1
|
Pan G, Chai L, Chen R, Yuan Q, Song Z, Feng W, Wei J, Yang Z, Zhang Y, Xie G, Yan A, Lv Q, Wang C, Zhao Y, Wang Y. Potential mechanism of Qinggong Shoutao pill alleviating age-associated memory decline based on integration strategy. PHARMACEUTICAL BIOLOGY 2024; 62:105-119. [PMID: 38145345 PMCID: PMC10763866 DOI: 10.1080/13880209.2023.2291689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 11/30/2023] [Indexed: 12/26/2023]
Abstract
CONTEXT Qinggong Shoutao Wan (QGSTW) is a pill used as a traditional medicine to treat age-associated memory decline (AAMI). However, its potential mechanisms are unclear. OBJECTIVE This study elucidates the possible mechanisms of QGSTW in treating AAMI. MATERIALS AND METHODS Network pharmacology and molecular docking approaches were utilized to identify the potential pathway by which QGSTW alleviates AAMI. C57BL/6J mice were divided randomly into control, model, and QGSTW groups. A mouse model of AAMI was established by d-galactose, and the pathways that QGSTW acts on to ameliorate AAMI were determined by ELISA, immunofluorescence staining and Western blotting after treatment with d-gal (100 mg/kg) and QGSTW (20 mL/kg) for 12 weeks. RESULTS Network pharmacology demonstrated that the targets of the active components were significantly enriched in the cAMP signaling pathway. AKT1, FOS, GRIN2B, and GRIN1 were the core target proteins. QGSTW treatment increased the discrimination index from -16.92 ± 7.06 to 23.88 ± 15.94% in the novel location test and from -19.54 ± 5.71 to 17.55 ± 6.73% in the novel object recognition test. ELISA showed that QGSTW could increase the levels of cAMP. Western blot analysis revealed that QGSTW could upregulate the expression of PKA, CREB, c-Fos, GluN1, GluA1, CaMKII-α, and SYN. Immunostaining revealed that the expression of SYN was decreased in the CA1 and DG. DISCUSSION AND CONCLUSIONS This study not only provides new insights into the mechanism of QGSTW in the treatment of AAMI but also provides important information and new research ideas for the discovery of traditional Chinese medicine compounds that can treat AAMI.
Collapse
Affiliation(s)
- Guiyun Pan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lijuan Chai
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Yuan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Song
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanying Feng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinna Wei
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihua Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhang Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guinan Xie
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - An Yan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Lv
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Caijun Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingqiang Zhao
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
2
|
Hu Y, Lu F, Yang H, Pan Q, Wu X. Effect of artificial sugar supplement on the lifespan and learning memory ability of honey bee (Apis cerana) worker bee offspring. JOURNAL OF ECONOMIC ENTOMOLOGY 2024; 117:1723-1728. [PMID: 39120062 DOI: 10.1093/jee/toae176] [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: 10/31/2023] [Revised: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024]
Abstract
Honeybees maintain their growth and reproduction mainly by collecting nutrients from nectar-source plants. Apis cerana, a unique species of honeybee in China, is capable of sporadically collecting nectar. In traditional beekeeping, sugar syrup or a honey-water solution must be artificially fed to bees to supplement their diet during rainy weather or nectar-deficient periods. In this study, 2 groups of honeybee colonies were each fed sugar syrup or a honey-water solution, and a third group consisting of colonies that were allowed to naturally forage without any dietary supplement was used as the control. The effects of the 2 sugar sources on A. cerana worker bee offspring were compared. The results showed that the sugar source affected the lifespan and learning memory of the worker bee offspring. The lifespan, learning memory ability, and expression of related genes in the sugar syrup group were significantly lower than those in the honey-water solution and natural nectar foraging groups (P < 0.05). A honey-water solution supplement was more beneficial to the healthy development of worker bee offspring than a sugar syrup supplement when the colonies lacked dietary resources. These findings provide a theoretical basis that can guide beekeepers in choosing the appropriate dietary supplements for honeybees.
Collapse
Affiliation(s)
- Yueyang Hu
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, PR China
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, PR China
- Jiangxi Anyuan Honeybee Science and Technology Backyard, Anyuan 342100, PR China
| | - Fangming Lu
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, PR China
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, PR China
- Jiangxi Anyuan Honeybee Science and Technology Backyard, Anyuan 342100, PR China
| | - Heyan Yang
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, PR China
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, PR China
| | - Qizhong Pan
- Jiangxi Ganzhou Agricultural College, Ganzhou 341199, PR China
| | - Xiaobo Wu
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, PR China
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, PR China
- Jiangxi Anyuan Honeybee Science and Technology Backyard, Anyuan 342100, PR China
| |
Collapse
|
3
|
Ji J, Xu Y, Wang Y, Zhang G, Tian X, Zhang Y, Ren J. miR-351-5p regulation of CPEB3 affecting aluminium-induced learning and memory impairment in SD rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124973. [PMID: 39307336 DOI: 10.1016/j.envpol.2024.124973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/07/2024] [Accepted: 09/14/2024] [Indexed: 09/29/2024]
Abstract
Aluminium exposure has been found to impair learning and memory abilities; however, the underlying molecular mechanisms remain unclear. In this study we conducted a double luciferase reporter assay to determine whether miR-351-5p regulates cytoplasmic polyadenylation element binding protein (CPEB) 3 mRNA. To this end, we overexpressed and inhibited miR-351-5p via stereotaxic microinjections of adeno-associated virus (AAV) into the hippocampus of Sprague Dawley rats in a sub-chronic aluminium exposure model to examine learning and memory ability using Morris water maze. Ultrastructural electron microscopy and Golgi staining were used to examine morphological changes in hippocampal neurons. In addition, we examined the levels of synaptic plasticity-related proteins (PRPs) and CPEB3 to determine the involvement of the miR-351-5P/CPEB3/PRPs pathway in aluminium neurotoxicity. Sub-chronic aluminium exposure reduced the spatial learning and memory ability of rats. Overexpression of AAV-miR-351-5P in the hippocampus aggravated the impairment of spatial learning and memory abilities of aluminium-treated rats, whereas inhibition of AAV-miR-351-5p expression alleviated it. Western blotting suggested that sub-chronic aluminium exposure increased miR-351-5p levels and reduced the expression of CPEB3 and PRPs in the hippocampus. Treatment with an AAV-miR-351-5p inhibitor partially recovered CPEB3 and PRPs. Double luciferase reporter assay results showed that CPEB3 was a direct target of miR-351-5p, while electron microscopy suggested that aluminium could damage mitochondria and synapses in the CA1 of the hippocampus. Golgi staining results indicated that aluminium could reduce the number of dendritic spines in hippocampal neurons. Inhibition of miR-351-5p restored the synaptic structure and growth of dendritic spines in the hippocampus. The involvement of the miR-351-5P/CPEB3/RPPs pathway in aluminium neurotoxicity was confirmed. Our findings suggest that inhibition of miR-351-5p can alleviate learning and memory impairments by increasing CPEB3 and PRPs.
Collapse
Affiliation(s)
- Jingjing Ji
- Shanxi Medical University Fenyang College, China; Pathology Department, Shanxi Fenyang Hospital, China
| | - Yirong Xu
- Shanxi Medical University Fenyang College, China; Pathology Department, Shanxi Fenyang Hospital, China.
| | - Yanni Wang
- School of Public Health, Shanxi Medical University, China
| | | | - Xiaoai Tian
- Pathology Department, Shanxi Fenyang Hospital, China
| | - Yeping Zhang
- Shanxi Medical University Fenyang College, China
| | - Jiaxuan Ren
- Shanxi Medical University Fenyang College, China
| |
Collapse
|
4
|
Chowdhury MAR, Haq MM, Lee JH, Jeong S. Multi-faceted regulation of CREB family transcription factors. Front Mol Neurosci 2024; 17:1408949. [PMID: 39165717 PMCID: PMC11333461 DOI: 10.3389/fnmol.2024.1408949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
cAMP response element-binding protein (CREB) is a ubiquitously expressed nuclear transcription factor, which can be constitutively activated regardless of external stimuli or be inducibly activated by external factors such as stressors, hormones, neurotransmitters, and growth factors. However, CREB controls diverse biological processes including cell growth, differentiation, proliferation, survival, apoptosis in a cell-type-specific manner. The diverse functions of CREB appear to be due to CREB-mediated differential gene expression that depends on cAMP response elements and multi-faceted regulation of CREB activity. Indeed, the transcriptional activity of CREB is controlled at several levels including alternative splicing, post-translational modification, dimerization, specific transcriptional co-activators, non-coding small RNAs, and epigenetic regulation. In this review, we present versatile regulatory modes of CREB family transcription factors and discuss their functional consequences.
Collapse
Affiliation(s)
- Md Arifur Rahman Chowdhury
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Md Mazedul Haq
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jeong Hwan Lee
- Division of Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Sangyun Jeong
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
5
|
Thiem J, Viskadourou M, Gaitanidis A, Stravopodis DJ, Strauß R, Duch C, Consoulas C. Biological aging of two innate behaviors of Drosophila melanogaster: Escape climbing versus courtship learning and memory. PLoS One 2024; 19:e0293252. [PMID: 38593121 PMCID: PMC11003613 DOI: 10.1371/journal.pone.0293252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Motor and cognitive aging can severely affect life quality of elderly people and burden health care systems. In search for diagnostic behavioral biomarkers, it has been suggested that walking speed can predict forms of cognitive decline, but in humans, it remains challenging to separate the effects of biological aging and lifestyle. We examined a possible association of motor and cognitive decline in Drosophila, a genetic model organism of healthy aging. Long term courtship memory is present in young male flies but absent already during mid life (4-8 weeks). By contrast, courtship learning index and short term memory (STM) are surprisingly robust and remain stable through mid (4-8 weeks) and healthy late life (>8 weeks), until courtship performance collapses suddenly at ~4.5 days prior to death. By contrast, climbing speed declines gradually during late life (>8 weeks). The collapse of courtship performance and short term memory close to the end of life occur later and progress with a different time course than the gradual late life decline in climbing speed. Thus, during healthy aging in male Drosophila, climbing and courtship motor behaviors decline differentially. Moreover, cognitive and motor performances decline at different time courses. Differential behavioral decline during aging may indicate different underlying causes, or alternatively, a common cause but different thresholds for defects in different behaviors.
