereby H. parasuis induced RANTES in the inflammatory response.
Collapse
Affiliation(s)
- Yushan Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shanshan Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Kexin Hua
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongde Xiao
- Hubei Center for Animal Disease Control and Prevention, Wuhan 430070, China
| | - Zili Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Mei Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Dingren Bi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
39
|
Crisafulli C, Drago A, Calabrò M, Spina E, Serretti A. A molecular pathway analysis informs the genetic background at risk for schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2015; 59:21-30. [PMID: 25554435 DOI: 10.1016/j.pnpbp.2014.12.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND Schizophrenia is a complex mental disorder marked by severely impaired thinking, delusional thoughts, hallucinations and poor emotional responsiveness. The biological mechanisms that lead to schizophrenia may be related to the genetic background of patients. Thus, a genetic perspective may help to unravel the molecular pathways disrupted in schizophrenia. METHODS In the present work, we used a molecular pathway analysis to identify the molecular pathways associated with schizophrenia. We collected data of genetic loci previously associated with schizophrenia, identified the genes located in those positions and created the metabolic pathways that are related to those genes' products. These pathways were tested for enrichment (a number of SNPs associated with the phenotype significantly higher than expected by chance) in a sample of schizophrenic patients and controls (4486 and 4477, respectively). RESULTS The molecular pathway that resulted from the identification of all the genes located in the loci previously found to be associated with schizophrenia was found to be enriched, as expected (permutated p(10(6))=9.9999e-06).We found 60 SNPs amongst 30 different genes with a strong association with schizophrenia. The genes are related to the pathways related to neurodevelopment, apoptosis, vesicle traffic, immune response and MAPK cascade. CONCLUSIONS The pathway related to the toll-like receptor family seemed to play a central role in the modulation/connection of various pathways whose disruption leads to schizophrenia. This pathway is related to the innate immune system, further stressing the role of immunological-related events in increasing the risk to schizophrenia.
Collapse
Affiliation(s)
- Concetta Crisafulli
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Antonio Drago
- Department of Biomedical and Neuromotor Sciences - DIBINEM, University of Bologna, Bologna, Italy; I.R.C.C.S. "San Giovanni di Dio", Fatebenefratelli, Brescia, Italy.
| | - Marco Calabrò
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Messina, Italy; Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy; IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Edoardo Spina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences - DIBINEM, University of Bologna, Bologna, Italy
| |
Collapse
|
40
|
Kreutzmann JC, Havekes R, Abel T, Meerlo P. Sleep deprivation and hippocampal vulnerability: changes in neuronal plasticity, neurogenesis and cognitive function. Neuroscience 2015; 309:173-90. [PMID: 25937398 DOI: 10.1016/j.neuroscience.2015.04.053] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/31/2015] [Accepted: 04/21/2015] [Indexed: 01/19/2023]
Abstract
Despite the ongoing fundamental controversy about the physiological function of sleep, there is general consensus that sleep benefits neuronal plasticity, which ultimately supports brain function and cognition. In agreement with this are numerous studies showing that sleep deprivation (SD) results in learning and memory impairments. Interestingly, such impairments appear to occur particularly when these learning and memory processes require the hippocampus, suggesting that this brain region may be particularly sensitive to the consequences of sleep loss. Although the molecular mechanisms underlying sleep and memory formation remain to be investigated, available evidence suggests that SD may impair hippocampal neuronal plasticity and memory processes by attenuating intracellular cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling which may lead to alterations in cAMP response element binding protein (CREB)-mediated gene transcription, neurotrophic signaling, and glutamate receptor expression. When restricted sleep becomes a chronic condition, it causes a reduction of hippocampal cell proliferation and neurogenesis, which may eventually lead to a reduction in hippocampal volume. Ultimately, by impairing hippocampal plasticity and function, chronically restricted and disrupted sleep contributes to cognitive disorders and psychiatric diseases.