Collapse
Affiliation(s)
- Jessica Thiem
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Rhineland-Palatinate, Germany
| | - Maria Viskadourou
- Laboratory of Experimental Physiology, Medical School, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Alexandros Gaitanidis
- Laboratory of Experimental Physiology, Medical School, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Dimitrios J. Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Roland Strauß
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Rhineland-Palatinate, Germany
| | - Carsten Duch
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Rhineland-Palatinate, Germany
| | - Christos Consoulas
- Laboratory of Experimental Physiology, Medical School, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| |
Collapse
|
6
|
Dow LF, Case AM, Paustian MP, Pinkerton BR, Simeon P, Trippier PC. The evolution of small molecule enzyme activators. RSC Med Chem 2023; 14:2206-2230. [PMID: 37974956 PMCID: PMC10650962 DOI: 10.1039/d3md00399j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 11/19/2023] Open
Abstract
There is a myriad of enzymes within the body responsible for maintaining homeostasis by providing the means to convert substrates to products as and when required. Physiological enzymes are tightly controlled by many signaling pathways and their products subsequently control other pathways. Traditionally, most drug discovery efforts focus on identifying enzyme inhibitors, due to upregulation being prevalent in many diseases and the existence of endogenous substrates that can be modified to afford inhibitor compounds. As enzyme downregulation and reduction of endogenous activators are observed in multiple diseases, the identification of small molecules with the ability to activate enzymes has recently entered the medicinal chemistry toolbox to afford chemical probes and potential therapeutics as an alternative means to intervene in diseases. In this review we highlight the progress made in the identification and advancement of non-kinase enzyme activators and their potential in treating various disease states.
Collapse
Affiliation(s)
- Louise F Dow
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Alfie M Case
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Megan P Paustian
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Braeden R Pinkerton
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Princess Simeon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center Omaha NE 68106 USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center Omaha NE 68106 USA
| |
Collapse
|
7
|
Wang CM, Wu CY, Lin CE, Hsu MC, Lin JC, Huang CC, Lien TY, Lin HK, Chang TW, Chiang HC. Forgotten memory storage and retrieval in Drosophila. Nat Commun 2023; 14:7153. [PMID: 37935667 PMCID: PMC10630420 DOI: 10.1038/s41467-023-42753-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/20/2023] [Indexed: 11/09/2023] Open
Abstract
Inaccessibility of stored memory in ensemble cells through the forgetting process causes animals to be unable to respond to natural recalling cues. While accumulating evidence has demonstrated that reactivating memory-stored cells can switch cells from an inaccessible state to an accessible form and lead to recall of previously learned information, the underlying cellular and molecular mechanisms remain elusive. The current study used Drosophila as a model to demonstrate that the memory of one-trial aversive olfactory conditioning, although inaccessible within a few hours after learning, is stored in KCαβ and retrievable after mild retraining. One-trial aversive conditioning triggers protein synthesis to form a long-lasting cellular memory trace, approximately 20 days, via creb in KCαβ, and a transient cellular memory trace, approximately one day, via orb in MBON-α3. PPL1-α3 negatively regulates forgotten one-trial conditioning memory retrieval. The current study demonstrated that KCαβ, PPL1-α3, and MBON-α3 collaboratively regulate the formation of forgotten one-cycle aversive conditioning memory formation and retrieval.
Collapse
Affiliation(s)
- Chih-Ming Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Chun-Yuan Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Chen-En Lin
- Department of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Ming-Chi Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Jing-Chun Lin
- Department of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Chuan-Chin Huang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Ting-Yu Lien
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Hsin-Kai Lin
- Department of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Ting-Wei Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Hsueh-Cheng Chiang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC.
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC.
| |
Collapse
|
8
|
Axelrod S, Li X, Sun Y, Lincoln S, Terceros A, O’Neil J, Wang Z, Nguyen A, Vora A, Spicer C, Shapiro B, Young MW. The Drosophila blood-brain barrier regulates sleep via Moody G protein-coupled receptor signaling. Proc Natl Acad Sci U S A 2023; 120:e2309331120. [PMID: 37831742 PMCID: PMC10589661 DOI: 10.1073/pnas.2309331120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 10/15/2023] Open
Abstract
Sleep is vital for most animals, yet its mechanism and function remain unclear. We found that permeability of the BBB (blood-brain barrier)-the organ required for the maintenance of homeostatic levels of nutrients, ions, and other molecules in the brain-is modulated by sleep deprivation (SD) and can cell-autonomously effect sleep changes. We observed increased BBB permeability in known sleep mutants as well as in acutely sleep-deprived animals. In addition to molecular tracers, SD-induced BBB changes also increased the penetration of drugs used in the treatment of brain pathologies. After chronic/genetic or acute SD, rebound sleep or administration of the sleeping aid gaboxadol normalized BBB permeability, showing that SD effects on the BBB are reversible. Along with BBB permeability, RNA levels of the BBB master regulator moody are modulated by sleep. Conversely, altering BBB permeability alone through glia-specific modulation of moody, gαo, loco, lachesin, or neuroglian-each a well-studied regulator of BBB function-was sufficient to induce robust sleep phenotypes. These studies demonstrate a tight link between BBB permeability and sleep and indicate a unique role for the BBB in the regulation of sleep.
Collapse
Affiliation(s)
- Sofia Axelrod
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Xiaoling Li
- International Personalized Cancer Center, Tianjin Cancer Hospital Airport Hospital, Tianjin300308, China
| | - Yingwo Sun
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Samantha Lincoln
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrea Terceros
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Jenna O’Neil
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrew Nguyen
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Aabha Vora
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Carmen Spicer
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Benjamin Shapiro
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Michael W. Young
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| |
Collapse
|
9
|
Chen CC, Lin HW, Feng KL, Tseng DW, de Belle JS, Chiang AS. A subset of cholinergic mushroom body neurons blocks long-term memory formation in Drosophila. Cell Rep 2023; 42:112974. [PMID: 37590142 DOI: 10.1016/j.celrep.2023.112974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/22/2022] [Accepted: 07/29/2023] [Indexed: 08/19/2023] Open
Abstract
Long-term memory (LTM) requires learning-induced synthesis of new proteins allocated to specific neurons and synapses in a neural circuit. Not all learned information, however, becomes permanent memory. How the brain gates relevant information into LTM remains unclear. In Drosophila adults, weak learning after a single training session in an olfactory aversive task typically does not induce protein-synthesis-dependent LTM. Instead, strong learning after multiple spaced training sessions is required. Here, we report that pre-synaptic active-zone protein synthesis and cholinergic signaling from the early α/β subset of mushroom body (MB) neurons produce a downstream inhibitory effect on LTM formation. When we eliminated inhibitory signaling from these neurons, weak learning was then sufficient to form LTM. This bidirectional circuit mechanism modulates the transition between distinct memory phase functions in different subpopulations of MB neurons in the olfactory memory circuit.
Collapse
Affiliation(s)
- Chun-Chao Chen
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Hsuan-Wen Lin
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kuan-Lin Feng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Der-Wan Tseng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - J Steven de Belle
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA; Department of Psychological Sciences, University of San Diego, San Diego, CA 92110, USA; School of Life Sciences, University of Nevada, Las Vegas, NV 89154, USA; MnemOdyssey LLC, Escondido, CA 92027, USA
| | - Ann-Shyn Chiang
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80780, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan; Kavli Institute for Brain and Mind, University of California at San Diego, La Jolla, CA 92093-0526, USA.
| |
Collapse
|
10
|
Davis RL. Learning and memory using Drosophila melanogaster: a focus on advances made in the fifth decade of research. Genetics 2023; 224:iyad085. [PMID: 37212449 PMCID: PMC10411608 DOI: 10.1093/genetics/iyad085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023] Open
Abstract
In the last decade, researchers using Drosophila melanogaster have made extraordinary progress in uncovering the mysteries underlying learning and memory. This progress has been propelled by the amazing toolkit available that affords combined behavioral, molecular, electrophysiological, and systems neuroscience approaches. The arduous reconstruction of electron microscopic images resulted in a first-generation connectome of the adult and larval brain, revealing complex structural interconnections between memory-related neurons. This serves as substrate for future investigations on these connections and for building complete circuits from sensory cue detection to changes in motor behavior. Mushroom body output neurons (MBOn) were discovered, which individually forward information from discrete and non-overlapping compartments of the axons of mushroom body neurons (MBn). These neurons mirror the previously discovered tiling of mushroom body axons by inputs from dopamine neurons and have led to a model that ascribes the valence of the learning event, either appetitive or aversive, to the activity of different populations of dopamine neurons and the balance of MBOn activity in promoting avoidance or approach behavior. Studies of the calyx, which houses the MBn dendrites, have revealed a beautiful microglomeruluar organization and structural changes of synapses that occur with long-term memory (LTM) formation. Larval learning has advanced, positioning it to possibly lead in producing new conceptual insights due to its markedly simpler structure over the adult brain. Advances were made in how cAMP response element-binding protein interacts with protein kinases and other transcription factors to promote the formation of LTM. New insights were made on Orb2, a prion-like protein that forms oligomers to enhance synaptic protein synthesis required for LTM formation. Finally, Drosophila research has pioneered our understanding of the mechanisms that mediate permanent and transient active forgetting, an important function of the brain along with acquisition, consolidation, and retrieval. This was catalyzed partly by the identification of memory suppressor genes-genes whose normal function is to limit memory formation.
Collapse
Affiliation(s)
- Ronald L Davis
- Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, 130 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
11
|
Chowdhury MAR, An J, Jeong S. The Pleiotropic Face of CREB Family Transcription Factors. Mol Cells 2023; 46:399-413. [PMID: 37013623 PMCID: PMC10336275 DOI: 10.14348/molcells.2023.2193] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 04/05/2023] Open
Abstract
cAMP responsive element-binding protein (CREB) is one of the most intensively studied phosphorylation-dependent transcription factors that provide evolutionarily conserved mechanisms of differential gene expression in vertebrates and invertebrates. Many cellular protein kinases that function downstream of distinct cell surface receptors are responsible for the activation of CREB. Upon functional dimerization of the activated CREB to cis-acting cAMP responsive elements within the promoters of target genes, it facilitates signal-dependent gene expression. From the discovery of CREB, which is ubiquitously expressed, it has been proven to be involved in a variety of cellular processes that include cell proliferation, adaptation, survival, differentiation, and physiology, through the control of target gene expression. In this review, we highlight the essential roles of CREB proteins in the nervous system, the immune system, cancer development, hepatic physiology, and cardiovascular function and further discuss a wide range of CREB-associated diseases and molecular mechanisms underlying the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Md. Arifur Rahman Chowdhury
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| | - Jungeun An
- Division of Life Sciences (Life Sciences Major), Jeonbuk National University, Jeonju 54896, Korea
| | - Sangyun Jeong
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
12
|
Yu JX, Hui YM, Xue JA, Qu JB, Ling SQ, Wang W, Zeng XN, Liu JL. Formation characteristics of long-term memory in Bactrocera dorsalis. INSECT SCIENCE 2023; 30:829-843. [PMID: 36151856 DOI: 10.1111/1744-7917.13119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/23/2022] [Accepted: 09/12/2022] [Indexed: 06/15/2023]
Abstract
Studies on insects have contributed significantly to a better understanding of learning and memory, which is a necessary cognitive capability for all animals. Although the formation of memory has been studied in some model insects, more evidence is required to clarify the characteristics of memory formation, especially long-term memory (LTM), which is important for reliably storing information. Here, we explored this question by examining Bactrocera dorsalis, an agricultural pest with excellent learning abilities. Using the classical conditioning paradigm of the olfactory proboscis extension reflex (PER), we found that paired conditioning with multiple trials (>3) spaced with an intertrial interval (≥10 min) resulted in stable memory that lasted for at least 3 d. Furthermore, even a single conditioning trial was sufficient for the formation of a 2-d memory. With the injection of protein inhibitors, protein-synthesis-dependent memory was confirmed to start 4 h after training, and its dependence on translation and transcription differed. Moreover, the results revealed that the dependence of memory on protein translation exhibited a time-window effect (4-6 h). Our findings provide an integrated view of LTM in insects, suggesting common mechanisms in LTM formation that play a key role in the biological basis of memory.
Collapse
Affiliation(s)
- Jin-Xin Yu
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Yan-Min Hui
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Jun-Ao Xue
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Jia-Bao Qu
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Si-Quan Ling
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Silviculture, Protection, and Utilization, Guangdong Academy of Forestry, Guangzhou, China
| | - Wei Wang
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Xin-Nian Zeng
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Jia-Li Liu
- Guangdong Engineering Research Center for Insect Behavior Regulation, College of Plant Protection, South China Agricultural University, Guangzhou, China
| |
Collapse
|
13
|
Brunswick CA, Baldwin DJ, Bodinayake KK, McKenna AR, Lo CY, Bellfy L, Urban MW, Stuart EM, Murakami S, Smies CW, Kwapis JL. The clock gene Per1 is necessary in the retrosplenial cortex-but not in the suprachiasmatic nucleus-for incidental learning in young and aging male mice. Neurobiol Aging 2023; 126:77-90. [PMID: 36958103 PMCID: PMC10106450 DOI: 10.1016/j.neurobiolaging.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/03/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023]
Abstract
Aging impairs both circadian rhythms and memory, though the relationship between these impairments is not fully understood. Circadian rhythms are largely dictated by clock genes within the body's central pacemaker, the suprachiasmatic nucleus (SCN), though these genes are also expressed in local clocks throughout the body. As circadian rhythms can directly affect memory performance, one possibility is that memory deficits observed with age are downstream of global circadian rhythm disruptions stemming from the SCN. Here, we demonstrate that expression of clock gene Period1 within a memory-relevant cortical structure, the retrosplenial cortex (RSC), is necessary for incidental learning, and that age-related disruption of Period1 within the RSC-but not necessarily the SCN-contributes to cognitive decline. These data expand the known functions of clock genes beyond maintaining circadian rhythms and suggests that age-associated changes in clock gene expression modulates circadian rhythms and memory performance in a brain region-dependent manner.
Collapse
Affiliation(s)
- Chad A Brunswick
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Derek J Baldwin
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Kasuni K Bodinayake
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA
| | | | - Chen-Yu Lo
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Lauren Bellfy
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Mark W Urban
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Emily M Stuart
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Shoko Murakami
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Chad W Smies
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, University Park, PA.
| |
Collapse
|
14
|
de Andrade CV, Soliani AG, Cerutti SM. Standardized extract of Ginkgo biloba treatment and novelty on the weak encoding of spatial recognition memory in rats. Learn Mem 2023; 30:85-95. [PMID: 37072140 PMCID: PMC10165992 DOI: 10.1101/lm.053755.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 04/20/2023]
Abstract
Long-term memory (LTM) formation is dependent on neurochemical changes that guarantee that a recently formed memory (short-term memory [STM]) remains in the specific neural circuitry via the consolidation process. The persistence of recognition memory has been evidenced by using behavioral tagging in young adult rats, but it has not been effective on aging. Here, we investigated the effects of treatment with a standardized extract of Ginkgo biloba (EGb) associated with novelty on the consolidation of object location memory (OLM) and its persistence after weak training of spatial object preference in young adult and aged rats. The object location task used in this study included two habituation sessions, training sessions associated or not associated with EGb treatment and contextual novelty, and short-term or long-term retention testing sessions. Altogether, our data showed that treatment with EGb associated with novelty close to the time of encoding resulted in STM that lasted for 1 h and persisted for 24 h for both young adult and aged rats. In aged rats, the cooperative mechanisms induced robust long-term OLM. Our findings support and extend our knowledge about recognition memory in aged rats and the modulating effects of EGb treatment and contextual novelty on the persistence of memory.
Collapse
Affiliation(s)
- Carla Vitor de Andrade
- The Graduate Program in Structural and Functional Biology, Universidade Federal de São Paulo, São Paulo 09972-270, Brazil
- Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de São Paulo, São Paulo 09972-270, Brazil
| | - Andressa Gabriela Soliani
- Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de São Paulo, São Paulo 09972-270, Brazil
- the Graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, São Paulo 09972-270, Brazil
| | - Suzete Maria Cerutti
- The Graduate Program in Structural and Functional Biology, Universidade Federal de São Paulo, São Paulo 09972-270, Brazil
- Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de São Paulo, São Paulo 09972-270, Brazil
- the Graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, São Paulo 09972-270, Brazil
| |
Collapse
|
15
|
Yin JCP, Cui E, Hardin PE, Zhou H. Circadian disruption of memory consolidation in Drosophila. Front Syst Neurosci 2023; 17:1129152. [PMID: 37034015 PMCID: PMC10073699 DOI: 10.3389/fnsys.2023.1129152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
The role of the circadian system in memory formation is an important question in neurobiology. Despite this hypothesis being intuitively appealing, the existing data is confusing. Recent work in Drosophila has helped to clarify certain aspects of the problem, but the emerging sense is that the likely mechanisms are more complex than originally conceptualized. In this report, we identify a post-training window of time (during consolidation) when the circadian clock and its components are involved in memory formation. In the broader context, our data suggest that circadian biology might have multiple roles during memory formation. Testing for its roles at multiple timepoints, and in different cells, will be necessary to resolve some of the conflicting data.
Collapse
Affiliation(s)
- Jerry C. P. Yin
- Laboratory of Genetics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Neurology Department, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- *Correspondence: Jerry C. P. Yin
| | - Ethan Cui
- Laboratory of Genetics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Paul E. Hardin
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, College Station, TX, United States
| | - Hong Zhou
- Laboratory of Genetics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| |
Collapse
|
16
|
Neurogenetics of Memory, Learning, and Forgetting. Neurogenetics 2023. [DOI: 10.1007/978-3-031-07793-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Gupta VS, Kale PP. Combinatory Approaches Targeting Cognitive Impairments and Memory Enhancement: A Review. Curr Drug Targets 2023; 24:55-70. [PMID: 36173073 DOI: 10.2174/1389450123666220928152743] [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: 04/01/2022] [Revised: 06/21/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022]
Abstract
The objective of this paper is to look at how natural medicines can improve cognition and memory when used with sildenafil, a popular erectile dysfunction medicine that also has nootropic properties. Newer treatment strategies to treat the early stages of these diseases need to be developed. Multiple factors lead to complex pathophysiological conditions, which are responsible for various long-term complications. In this review, a combination of treatments targeting these pathologies is discussed. These combinations may help manage early and later phases of cognitive impairments. The purpose of this article is to discuss a link between these pathologies and a combinational approach with the objective of considering newer therapeutic strategies in the treatment of cognitive impairments. The natural drugs and their ingredients play a major role in the management of disease progression. Additionally, their combination with sildenafil allows for more efficacy and better response. Studies showing the effectiveness of natural drugs and sildenafil are mentioned, and how these combinations could be beneficial for the treatment of cognitive impairments and amnesia are summarised. Furthermore, preclinical and clinical trials are required to explore the medicinal potential of these drug combinations.
Collapse
Affiliation(s)
- Varun Santosh Gupta
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai 400056, India
| | - Pravin Popatrao Kale
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai 400056, India
| |
Collapse
|
18
|
Pribadi AK, Chalasani SH. Fear conditioning in invertebrates. Front Behav Neurosci 2022; 16:1008818. [PMID: 36439964 PMCID: PMC9686301 DOI: 10.3389/fnbeh.2022.1008818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/19/2022] [Indexed: 09/30/2023] Open
Abstract
Learning to identify and predict threats is a basic skill that allows animals to avoid harm. Studies in invertebrates like Aplysia californica, Drosophila melanogaster, and Caenorhabditis elegans have revealed that the basic mechanisms of learning and memory are conserved. We will summarize these studies and highlight the common pathways and mechanisms in invertebrate fear-associated behavioral changes. Fear conditioning studies utilizing electric shock in Aplysia and Drosophila have demonstrated that serotonin or dopamine are typically involved in relaying aversive stimuli, leading to changes in intracellular calcium levels and increased presynaptic neurotransmitter release and short-term changes in behavior. Long-term changes in behavior typically require multiple, spaced trials, and involve changes in gene expression. C. elegans studies have demonstrated these basic aversive learning principles as well; however, fear conditioning has yet to be explicitly demonstrated in this model due to stimulus choice. Because predator-prey relationships can be used to study learned fear in a naturalistic context, this review also summarizes what is known about predator-induced behaviors in these three organisms, and their potential applications for future investigations into fear conditioning.
Collapse
Affiliation(s)
- Amy K. Pribadi
- Biological Sciences Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, United States
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Sreekanth H. Chalasani
- Biological Sciences Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, United States
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
19
|
Kelvington BA, Nickl-Jockschat T, Abel T. Neurobiological insights into twice-exceptionality: Circuits, cells, and molecules. Neurobiol Learn Mem 2022; 195:107684. [PMID: 36174887 PMCID: PMC9888516 DOI: 10.1016/j.nlm.2022.107684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 02/02/2023]
Abstract
Twice-exceptional learners face a unique set of challenges arising from the intersection of extraordinary talent and disability. Neurobiology research has the capacity to complement pedagogical research and provide support for twice-exceptional learners. Very few studies have attempted to specifically address the neurobiological underpinnings of twice-exceptionality. However, neurobiologists have built a broad base of knowledge in nervous system function spanning from the level of neural circuits to the molecular basis of behavior. It is known that distinct neural circuits mediate different neural functions, which suggests that 2e learning may result from enhancement in one circuit and disruption in another. Neural circuits are known to adapt and change in response to experience, a cellular process known as neuroplasticity. Plasticity is controlled by a bidirectional connection between the synapse, where neural signals are received, and the nucleus, where regulated gene expression can return to alter synaptic function. Complex molecular mechanisms compose this connection in distinct neural circuits, and genetic alterations in these mechanisms are associated with both memory enhancements and psychiatric disorder. Understanding the consequences of these changes at the molecular, cellular, and circuit levels will provide critical insights into the neurobiological bases of twice-exceptionality.