Collapse
Affiliation(s)
- J C Kreutzmann
- Center for Behavior and Neurosciences, University of Groningen, The Netherlands; Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - R Havekes
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - T Abel
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| | - P Meerlo
- Center for Behavior and Neurosciences, University of Groningen, The Netherlands.
| |
Collapse
|
41
|
Abstract
In the past decades, a large number of neuropeptides with unknown functions have been identified in the brain. Among the newly discovered peptides, nociceptin or orphanin-FQ (N/OFQ) peptide has attracted considerable attention because of its sequence homology with the opioid peptide family. N/OFQ and its cognate receptor (NOP receptor) are distributed widely in the mammalian central nervous system, though particularly intense expression is found in corticolimbic structures. Such distinctive pattern of expression suggests a key role of N/OFQ system in higher brain functions, such as cognition and emotion. In this chapter, we will outline the findings supporting the role played by N/OFQ and NOP receptors in learning and memory and discuss the underlying mechanisms.
Collapse
|
42
|
Hippocampal cAMP/PKA/CREB is required for neuroprotective effect of acupuncture. Physiol Behav 2014; 139:482-90. [PMID: 25481359 DOI: 10.1016/j.physbeh.2014.12.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 11/21/2014] [Accepted: 12/02/2014] [Indexed: 01/18/2023]
Abstract
Acupuncture has beneficial effects in vascular dementia (VaD) patients. The underlying mechanism, however, remains unknown. The present study was designed to investigate whether the cAMP/PKA/CREB cascade is involved in the mechanism of acupuncture in cerebral multi-infarction rats. In this study, cerebral multi-infarction was modeled in adult Wistar rats by homologous blood clot emboli. After a two-week acupuncture treatment at Zusanli (ST36), hippocampal-dependent memory was tested by employing a radial arm maze test. The hippocampus was isolated for analyses of cAMP concentration, phosphodiesterase (PDE) activity and CREB/pCREB and ERK/pERK expressions. The Morris water maze (MWM) task and CREB phosphorylation were evaluated in the presence of PKA-selective peptide inhibitor (H89). The radial arm maze test results demonstrated that acupuncture treatment at ST36 reversed hippocampal-dependent memory in impaired animals. Compared to those of the impaired group, cAMP concentration, PKA activity and pCREB and pERK expressions were increased following acupuncture therapy. Finally, the blockade of PKA reversed the increase in CREB phosphorylation and the improvement in recognitive function induced by acupuncture treatment. These results suggest that acupuncture could improve hippocampus function by modulating the cAMP/PKA/CREB signaling pathway, which represents a molecular mechanism of acupuncture for recognitive function in cerebral multi-infarction rats.
Collapse
|
43
|
Insulin resistance in Alzheimer's disease. Neurobiol Dis 2014; 72 Pt A:92-103. [PMID: 25237037 DOI: 10.1016/j.nbd.2014.09.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 09/02/2014] [Accepted: 09/05/2014] [Indexed: 12/16/2022] Open
Abstract
Insulin is a key hormone regulating metabolism. Insulin binding to cell surface insulin receptors engages many signaling intermediates operating in parallel and in series to control glucose, energy, and lipids while also regulating mitogenesis and development. Perturbations in the function of any of these intermediates, which occur in a variety of diseases, cause reduced sensitivity to insulin and insulin resistance with consequent metabolic dysfunction. Chronic inflammation ensues which exacerbates compromised metabolic homeostasis. Since insulin has a key role in learning and memory as well as directly regulating ERK, a kinase required for the type of learning and memory compromised in early Alzheimer's disease (AD), insulin resistance has been identified as a major risk factor for the onset of AD. Animal models of AD or insulin resistance or both demonstrate that AD pathology and impaired insulin signaling form a reciprocal relationship. Of note are human and animal model studies geared toward improving insulin resistance that have led to the identification of the nuclear receptor and transcription factor, peroxisome proliferator-activated receptor gamma (PPARγ) as an intervention tool for early AD. Strategic targeting of alternate nodes within the insulin signaling network has revealed disease-stage therapeutic windows in animal models that coalesce with previous and ongoing clinical trial approaches. Thus, exploiting the connection between insulin resistance and AD provides powerful opportunities to delineate therapeutic interventions that slow or block the pathogenesis of AD.