Collapse
Affiliation(s)
- Benjamin A Kelvington
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
20
|
Ojea Ramos S, Feld M, Fustiñana MS. Contributions of extracellular-signal regulated kinase 1/2 activity to the memory trace. Front Mol Neurosci 2022; 15:988790. [PMID: 36277495 PMCID: PMC9580372 DOI: 10.3389/fnmol.2022.988790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/02/2022] [Indexed: 11/15/2022] Open
Abstract
The ability to learn from experience and consequently adapt our behavior is one of the most fundamental capacities enabled by complex and plastic nervous systems. Next to cellular and systems-level changes, learning and memory formation crucially depends on molecular signaling mechanisms. In particular, the extracellular-signal regulated kinase 1/2 (ERK), historically studied in the context of tumor growth and proliferation, has been shown to affect synaptic transmission, regulation of neuronal gene expression and protein synthesis leading to structural synaptic changes. However, to what extent the effects of ERK are specifically related to memory formation and stabilization, or merely the result of general neuronal activation, remains unknown. Here, we review the signals leading to ERK activation in the nervous system, the subcellular ERK targets associated with learning-related plasticity, and how neurons with activated ERK signaling may contribute to the formation of the memory trace.
Collapse
Affiliation(s)
- Santiago Ojea Ramos
- Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mariana Feld
- Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | |
Collapse
|
21
|
Andreani T, Rosensweig C, Sisobhan S, Ogunlana E, Kath W, Allada R. Circadian programming of the ellipsoid body sleep homeostat in Drosophila. eLife 2022; 11:e74327. [PMID: 35735904 PMCID: PMC9270026 DOI: 10.7554/elife.74327] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Homeostatic and circadian processes collaborate to appropriately time and consolidate sleep and wake. To understand how these processes are integrated, we scheduled brief sleep deprivation at different times of day in Drosophila and find elevated morning rebound compared to evening. These effects depend on discrete morning and evening clock neurons, independent of their roles in circadian locomotor activity. In the R5 ellipsoid body sleep homeostat, we identified elevated morning expression of activity dependent and presynaptic gene expression as well as the presynaptic protein BRUCHPILOT consistent with regulation by clock circuits. These neurons also display elevated calcium levels in response to sleep loss in the morning, but not the evening consistent with the observed time-dependent sleep rebound. These studies reveal the circuit and molecular mechanisms by which discrete circadian clock neurons program a homeostatic sleep center.
Collapse
Affiliation(s)
- Tomas Andreani
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Clark Rosensweig
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Shiju Sisobhan
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Emmanuel Ogunlana
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - William Kath
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States
| | - Ravi Allada
- Department of Neurobiology, Northwestern UniversityChicagoUnited States
| |
Collapse
|
22
|
Nakai J, Chikamoto N, Fujimoto K, Totani Y, Hatakeyama D, Dyakonova VE, Ito E. Insulin and Memory in Invertebrates. Front Behav Neurosci 2022; 16:882932. [PMID: 35558436 PMCID: PMC9087806 DOI: 10.3389/fnbeh.2022.882932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Insulin and insulin-like peptides (ILP) help to maintain glucose homeostasis, whereas insulin-like growth factor (IGF) promotes the growth and differentiation of cells in both vertebrates and invertebrates. It is sometimes difficult to distinguish between ILP and IGF in invertebrates, however, because in some cases ILP has the same function as IGF. In the present review, therefore, we refer to these peptides as ILP/IGF signaling (IIS) in invertebrates, and discuss the role of IIS in memory formation after classical conditioning in invertebrates. In the arthropod Drosophila melanogaster, IIS is involved in aversive olfactory memory, and in the nematode Caenorhabditis elegans, IIS controls appetitive/aversive response to NaCl depending on the duration of starvation. In the mollusk Lymnaea stagnalis, IIS has a critical role in conditioned taste aversion. Insulin in mammals is also known to play an important role in cognitive function, and many studies in humans have focused on insulin as a potential treatment for Alzheimer’s disease. Although analyses of tissue and cellular levels have progressed in mammals, the molecular mechanisms, such as transcriptional and translational levels, of IIS function in cognition have been far advanced in studies using invertebrates. We anticipate that the present review will help to pave the way for studying the effects of insulin, ILPs, and IGFs in cognitive function across phyla.
Collapse
Affiliation(s)
- Junko Nakai
- Department of Biology, Waseda University, Tokyo, Japan
| | | | | | - Yuki Totani
- Department of Biology, Waseda University, Tokyo, Japan
| | - Dai Hatakeyama
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Varvara E. Dyakonova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Etsuro Ito
- Department of Biology, Waseda University, Tokyo, Japan
- Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Etsuro Ito
| |
Collapse
|
23
|
Adaptation of vestibulo-ocular and optokinetic reflexes after massed and spaced vestibulo-ocular motor learning. Behav Brain Res 2022; 426:113837. [DOI: 10.1016/j.bbr.2022.113837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/06/2022] [Accepted: 03/07/2022] [Indexed: 11/21/2022]
|
24
|
Lesar A, Tahir J, Wolk J, Gershow M. Switch-like and persistent memory formation in individual Drosophila larvae. eLife 2021; 10:e70317. [PMID: 34636720 PMCID: PMC8510578 DOI: 10.7554/elife.70317] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/27/2021] [Indexed: 11/15/2022] Open
Abstract
Associative learning allows animals to use past experience to predict future events. The circuits underlying memory formation support immediate and sustained changes in function, often in response to a single example. Larval Drosophila is a genetic model for memory formation that can be accessed at molecular, synaptic, cellular, and circuit levels, often simultaneously, but existing behavioral assays for larval learning and memory do not address individual animals, and it has been difficult to form long-lasting memories, especially those requiring synaptic reorganization. We demonstrate a new assay for learning and memory capable of tracking the changing preferences of individual larvae. We use this assay to explore how activation of a pair of reward neurons changes the response to the innately aversive gas carbon dioxide (CO2). We confirm that when coupled to CO2 presentation in appropriate temporal sequence, optogenetic reward reduces avoidance of CO2. We find that learning is switch-like: all-or-none and quantized in two states. Memories can be extinguished by repeated unrewarded exposure to CO2 but are stabilized against extinction by repeated training or overnight consolidation. Finally, we demonstrate long-lasting protein synthesis dependent and independent memory formation.
Collapse
Affiliation(s)
- Amanda Lesar
- Department of Physics, New York UniversityNew YorkUnited States
| | - Javan Tahir
- Department of Physics, New York UniversityNew YorkUnited States
| | - Jason Wolk
- Department of Physics, New York UniversityNew YorkUnited States
| | - Marc Gershow
- Department of Physics, New York UniversityNew YorkUnited States
- Center for Neural Science, New York UniversityNew YorkUnited States
- NYU Neuroscience Institute, New York University Langone Medical CenterNew YorkUnited States
| |
Collapse
|
25
|
CREBA and CREBB in two identified neurons gate long-term memory formation in Drosophila. Proc Natl Acad Sci U S A 2021; 118:2100624118. [PMID: 34507985 PMCID: PMC8449312 DOI: 10.1073/pnas.2100624118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2021] [Indexed: 11/18/2022] Open
Abstract
Episodic events are frequently consolidated into labile memory but are not necessarily transferred to persistent long-term memory (LTM). Regulatory mechanisms leading to LTM formation are poorly understood, however, especially at the resolution of identified neurons. Here, we demonstrate enhanced LTM following aversive olfactory conditioning in Drosophila when the transcription factor cyclic AMP response element binding protein A (CREBA) is induced in just two dorsal-anterior-lateral (DAL) neurons. Our experiments show that this process is regulated by protein-gene interactions in DAL neurons: (1) crebA transcription is induced by training and repressed by crebB overexpression, (2) CREBA bidirectionally modulates LTM formation, (3) crebA overexpression enhances training-induced gene transcription, and (4) increasing membrane excitability enhances LTM formation and gene expression. These findings suggest that activity-dependent gene expression in DAL neurons during LTM formation is regulated by CREB proteins.
Collapse
|
26
|
How CM, Lin TA, Liao VHC. Early-life chronic di(2-ethylhexyl)phthalate exposure worsens age-related long-term associative memory decline associated with insulin/IGF-1 signaling and CRH-1/CREB in Caenorhabditis elegans. JOURNAL OF HAZARDOUS MATERIALS 2021; 417:126044. [PMID: 34229382 DOI: 10.1016/j.jhazmat.2021.126044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 06/13/2023]
Abstract
The ubiquitous contamination of di(2-ethylhexyl)phthalate (DEHP) in the environment, biota, and food poses potential ecological and human health risks. DEHP exposure can adversely affect learning and memory, yet the underlying mechanisms remain unclear. In this study, Caenorhabditis elegans was used to investigate the effect of early-life DEHP exposure on age-related long-term associative memory (LTAM) decline, as well as the associations with the cAMP-responsive element-binding protein (CREB) transcription factor and insulin/IGF-1 signaling (IIS). We showed that early-life exposure to DEHP reduced LTAM in wild-type worms at day-0 adulthood. Chronic exposure to DEHP from the L1 stage to day-5 adulthood worsened the age-dependent decline of LTAM. Moreover, the effect of DEHP on age-related LTAM requires CRH-1, a homolog of CREB. Mutations in daf-2, the sole receptor of C. elegans IIS, ameliorated the inhibition of LTAM by DEHP, and the effect depended on daf-16. In addition, daf-2 mutation restored the CRH-1 level in DEHP-exposed worms, and the effect required daf-16. Our study suggests that early-life chronic exposure to DEHP worsens age-related LTAM decline and the effect is associated with CRH-1 and IIS in C. elegans. The evolutionary conservation of IIS and CREB implies possible adverse effects by DEHP across species.
Collapse
Affiliation(s)
- Chun Ming How
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Ting-An Lin
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
27
|
Wang L, Zhang L, Gong X, Fu J, Gan Y, Hou M, Nie Q, Xiang J, Xiao Y, Wang Y, Zheng S, Yang L, Chen H, Xiang M, Liu Y, Li DW. PP-1β and PP-2Aα modulate cAMP response element-binding protein (CREB) functions in aging control and stress response through de-regulation of αB-crystallin gene and p300-p53 signaling axis. Aging Cell 2021; 20:e13458. [PMID: 34425033 PMCID: PMC8441381 DOI: 10.1111/acel.13458] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 07/04/2021] [Accepted: 08/06/2021] [Indexed: 02/06/2023] Open
Abstract
The function of the transcription factor, cAMP response element‐binding protein (CREB), is activated through S133 phosphorylation by PKA and others. Regarding its inactivation, it is not well defined. cAMP response element‐binding protein plays an essential role in promoting cell proliferation, neuronal survival and the synaptic plasticity associated with long‐term memory. Our recent studies have shown that CREB is an important player in mediating stress response. Here, we have demonstrated that CREB regulates aging process through suppression of αB‐crystallin and activation of the p300‐p53‐Bak/Bax signaling axis. First, we determined that two specific protein phosphatases, PP‐1β and PP‐2Aα, can inactivate CREB through S133 dephosphorylation. Subsequently, we demonstrated that cells expressing the S133A‐CREB, a mutant mimicking constant dephosphorylation at S133, suppress CREB functions in aging control and stress response. Mechanistically, S133A‐CREB not only significantly suppresses CREB control of αB‐crystallin gene, but also represses CREB‐mediated activation of p53 acetylation and downstream Bak/Bax genes. cAMP response element‐binding protein suppression of αB‐crystallin and its activation of p53 acetylation are major molecular events observed in human cataractous lenses of different age groups. Together, our results demonstrate that PP‐1β and PP‐2Aα modulate CREB functions in aging control and stress response through de‐regulation of αB‐crystallin gene and p300‐p53‐Bax/Bak signaling axis, which regulates human cataractogenesis in the aging lens.
Collapse
Affiliation(s)
- Ling Wang
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Lan Zhang
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Xiao‐Dong Gong
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Jia‐Ling Fu
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yu‐Wen Gan
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Min Hou
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Qian Nie
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Jia‐Wen Xiang
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yuan Xiao
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yan Wang
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Shu‐Yu Zheng
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Lan Yang
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Huimin Chen
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Meng‐Qing Xiang
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yizhi Liu
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - David Wan‐Cheng Li
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| |
Collapse
|
28
|
Zhang Y, Zhang J, Wang S. The Role of Rapamycin in Healthspan Extension via the Delay of Organ Aging. Ageing Res Rev 2021; 70:101376. [PMID: 34089901 DOI: 10.1016/j.arr.2021.101376] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 05/07/2021] [Accepted: 05/30/2021] [Indexed: 12/17/2022]
Abstract
Aging can not only shorten a healthy lifespan, but can also lead to multi-organ dysfunction and failure. Anti-aging is a complex and worldwide conundrum for eliminating the various pathologies of senility. The past decade has seen great progress in the understanding of the aging-associated signaling pathways and their application for developing anti-aging approaches. Currently, some drugs can improve quality of life. The activation of mammalian target of rapamycin (mTOR) signaling is one of the core and detrimental mechanisms related to aging; rapamycin can reduce the rate of aging, improve age-related diseases by inhibiting the mTOR pathway, and prolong lifespan and healthspan effectively. However, the current evidence for rapamycin in lifespan extension and organ aging is fragmented and scattered. In this review, we summarize the efficacy and safety of rapamycin in prolonging a healthy lifespan by systematically alleviating aging in multiple organ systems, i.e., the nervous, urinary, digestive, circulatory, motor, respiratory, endocrine, reproductive, integumentary and immune systems, to provide a theoretical basis for the future clinical application of rapamycin in anti-aging.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
29
|
Potential evidence of peripheral learning and memory in the arms of dwarf cuttlefish, Sepia bandensis. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2021; 207:575-594. [PMID: 34121131 DOI: 10.1007/s00359-021-01499-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
CREB (cAMP response element-binding) transcription factors are conserved markers of memory formation in the brain and peripheral circuits. We provide immunohistochemical evidence of CREB phosphorylation in the dwarf cuttlefish, Sepia bandensis, following the inaccessible prey (IP) memory experiment. During the IP experiment, cuttlefish are shown prey enclosed in a transparent tube, and tentacle strikes against the tube decrease over time as the cuttlefish learns the prey is inaccessible. The cues driving IP learning are unclear but may include sensory inputs from arms touching the tube. The neural activity marker, anti-phospho-CREB (anti-pCREB) was used to determine whether IP training stimulated cuttlefish arm sensory neurons. pCREB immunoreactivity occurred along the oral surface of the arms, including the suckers and epithelial folds surrounding the suckers. pCREB increased in the epithelial folds and suckers of trained cuttlefish. We found differential pCREB immunoreactivity along the distal-proximal axis of trained arms, with pCREB concentrated distally. Unequal CREB phosphorylation occurred among the 4 trained arm pairs, with arm pairs 1 and 2 containing more pCREB. The resulting patterns of pCREB in trained arms suggest that the arms obtain cues that may be salient for learning and memory of the IP experiment.
Collapse
|
30
|
Zia A, Pourbagher-Shahri AM, Farkhondeh T, Samarghandian S. Molecular and cellular pathways contributing to brain aging. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2021; 17:6. [PMID: 34118939 PMCID: PMC8199306 DOI: 10.1186/s12993-021-00179-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Aging is the leading risk factor for several age-associated diseases such as neurodegenerative diseases. Understanding the biology of aging mechanisms is essential to the pursuit of brain health. In this regard, brain aging is defined by a gradual decrease in neurophysiological functions, impaired adaptive neuroplasticity, dysregulation of neuronal Ca2+ homeostasis, neuroinflammation, and oxidatively modified molecules and organelles. Numerous pathways lead to brain aging, including increased oxidative stress, inflammation, disturbances in energy metabolism such as deregulated autophagy, mitochondrial dysfunction, and IGF-1, mTOR, ROS, AMPK, SIRTs, and p53 as central modulators of the metabolic control, connecting aging to the pathways, which lead to neurodegenerative disorders. Also, calorie restriction (CR), physical exercise, and mental activities can extend lifespan and increase nervous system resistance to age-associated neurodegenerative diseases. The neuroprotective effect of CR involves increased protection against ROS generation, maintenance of cellular Ca2+ homeostasis, and inhibition of apoptosis. The recent evidence about the modem molecular and cellular methods in neurobiology to brain aging is exhibiting a significant potential in brain cells for adaptation to aging and resistance to neurodegenerative disorders.
Collapse
Affiliation(s)
- Aliabbas Zia
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ali Mohammad Pourbagher-Shahri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), 9717853577 Birjand, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
31
|
Mozolewski P, Jeziorek M, Schuster CM, Bading H, Frost B, Dobrowolski R. The role of nuclear Ca2+ in maintaining neuronal homeostasis and brain health. J Cell Sci 2021; 134:jcs254904. [PMID: 33912918 PMCID: PMC8084578 DOI: 10.1242/jcs.254904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nuclear Ca2+ has emerged as one of the most potent mediators of the dialogue between neuronal synapses and the nucleus that regulates heterochromatin states, transcription factor activity, nuclear morphology and neuronal gene expression induced by synaptic activity. Recent studies underline the importance of nuclear Ca2+ signaling in long-lasting, activity-induced adaptation and maintenance of proper brain function. Diverse forms of neuroadaptation require transient nuclear Ca2+ signaling and cyclic AMP-responsive element-binding protein (CREB1, referred to here as CREB) as its prime target, which works as a tunable switch to drive and modulate specific gene expression profiles associated with memory, pain, addiction and neuroprotection. Furthermore, a reduction of nuclear Ca2+ levels has been shown to be neurotoxic and a causal factor driving the progression of neurodegenerative disorders, as well as affecting neuronal autophagy. Because of its central role in the brain, deficits in nuclear Ca2+ signaling may underlie a continuous loss of neuroprotection in the aging brain, contributing to the pathophysiology of Alzheimer's disease. In this Review, we discuss the principles of the 'nuclear calcium hypothesis' in the context of human brain function and its role in controlling diverse forms of neuroadaptation and neuroprotection. Furthermore, we present the most relevant and promising perspectives for future studies.
Collapse
Affiliation(s)
- Pawel Mozolewski
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Maciej Jeziorek
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Christoph M. Schuster
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 345 and INF 366, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 345 and INF 366, 69120 Heidelberg, Germany
| | - Bess Frost
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health, San Antonio, San Antonio, TX 78229, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, San Antonio, TX 78229, USA
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health, San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
32
|
Pham NC, Kim YG, Kim SJ, Kim CH. Efficacy of spaced learning in adaptation of optokinetic response. Brain Behav 2021; 11:e01944. [PMID: 33185985 PMCID: PMC7821562 DOI: 10.1002/brb3.1944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION The superiority of spaced training, in which repeated training sessions are given with resting intervals, over massed training in learning efficacy has been well established. However, longer duration of total training time has been required for spaced training than massed training because spacing intervals intervene between training sessions in spaced training. Thus, the learning efficacy may not be simply compared between spaced and massed training in terms of "time efficiency." The aim of the present study was to investigate the efficacy of spaced and massed training using adaptation of horizontal optokinetic reflex (hOKR) in mice. METHODS Training paradigms were categorized into seven groups according to the duration of spacing interval, keeping total duration of hOKR training including spacing almost equal in all training paradigms. RESULTS The amount of short-term hOKR gain increase immediately after the 60 min hOKR training was not significantly different among seven training paradigms. The hOKR adaptation was still in progress during a spacing interval, and the increment in hOKR gain tended to be greater with the longer spacing interval. The increase in hOKR gain was maintained until 48 hr after the end of training in both massed and spaced training. CONCLUSION The short-term learning effect was not significantly different among training paradigms regardless of spacing interval in hOKR adaptation, which suggests that the spacing effect is robust enough to overcome the shortage of optokinetic training cycles in hOKR adaptation.
Collapse
Affiliation(s)
- Ngoc Chien Pham
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University Medical Center, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Yong Gyu Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Memory Network Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Memory Network Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea.,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang-Hee Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Konkuk University Medical Center, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Cattaneo V, San Martin A, Lew SE, Gelb BD, Pagani MR. Repeating or spacing learning sessions are strategies for memory improvement with shared molecular and neuronal components. Neurobiol Learn Mem 2020; 172:107233. [PMID: 32360730 PMCID: PMC7451235 DOI: 10.1016/j.nlm.2020.107233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 03/05/2020] [Accepted: 04/25/2020] [Indexed: 11/21/2022]
Abstract
Intellectual disability is a common feature in genetic disorders with enhanced RAS-ERK1/2 signaling, including neurofibromatosis type 1 (NF1) and Noonan syndrome (NS). Additional training trials and additional spacing between trials, respectively, restores memory deficits in animal models of NF1 and NS. However, the relationship between the underlying mechanisms in these strategies remain obscure. Here, we developed an approach to examine the effect of adding training trials or spacing to a weak training protocol and used genetic and behavioral manipulations in Drosophila to explore such question. We found that repetition and spacing effects are highly related, being equally effective to improve memory in control flies and sharing mechanistic bases, including the requirement of RAS activity in mushroom body neurons and protein synthesis dependence. After spacing or repeating learning trials, memory improvement depends on the formation of long-term memory (LTM). Moreover, a disease-related gain-of-function RasV152G allele impaired LTM. Using minimal training protocols, we established that both learning strategies were also equally effective for memory rescue in the RasV152G mutant and showed non-additive interaction of the spacing and repetition effects. Memory improvement was never detected after Ras inhibition. We conclude that memory improvement by spacing or repeating training trials are two ways of using the same molecular resources, including RAS-ERK1/2-dependent signaling. This evidence supports the concept that learning problems in RAS-related disorders depend on the impaired ability to exploit the repetition and the spacing effect required for long-term memory induction.