Collapse
|
44
|
Roohbakhsh A, Parhiz H, Soltani F, Rezaee R, Iranshahi M. Neuropharmacological properties and pharmacokinetics of the citrus flavonoids hesperidin and hesperetin — A mini-review. Life Sci 2014; 113:1-6. [DOI: 10.1016/j.lfs.2014.07.029] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/16/2014] [Accepted: 07/21/2014] [Indexed: 12/24/2022]
|
45
|
Roy A, Modi KK, Khasnavis S, Ghosh S, Watson R, Pahan K. Enhancement of morphological plasticity in hippocampal neurons by a physically modified saline via phosphatidylinositol-3 kinase. PLoS One 2014; 9:e101883. [PMID: 25007337 PMCID: PMC4090203 DOI: 10.1371/journal.pone.0101883] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022] Open
Abstract
Increase of the density of dendritic spines and enhancement of synaptic transmission through ionotropic glutamate receptors are important events, leading to synaptic plasticity and eventually hippocampus-dependent spatial learning and memory formation. Here we have undertaken an innovative approach to upregulate hippocampal plasticity. RNS60 is a 0.9% saline solution containing charge-stabilized nanobubbles that are generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not NS (normal saline), PNS60 (saline containing a comparable level of oxygen without the TCP modification), or RNS10.3 (TCP-modified normal saline without excess oxygen), stimulated morphological plasticity and synaptic transmission via NMDA- and AMPA-sensitive calcium influx in cultured mouse hippocampal neurons. Using mRNA-based targeted gene array, real-time PCR, immunoblot, and immunofluorescence analyses, we further demonstrate that RNS60 stimulated the expression of many plasticity-associated genes in cultured hippocampal neurons. Activation of type IA, but not type IB, phosphatidylinositol-3 (PI-3) kinase by RNS60 together with abrogation of RNS60-mediated upregulation of plasticity-related proteins (NR2A and GluR1) and increase in spine density, neuronal size, and calcium influx by LY294002, a specific inhibitor of PI-3 kinase, suggest that RNS60 upregulates hippocampal plasticity via activation of PI-3 kinase. Finally, in the 5XFAD transgenic model of Alzheimer's disease (AD), RNS60 treatment upregulated expression of plasticity-related proteins PSD95 and NR2A and increased AMPA- and NMDA-dependent hippocampal calcium influx. These results describe a novel property of RNS60 in stimulating hippocampal plasticity, which may help AD and other dementias.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Khushbu K. Modi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Saurabh Khasnavis
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Supurna Ghosh
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Richard Watson
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
46
|
San Martín A, Pagani MR. Understanding intellectual disability through RASopathies. ACTA ACUST UNITED AC 2014; 108:232-9. [PMID: 24859216 DOI: 10.1016/j.jphysparis.2014.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 04/20/2014] [Accepted: 05/13/2014] [Indexed: 12/18/2022]
Abstract
Intellectual disability, commonly known as mental retardation in the International Classification of Disease from World Health Organization, is the term that describes an intellectual and adaptive cognitive disability that begins in early life during the developmental period. Currently the term intellectual disability is the preferred one. Although our understanding of the physiological basis of learning and learning disability is poor, a general idea is that such condition is quite permanent. However, investigations in animal models suggest that learning disability can be functional in nature and as such reversible through pharmacology or appropriate learning paradigms. A fraction of the cases of intellectual disability is caused by point mutations or deletions in genes that encode for proteins of the RAS/MAP kinase signaling pathway known as RASopathies. Here we examined the current understanding of the molecular mechanisms involved in this group of genetic disorders focusing in studies which provide evidence that intellectual disability is potentially treatable and curable. The evidence presented supports the idea that with the appropriate understanding of the molecular mechanisms involved, intellectual disability could be treated pharmacologically and perhaps through specific mechanistic-based teaching strategies.