Collapse
Affiliation(s)
- Verónica Cattaneo
- IFIBIO-Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires - CONICET, 2155 Paraguay Street, Buenos Aires, Argentina
| | - Alvaro San Martin
- IFIBIO-Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires - CONICET, 2155 Paraguay Street, Buenos Aires, Argentina
| | - Sergio E Lew
- Instituto de Ingeniería Biomédica, Facultad de Ingeniería, Universidad de Buenos Aires, Argentina
| | - Bruce D Gelb
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mario R Pagani
- IFIBIO-Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires - CONICET, 2155 Paraguay Street, Buenos Aires, Argentina.
| |
Collapse
|
34
|
Wang L, Nie Q, Gao M, Yang L, Xiang JW, Xiao Y, Liu FY, Gong XD, Fu JL, Wang Y, Nguyen QD, Liu Y, Liu M, Li DWC. The transcription factor CREB acts as an important regulator mediating oxidative stress-induced apoptosis by suppressing αB-crystallin expression. Aging (Albany NY) 2020; 12:13594-13617. [PMID: 32554860 PMCID: PMC7377838 DOI: 10.18632/aging.103474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/03/2020] [Indexed: 12/17/2022]
Abstract
The general transcription factor, CREB has been shown to play an essential role in promoting cell proliferation, neuronal survival and synaptic plasticity in the nervous system. However, its function in stress response remains to be elusive. In the present study, we demonstrated that CREB plays a major role in mediating stress response. In both rat lens organ culture and mouse lens epithelial cells (MLECs), CREB promotes oxidative stress-induced apoptosis. To confirm that CREB is a major player mediating the above stress response, we established stable lines of MLECs stably expressing CREB and found that they are also very sensitive to oxidative stress-induced apoptosis. To define the underlying mechanism, RNAseq analysis was conducted. It was found that CREB significantly suppressed expression of the αB-crystallin gene to sensitize CREB-expressing cells undergoing oxidative stress-induced apoptosis. CREB knockdown via CRISPR/CAS9 technology led to upregulation of αB-crystallin and enhanced resistance against oxidative stress-induced apoptosis. Moreover, overexpression of exogenous human αB-crystallin can restore the resistance against oxidative stress-induced apoptosis. Finally, we provided first evidence that CREB directly regulates αB-crystallin gene. Together, our results demonstrate that CREB is an important transcription factor mediating stress response, and it promotes oxidative stress-induced apoptosis by suppressing αB-crystallin expression.
Collapse
Affiliation(s)
- Ling Wang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Qian Nie
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Meng Gao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
- Medical College, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Lan Yang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Jia-Wen Xiang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Yuan Xiao
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Fang-Yuan Liu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Xiao-Dong Gong
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Jia-Ling Fu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Yan Wang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Quan Dong Nguyen
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Yizhi Liu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - David Wan-Cheng Li
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| |
Collapse
|
35
|
Spiking activity in the human hippocampus prior to encoding predicts subsequent memory. Proc Natl Acad Sci U S A 2020; 117:13767-13770. [PMID: 32482860 PMCID: PMC7306990 DOI: 10.1073/pnas.2001338117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Encoding activity in the medial temporal lobe, presumably evoked by the presentation of stimuli (postonset activity), is known to predict subsequent memory. However, several independent lines of research suggest that preonset activity also affects subsequent memory. We investigated the role of preonset and postonset single-unit and multiunit activity recorded from epilepsy patients as they completed a continuous recognition task. In this task, words were presented in a continuous series and eventually began to repeat. For each word, the patient's task was to decide whether it was novel or repeated. We found that preonset spiking activity in the hippocampus (when the word was novel) predicted subsequent memory (when the word was later repeated). Postonset activity during encoding also predicted subsequent memory, but was simply a continuation of preonset activity. The predictive effect of preonset spiking activity was much stronger in the hippocampus than in three other brain regions (amygdala, anterior cingulate, and prefrontal cortex). In addition, preonset and postonset activity around the encoding of novel words did not predict memory performance for novel words (i.e., correctly classifying the word as novel), and preonset and postonset activity around the time of retrieval did not predict memory performance for repeated words (i.e., correctly classifying the word as repeated). Thus, the only predictive effect was between preonset activity (along with its postonset continuation) at the time of encoding and subsequent memory. Taken together, these findings indicate that preonset hippocampal activity does not reflect general arousal/attention but instead reflects what we term "attention to encoding."
Collapse
|
36
|
Haubrich J, Bernabo M, Baker AG, Nader K. Impairments to Consolidation, Reconsolidation, and Long-Term Memory Maintenance Lead to Memory Erasure. Annu Rev Neurosci 2020; 43:297-314. [PMID: 32097575 DOI: 10.1146/annurev-neuro-091319-024636] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An enduring problem in neuroscience is determining whether cases of amnesia result from eradication of the memory trace (storage impairment) or if the trace is present but inaccessible (retrieval impairment). The most direct approach to resolving this question is to quantify changes in the brain mechanisms of long-term memory (BM-LTM). This approach argues that if the amnesia is due to a retrieval failure, BM-LTM should remain at levels comparable to trained, unimpaired animals. Conversely, if memories are erased, BM-LTM should be reduced to resemble untrained levels. Here we review the use of BM-LTM in a number of studies that induced amnesia by targeting memory maintenance or reconsolidation. The literature strongly suggests that such amnesia is due to storage rather than retrieval impairments. We also describe the shortcomings of the purely behavioral protocol that purports to show recovery from amnesia as a method of understanding the nature of amnesia.
Collapse
Affiliation(s)
- Josué Haubrich
- Department of Psychology, McGill University, Montreal, Quebec H3A 1B1, Canada;
| | - Matteo Bernabo
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Andrew G Baker
- Department of Psychology, McGill University, Montreal, Quebec H3A 1B1, Canada;
| | - Karim Nader
- Department of Psychology, McGill University, Montreal, Quebec H3A 1B1, Canada;
| |
Collapse
|
37
|
Syntaxin1A Neomorphic Mutations Promote Rapid Recovery from Isoflurane Anesthesia in Drosophila melanogaster. Anesthesiology 2020; 131:555-568. [PMID: 31356232 DOI: 10.1097/aln.0000000000002850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Mutations in the presynaptic protein syntaxin1A modulate general anesthetic effects in vitro and in vivo. Coexpression of a truncated syntaxin1A protein confers resistance to volatile and intravenous anesthetics, suggesting a target mechanism distinct from postsynaptic inhibitory receptor processes. Hypothesizing that recovery from anesthesia may involve a presynaptic component, the authors tested whether syntaxin1A mutations facilitated recovery from isoflurane anesthesia in Drosophila melanogaster. METHODS A truncated syntaxin1A construct was expressed in Drosophila neurons. The authors compared effects on isoflurane induction versus recovery in syntaxin1A mutant animals by probing behavioral responses to mechanical stimuli. The authors also measured synaptic responses from the larval neuromuscular junction using sharp intracellular recordings, and performed Western blots to determine whether the truncated syntaxin1A is associated with presynaptic core complexes. RESULTS Drosophila expressing a truncated syntaxin1A (syx, n = 40) were resistant to isoflurane induction for a behavioral responsiveness endpoint (ED50 0.30 ± 0.01% isoflurane, P < 0.001) compared with control (0.240 ± 0.002% isoflurane, n = 40). Recovery from isoflurane anesthesia was also faster, with syx-expressing flies showing greater levels of responsiveness earlier in recovery (reaction proportion 0.66 ± 0.48, P < 0.001, n = 68) than controls (0.22 ± 0.42, n = 68 and 0.33 ± 0.48, n = 66). Measuring excitatory junction potentials of larvae coexpressing the truncated syntaxin1A protein showed a greater recovery of synaptic function, compared with controls (17.39 ± 3.19 mV and 10.29 ± 4.88 mV, P = 0.014, n = 8 for both). The resistance-promoting truncated syntaxin1A was not associated with presynaptic core complexes, in the presence or absence of isoflurane anesthesia. CONCLUSIONS The same neomorphic syntaxin1A mutation that confers isoflurane resistance in cell culture and nematodes also produces isoflurane resistance in Drosophila. Resistance in Drosophila is, however, most evident at the level of recovery from anesthesia, suggesting that the syntaxin1A target affects anesthesia maintenance and recovery processes rather than induction. The absence of truncated syntaxin1A from the presynaptic complex suggests that the resistance-promoting effect of this molecule occurs before core complex formation.
Collapse
|
38
|
Elliott T. Dynamic Integrative Synaptic Plasticity Explains the Spacing Effect in the Transition from Short- to Long-Term Memory. Neural Comput 2019; 31:2212-2251. [PMID: 31525308 DOI: 10.1162/neco_a_01227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Repeated stimuli that are spaced apart in time promote the transition from short- to long-term memory, while massing repetitions together does not. Previously, we showed that a model of integrative synaptic plasticity, in which plasticity induction signals are integrated by a low-pass filter before plasticity is expressed, gives rise to a natural timescale at which to repeat stimuli, hinting at a partial account of this spacing effect. The account was only partial because the important role of neuromodulation was not considered. We now show that by extending the model to allow dynamic integrative synaptic plasticity, the model permits synapses to robustly discriminate between spaced and massed repetition protocols, suppressing the response to massed stimuli while maintaining that to spaced stimuli. This is achieved by dynamically coupling the filter decay rate to neuromodulatory signaling in a very simple model of the signaling cascades downstream from cAMP production. In particular, the model's parameters may be interpreted as corresponding to the duration and amplitude of the waves of activity in the MAPK pathway. We identify choices of parameters and repetition times for stimuli in this model that optimize the ability of synapses to discriminate between spaced and massed repetition protocols. The model is very robust to reasonable changes around these optimal parameters and times, but for large changes in parameters, the model predicts that massed and spaced stimuli cannot be distinguished or that the responses to both patterns are suppressed. A model of dynamic integrative synaptic plasticity therefore explains the spacing effect under normal conditions and also predicts its breakdown under abnormal conditions.
Collapse
Affiliation(s)
- Terry Elliott
- Department of Electronics and Computer Science, University of Southampton, Highfield, Southampton, SO17 1BJ, U.K.
| |
Collapse
|
39
|
Mariottini C, Munari L, Gunzel E, Seco JM, Tzavaras N, Hansen J, Stern SA, Gao V, Aleyasin H, Sharma A, Azeloglu EU, Hodes GE, Russo SJ, Huff V, Birtwistle MR, Blitzer RD, Alberini CM, Iyengar R. Wilm's tumor 1 promotes memory flexibility. Nat Commun 2019; 10:3756. [PMID: 31434897 PMCID: PMC6704057 DOI: 10.1038/s41467-019-11781-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Under physiological conditions, strength and persistence of memory must be regulated in order to produce behavioral flexibility. In fact, impairments in memory flexibility are associated with pathologies such as post-traumatic stress disorder or autism; however, the underlying mechanisms that enable memory flexibility are still poorly understood. Here, we identify transcriptional repressor Wilm's Tumor 1 (WT1) as a critical synaptic plasticity regulator that decreases memory strength, promoting memory flexibility. WT1 is activated in the hippocampus following induction of long-term potentiation (LTP) or learning. WT1 knockdown enhances CA1 neuronal excitability, LTP and long-term memory whereas its overexpression weakens memory retention. Moreover, forebrain WT1-deficient mice show deficits in both reversal, sequential learning tasks and contextual fear extinction, exhibiting impaired memory flexibility. We conclude that WT1 limits memory strength or promotes memory weakening, thus enabling memory flexibility, a process that is critical for learning from new experiences.
Collapse
Affiliation(s)
- Chiara Mariottini
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
| | - Leonardo Munari
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Ellen Gunzel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Joseph M Seco
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Nikos Tzavaras
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Jens Hansen
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Sarah A Stern
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Center for Neural Science, New York University, New York, 10003, NY, USA
| | - Virginia Gao
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Center for Neural Science, New York University, New York, 10003, NY, USA
| | - Hossein Aleyasin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Ali Sharma
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Evren U Azeloglu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Georgia E Hodes
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Scott J Russo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Vicki Huff
- Department of Genetics, M.D. Anderson Cancer Center, University of Texas, Houston, 77030, TX, USA
| | - Marc R Birtwistle
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Robert D Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA
| | - Cristina M Alberini
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
- Center for Neural Science, New York University, New York, 10003, NY, USA.
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
- Systems Biology Center, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, NY, USA.
| |
Collapse
|
40
|
Differential Regulation of Innate and Learned Behavior by Creb1/Crh-1 in Caenorhabditis elegans. J Neurosci 2019; 39:7934-7946. [PMID: 31413073 PMCID: PMC6774408 DOI: 10.1523/jneurosci.0006-19.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/17/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022] Open
Abstract
Memory formation is crucial for the survival of animals. Here, we study the effect of different crh-1 [Caenorhabditis elegans homolog of mammalian cAMP response element binding protein 1 (CREB1)] isoforms on the ability of C. elegans to form long-term memory (LTM). Null mutants in creb1/crh-1 are defective in LTM formation across phyla. We show that a specific isoform of CREB1/CRH-1, CRH-1e, is primarily responsible for memory related functions of the transcription factor in C. elegans. Silencing of CRH-1e-expressing neurons during training for LTM formation abolishes the LTM of the animal. Further, CRH-1e expression in RIM neurons is sufficient to rescue LTM defects of creb1/crh-1-null mutants. We go on to show that apart from being LTM defective, creb1/crh-1-null animals show defects in innate chemotaxis behavior. We further characterize the amino acids K247 and K266 as responsible for the LTM related functions of CREB1/CRH-1 while being dispensable for its innate chemotaxis behavior. These findings provide insight into the spatial and temporal workings of a crucial transcription factor that can be further exploited to find CREB1 targets involved in the process of memory formation. SIGNIFICANCE STATEMENT This study elucidates the role of a specific isoform of CREB1/CRH-1, CRH-1e, in Caenorhabditis elegans memory formation and chemosensation. Removal of this single isoform of creb1/crh-1 shows defects in long-term memory formation in the animal and expression of CREB1/CRH-1e in a single pair of neurons is sufficient to rescue the memory defects seen in the mutant animals. We further show that two specific amino acids of CRH-1 are required for the process of memory formation in the animal.
Collapse
|
41
|
Karam CS, Jones SK, Javitch JA. Come Fly with Me: An overview of dopamine receptors in Drosophila melanogaster. Basic Clin Pharmacol Toxicol 2019; 126 Suppl 6:56-65. [PMID: 31219669 DOI: 10.1111/bcpt.13277] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022]
Abstract
Dopamine (DA) receptors play critical roles in a wide range of behaviours, including sensory processing, motor function, reward and arousal. As such, aberrant DA signalling is associated with numerous neurological and psychiatric disorders. Therefore, understanding the mechanisms by which DA neurotransmission drives intracellular signalling pathways that modulate behaviour can provide critical insights to guide the development of targeted therapeutics. Drosophila melanogaster has emerged as a powerful model with unique advantages to study the mechanisms underlying DA neurotransmission and associated behaviours in a controlled and systematic manner. Many regions in the fly brain innervated by dopaminergic neurons have been mapped and linked to specific behaviours, including associative learning and arousal. Here, we provide an overview of the homology between human and Drosophila dopaminergic systems and review the current literature on the pharmacology, molecular signalling mechanisms and behavioural outcome of DA receptor activation in the Drosophila brain.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York City, New York, USA
| | - Sandra K Jones
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York City, New York, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York City, New York, USA.,Department of Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York, USA
| |
Collapse
|
42
|
Castora FJ. Mitochondrial function and abnormalities implicated in the pathogenesis of ASD. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:83-108. [PMID: 30599156 DOI: 10.1016/j.pnpbp.2018.12.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 12/18/2022]
Abstract
Mitochondria are the powerhouse that generate over 90% of the ATP produced in cells. In addition to its role in energy production, the mitochondrion also plays a major role in carbohydrate, fatty acid, amino acid and nucleotide metabolism, programmed cell death (apoptosis), generation of and protection against reactive oxygen species (ROS), immune response, regulation of intracellular calcium ion levels and even maintenance of gut microbiota. With its essential role in bio-energetic as well as non-energetic biological processes, it is not surprising that proper cellular, tissue and organ function is dependent upon proper mitochondrial function. Accordingly, mitochondrial dysfunction has been shown to be directly linked to a variety of medical disorders, particularly neuromuscular disorders and increasing evidence has linked mitochondrial dysfunction to neurodegenerative and neurodevelopmental disorders such as Alzheimer's Disease (AD), Parkinson's Disease (PD), Rett Syndrome (RS) and Autism Spectrum Disorders (ASD). Over the last 40 years there has been a dramatic increase in the diagnosis of ASD and, more recently, an increasing body of evidence indicates that mitochondrial dysfunction plays an important role in ASD development. In this review, the latest evidence linking mitochondrial dysfunction and abnormalities in mitochondrial DNA (mtDNA) to the pathogenesis of autism will be presented. This review will also summarize the results of several recent `approaches used for improving mitochondrial function that may lead to new therapeutic approaches to managing and/or treating ASD.
Collapse
Affiliation(s)
- Frank J Castora
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA; Department of Neurology, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
43
|
Ri H, Lee J, Sonn JY, Yoo E, Lim C, Choe J. Drosophila CrebB is a Substrate of the Nonsense-Mediated mRNA Decay Pathway that Sustains Circadian Behaviors. Mol Cells 2019; 42:301-312. [PMID: 31091556 PMCID: PMC6530642 DOI: 10.14348/molcells.2019.2451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/23/2022] Open
Abstract
Post-transcriptional regulation underlies the circadian control of gene expression and animal behaviors. However, the role of mRNA surveillance via the nonsense-mediated mRNA decay (NMD) pathway in circadian rhythms remains elusive. Here, we report that Drosophila NMD pathway acts in a subset of circadian pacemaker neurons to maintain robust 24 h rhythms of free-running locomotor activity. RNA interference-mediated depletion of key NMD factors in timeless-expressing clock cells decreased the amplitude of circadian locomotor behaviors. Transgenic manipulation of the NMD pathway in clock neurons expressing a neuropeptide PIGMENT-DISPERSING FACTOR (PDF) was sufficient to dampen or lengthen free-running locomotor rhythms. Confocal imaging of a transgenic NMD reporter revealed that arrhythmic Clock mutants exhibited stronger NMD activity in PDF-expressing neurons than wild-type. We further found that hypomorphic mutations in Suppressor with morphogenetic effect on genitalia 5 (Smg5 ) or Smg6 impaired circadian behaviors. These NMD mutants normally developed PDF-expressing clock neurons and displayed daily oscillations in the transcript levels of core clock genes. By contrast, the loss of Smg5 or Smg6 function affected the relative transcript levels of cAMP response element-binding protein B (CrebB ) in an isoform-specific manner. Moreover, the overexpression of a transcriptional repressor form of CrebB rescued free-running locomotor rhythms in Smg5-depleted flies. These data demonstrate that CrebB is a rate-limiting substrate of the genetic NMD pathway important for the behavioral output of circadian clocks in Drosophila.