Collapse
Affiliation(s)
- Alvaro San Martín
- Genetics of Learning Laboratory, Systems Neuroscience Section, Department of Physiology and Biophysics, School of Medicine, University of Buenos Aires, IFIBIO-Houssay-CONICET, C1121ABG Buenos Aires, Argentina
| | - Mario Rafael Pagani
- Genetics of Learning Laboratory, Systems Neuroscience Section, Department of Physiology and Biophysics, School of Medicine, University of Buenos Aires, IFIBIO-Houssay-CONICET, C1121ABG Buenos Aires, Argentina.
| |
Collapse
|
47
|
Abstract
Despite the ubiquity of sleep across phylogeny, its function remains elusive. In this review, we consider one compelling candidate: brain plasticity associated with memory processing. Focusing largely on hippocampus-dependent memory in rodents and humans, we describe molecular, cellular, network, whole-brain and behavioral evidence establishing a role for sleep both in preparation for initial memory encoding, and in the subsequent offline consolidation of memory. Sleep and sleep deprivation bidirectionally alter molecular signaling pathways that regulate synaptic strength and control plasticity-related gene transcription and protein translation. At the cellular level, sleep deprivation impairs cellular excitability necessary for inducing synaptic potentiation and accelerates the decay of long-lasting forms of synaptic plasticity. In contrast, rapid eye movement (REM) and non-rapid eye movement (NREM) sleep enhance previously induced synaptic potentiation, although synaptic de-potentiation during sleep has also been observed. Beyond single cell dynamics, large-scale cell ensembles express coordinated replay of prior learning-related firing patterns during subsequent NREM sleep. At the whole-brain level, somewhat analogous learning-associated hippocampal (re)activation during NREM sleep has been reported in humans. Moreover, the same cortical NREM oscillations associated with replay in rodents also promote human hippocampal memory consolidation, and this process can be manipulated using exogenous reactivation cues during sleep. Mirroring molecular findings in rodents, specific NREM sleep oscillations before encoding refresh human hippocampal learning capacity, while deprivation of sleep conversely impairs subsequent hippocampal activity and associated encoding. Together, these cross-descriptive level findings demonstrate that the unique neurobiology of sleep exerts powerful effects on molecular, cellular and network mechanisms of plasticity that govern both initial learning and subsequent long-term memory consolidation.
Collapse
|
48
|
Stuchlik A. Dynamic learning and memory, synaptic plasticity and neurogenesis: an update. Front Behav Neurosci 2014; 8:106. [PMID: 24744707 PMCID: PMC3978286 DOI: 10.3389/fnbeh.2014.00106] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/13/2014] [Indexed: 01/17/2023] Open
Abstract
Mammalian memory is the result of the interaction of millions of neurons in the brain and their coordinated activity. Candidate mechanisms for memory are synaptic plasticity changes, such as long-term potentiation (LTP). LTP is essentially an electrophysiological phenomenon manifested in hours-lasting increase on postsynaptic potentials after synapse tetanization. It is thought to ensure long-term changes in synaptic efficacy in distributed networks, leading to persistent changes in the behavioral patterns, actions and choices, which are often interpreted as the retention of information, i.e., memory. Interestingly, new neurons are born in the mammalian brain and adult hippocampal neurogenesis is proposed to provide a substrate for dynamic and flexible aspects of behavior such as pattern separation, prevention of interference, flexibility of behavior and memory resolution. This work provides a brief review on the memory and involvement of LTP and adult neurogenesis in memory phenomena.
Collapse
Affiliation(s)
- Ales Stuchlik
- Institute of Physiology, Academy of Sciences of the Czech Republic Prague, Czech Republic
| |
Collapse
|
49
|
Early stress evokes temporally distinct consequences on the hippocampal transcriptome, anxiety and cognitive behaviour. Int J Neuropsychopharmacol 2014; 17:289-301. [PMID: 24025219 DOI: 10.1017/s1461145713001004] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The early stress of maternal separation (ES) exerts long-lasting effects on cognition and anxiety. Recent evidence indicates enhanced hippocampus-dependent spatial learning in young adult ES animals, which shifts towards a decline in long-term memory in middle-aged life. Further, we find that ES animals exhibit enhanced anxiety in young adulthood that does not persist into middle-aged life. Here, we demonstrate unique, predominantly non-overlapping, hippocampal transcriptomes in young adult and middle-aged ES animals that accompany the temporally-specific behavioural consequences. Strikingly, the extent of gene dysregulation in middle-aged ES animals was substantially higher than in young adulthood. Functional analysis revealed distinct biological processes enriched at the two ages, highlighting the temporal shift in ES-evoked gene regulation. Our results suggest that ES history interacts with aging to exacerbate age-associated transcriptional changes and cognitive decline. qPCR profiling of histone deacetylases (Hdacs) and histone methyltransferases (HMTs) revealed an age-dependent, opposing regulation with decreased expression noted in young adult ES animals (Hdac 2, 7, 8, 9 and Suv39h1) and enhanced levels in middle-aged life (Hdac 2, 6, 8 and Suv39h1). While altered expression of histone modifying enzymes did not translate into global histone acetylation or methylation changes, we noted differential enrichment of histone acetylation and methylation modifications at the promoters of multiple genes regulated in the hippocampi of young adult and middle-aged ES animals. Our results highlight the differential molecular and behavioural consequences of ES across a life-span, and suggest a possible role for epigenetic mechanisms in contributing to the temporally-specific transcriptional changes following ES.
Collapse
|
50
|
Chen JF. Adenosine receptor control of cognition in normal and disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 119:257-307. [PMID: 25175970 DOI: 10.1016/b978-0-12-801022-8.00012-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adenosine and adenosine receptors (ARs) are increasingly recognized as important therapeutic targets for controlling cognition under normal and disease conditions for its dual roles of neuromodulation as well as of homeostatic function in the brain. This chapter first presents the unique ability of adenosine, by acting on the inhibitory A1 and facilitating A2A receptor, to integrate dopamine, glutamate, and BNDF signaling and to modulate synaptic plasticity (e.g., long-term potentiation and long-term depression) in brain regions relevant to learning and memory, providing the molecular and cellular bases for adenosine receptor (AR) control of cognition. This led to the demonstration of AR modulation of social recognition memory, working memory, reference memory, reversal learning, goal-directed behavior/habit formation, Pavlovian fear conditioning, and effort-related behavior. Furthermore, human and animal studies support that AR activity can also, through cognitive enhancement and neuroprotection, reverse cognitive impairments in animal models of Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and schizophrenia. Lastly, epidemiological evidence indicates that regular human consumption of caffeine, the most widely used psychoactive drug and nonselective AR antagonists, is associated with the reduced cognitive decline in aging and AD patients, and with the reduced risk in developing PD. Thus, there is a convergence of the molecular studies revealing AR as molecular targets for integrating neurotransmitter signaling and controlling synaptic plasticity, with animal studies demonstrating the strong procognitive impact upon AR antagonism in normal and disease brains and with epidemiological and clinical evidences in support of caffeine and AR drugs for therapeutic modulation of cognition. Since some of adenosine A2A receptor antagonists are already in phase III clinical trials for motor benefits in PD patients with remarkable safety profiles, additional animal and human studies to better understand the mechanism underlying the AR-mediated control of cognition under normal and disease conditions will provide the required rationale to stimulate the necessary clinical investigation to rapidly translate adenosine and AR drug as a novel strategy to control memory impairment in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA; The Molecular Medicine Institute, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|