Collapse
Affiliation(s)
- Hwajung Ri
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| | - Jongbin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| | - Jun Young Sonn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| | - Eunseok Yoo
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919,
Korea
| | - Chunghun Lim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919,
Korea
| | - Joonho Choe
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| |
Collapse
|
44
|
Smolen P, Baxter DA, Byrne JH. How can memories last for days, years, or a lifetime? Proposed mechanisms for maintaining synaptic potentiation and memory. ACTA ACUST UNITED AC 2019; 26:133-150. [PMID: 30992383 PMCID: PMC6478248 DOI: 10.1101/lm.049395.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/12/2019] [Indexed: 01/24/2023]
Abstract
With memory encoding reliant on persistent changes in the properties of synapses, a key question is how can memories be maintained from days to months or a lifetime given molecular turnover? It is likely that positive feedback loops are necessary to persistently maintain the strength of synapses that participate in encoding. Such feedback may occur within signal-transduction cascades and/or the regulation of translation, and it may occur within specific subcellular compartments or within neuronal networks. Not surprisingly, numerous positive feedback loops have been proposed. Some posited loops operate at the level of biochemical signal-transduction cascades, such as persistent activation of Ca2+/calmodulin kinase II (CaMKII) or protein kinase Mζ. Another level consists of feedback loops involving transcriptional, epigenetic and translational pathways, and autocrine actions of growth factors such as BDNF. Finally, at the neuronal network level, recurrent reactivation of cell assemblies encoding memories is likely to be essential for late maintenance of memory. These levels are not isolated, but linked by shared components of feedback loops. Here, we review characteristics of some commonly discussed feedback loops proposed to underlie the maintenance of memory and long-term synaptic plasticity, assess evidence for and against their necessity, and suggest experiments that could further delineate the dynamics of these feedback loops. We also discuss crosstalk between proposed loops, and ways in which such interaction can facilitate the rapidity and robustness of memory formation and storage.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John H Byrne
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
45
|
Feldmann KG, Chowdhury A, Becker JL, McAlpin N, Ahmed T, Haider S, Richard Xia JX, Diaz K, Mehta MG, Mano I. Non-canonical activation of CREB mediates neuroprotection in a Caenorhabditis elegans model of excitotoxic necrosis. J Neurochem 2018; 148:531-549. [PMID: 30447010 DOI: 10.1111/jnc.14629] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/26/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022]
Abstract
Excitotoxicity, caused by exaggerated neuronal stimulation by Glutamate (Glu), is a major cause of neurodegeneration in brain ischemia. While we know that neurodegeneration is triggered by overstimulation of Glu-receptors (GluRs), the subsequent mechanisms that lead to cellular demise remain controversial. Surprisingly, signaling downstream of GluRs can also activate neuroprotective pathways. The strongest evidence involves activation of the transcription factor cAMP response element-binding protein (CREB), widely recognized for its importance in synaptic plasticity. Canonical views describe CREB as a phosphorylation-triggered transcription factor, where transcriptional activation involves CREB phosphorylation and association with CREB-binding protein. However, given CREB's ubiquitous cross-tissue expression, the multitude of cascades leading to CREB phosphorylation, and its ability to regulate thousands of genes, it remains unclear how CREB exerts closely tailored, differential neuroprotective responses in excitotoxicity. A non-canonical, alternative cascade for activation of CREB-mediated transcription involves the CREB co-factor cAMP-regulated transcriptional co-activator (CRTC), and may be independent of CREB phosphorylation. To identify cascades that activate CREB in excitotoxicity we used a Caenorhabditis elegans model of neurodegeneration by excitotoxic necrosis. We demonstrated that CREB's neuroprotective effect was conserved, and seemed most effective in neurons with moderate Glu exposure. We found that factors mediating canonical CREB activation were not involved. Instead, phosphorylation-independent CREB activation in nematode excitotoxic necrosis hinged on CRTC. CREB-mediated transcription that depends on CRTC, but not on CREB phosphorylation, might lead to expression of a specific subset of neuroprotective genes. Elucidating conserved mechanisms of excitotoxicity-specific CREB activation can help us focus on core neuroprotective programs in excitotoxicity. Cover Image for this issue: doi: 10.1111/jnc.14494.
Collapse
Affiliation(s)
- K Genevieve Feldmann
- Department of Molecular, Cellular and Biomedical Sciences, CDI Cluster on Neural Development and Repair, The CUNY School of Medicine, City College (CCNY), The City University of New York (CUNY), New York City, New York, USA.,The CUNY Neuroscience Collaborative PhD Program, CUNY Graduate Center, New York City, New York, USA
| | - Ayesha Chowdhury
- Department of Molecular, Cellular and Biomedical Sciences, CDI Cluster on Neural Development and Repair, The CUNY School of Medicine, City College (CCNY), The City University of New York (CUNY), New York City, New York, USA.,The CUNY Neuroscience Collaborative PhD Program, CUNY Graduate Center, New York City, New York, USA
| | - Jessica L Becker
- Undergraduate Program in Biology, CCNY, CUNY, New York City, New York, USA
| | - N'Gina McAlpin
- Undergraduate Program in Biology, CCNY, CUNY, New York City, New York, USA
| | - Taqwa Ahmed
- The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| | - Syed Haider
- Undergraduate Program in Biology, CCNY, CUNY, New York City, New York, USA
| | - Jian X Richard Xia
- The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| | - Karina Diaz
- The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| | - Monal G Mehta
- Robert Wood Johnson Medical School, Rutgers - The State University of New Jersey, Piscataway, New Jersey, USA
| | - Itzhak Mano
- Department of Molecular, Cellular and Biomedical Sciences, CDI Cluster on Neural Development and Repair, The CUNY School of Medicine, City College (CCNY), The City University of New York (CUNY), New York City, New York, USA.,The CUNY Neuroscience Collaborative PhD Program, CUNY Graduate Center, New York City, New York, USA.,The Sophie Davis BS/MD program, CUNY School of Medicine, New York City, New York, USA
| |
Collapse
|
46
|
Miyashita T, Kikuchi E, Horiuchi J, Saitoe M. Long-Term Memory Engram Cells Are Established by c-Fos/CREB Transcriptional Cycling. Cell Rep 2018; 25:2716-2728.e3. [DOI: 10.1016/j.celrep.2018.11.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/13/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
|
47
|
Savelieff MG, Nam G, Kang J, Lee HJ, Lee M, Lim MH. Development of Multifunctional Molecules as Potential Therapeutic Candidates for Alzheimer’s Disease, Parkinson’s Disease, and Amyotrophic Lateral Sclerosis in the Last Decade. Chem Rev 2018; 119:1221-1322. [DOI: 10.1021/acs.chemrev.8b00138] [Citation(s) in RCA: 270] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Masha G. Savelieff
- SciGency Science Communications, Ann Arbor, Michigan 48104, United States
| | - Geewoo Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Juhye Kang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Misun Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
48
|
Karri V, Ramos D, Martinez JB, Odena A, Oliveira E, Coort SL, Evelo CT, Mariman ECM, Schuhmacher M, Kumar V. Differential protein expression of hippocampal cells associated with heavy metals (Pb, As, and MeHg) neurotoxicity: Deepening into the molecular mechanism of neurodegenerative diseases. J Proteomics 2018; 187:106-125. [PMID: 30017948 DOI: 10.1016/j.jprot.2018.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 12/29/2022]
Abstract
Chronic exposure to heavy metals such as Pb, As, and MeHg can be associated with an increased risk of developing neurodegenerative diseases. Our in vitro bioassays results showed the potency of heavy metals in the order of Pb < As < MeHg on hippocampal cells. The main objective of this study was combining in vitro label free proteomics and systems biology approach for elucidating patterns of biological response, discovering underlying mechanisms of Pb, As, and MeHg toxicity in hippocampal cells. The omics data was refined by using different filters and normalization and multilevel analysis tools were employed to explore the data visualization. The functional and pathway visualization was performed by using Gene ontology and PathVisio tools. Using these all integrated approaches, we identified significant proteins across treatments within the mitochondrial dysfunction, oxidative stress, ubiquitin proteome dysfunction, and mRNA splicing related to neurodegenerative diseases. The systems biology analysis revealed significant alterations in proteins implicated in Parkinson's disease (PD) and Alzheimer's disease (AD). The current proteomics analysis of three metals support the insight into the proteins involved in neurodegeneration and the altered proteins can be useful for metal-specific biomarkers of exposure and its adverse effects. SIGNIFICANCE The proteomics techniques have been claimed to be more sensitive than the conventional toxicological assays, facilitating the measurement of responses to heavy metals (Pb, As, and MeHg) exposure before obvious harm has occurred demonstrating their predictive value. Also, proteomics allows for the comparison of responses between Pb, As, and MeHg metals, permitting the evaluation of potency differences hippocampal cells of the brain. Hereby, the molecular information provided by pathway and gene functional analysis can be used to develop a more thorough understanding of each metal mechanism at the protein level for different neurological adverse outcomes (e.g. Parkinson's disease, Alzheimer's diseases). Efforts are put into developing proteomics based toxicity testing methods using in vitro models for improving human risk assessment. Some of the key proteins identified can also potentially be used as biomarkers in epidemiologic studies. These heavy metal response patterns shed new light on the mechanisms of mRNA splicing, ubiquitin pathway role in neurodegeneration, and can be useful for the development of molecular biomarkers of heavy metals exposure.
Collapse
Affiliation(s)
- Venkatanaidu Karri
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - David Ramos
- Plataforma de Proteòmica, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Julia Bauzá Martinez
- Plataforma de Proteòmica, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Antonia Odena
- Plataforma de Proteòmica, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Eliandre Oliveira
- Unidad de Toxicologia, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Susan L Coort
- Department of Bioinformatics, BiGCaT, NUTRIM, Maastricht University, 6229, ER, Maastricht, the Netherlands
| | - Chris T Evelo
- Department of Bioinformatics, BiGCaT, NUTRIM, Maastricht University, 6229, ER, Maastricht, the Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Marta Schuhmacher
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - Vikas Kumar
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007 Tarragona, Spain; IISPV, Hospital Universitari Sant Joan de Reus, Universitat Rovira I Virgili, Reus, Spain.
| |
Collapse
|
49
|
Pak VM, Mazzotti DR, Keenan BT, Hirotsu C, Gehrman P, Bittencourt L, Pack AI, Tufik S. Candidate gene analysis in the São Paulo Epidemiologic Sleep Study (EPISONO) shows an association of variant in PDE4D and sleepiness. Sleep Med 2018; 47:106-112. [DOI: 10.1016/j.sleep.2017.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/30/2017] [Indexed: 12/24/2022]
|
50
|
Matsumoto Y, Matsumoto CS, Mizunami M. Signaling Pathways for Long-Term Memory Formation in the Cricket. Front Psychol 2018; 9:1014. [PMID: 29988479 PMCID: PMC6024501 DOI: 10.3389/fpsyg.2018.01014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/31/2018] [Indexed: 11/13/2022] Open
Abstract
Unraveling the molecular mechanisms underlying memory formation in insects and a comparison with those of mammals will contribute to a further understanding of the evolution of higher-brain functions. As it is for mammals, insect memory can be divided into at least two distinct phases: protein-independent short-term memory and protein-dependent long-term memory (LTM). We have been investigating the signaling pathway of LTM formation by behavioral-pharmacological experiments using the cricket Gryllus bimaculatus, whose olfactory learning and memory abilities are among the highest in insect species. Our studies revealed that the NO-cGMP signaling pathway, CaMKII and PKA play crucial roles in LTM formation in crickets. These LTM formation signaling pathways in crickets share a number of attributes with those of mammals, and thus we conclude that insects, with relatively simple brain structures and neural circuitry, will also be beneficial in exploratory experiments to predict the molecular mechanisms underlying memory formation in mammals.
Collapse
Affiliation(s)
- Yukihisa Matsumoto
- College of Liberal Arts and Sciences, Tokyo Medical and Dental University, Ichikawa, Japan
| | - Chihiro S Matsumoto
- College of Liberal Arts and Sciences, Tokyo Medical and Dental University, Ichikawa, Japan
| | - Makoto Mizunami
- Graduate School of Life Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|