51
|
Ko SY, Frankland PW. Neurogenesis-dependent transformation of hippocampal engrams. Neurosci Lett 2021; 762:136176. [PMID: 34400284 DOI: 10.1016/j.neulet.2021.136176] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/29/2021] [Accepted: 08/11/2021] [Indexed: 11/29/2022]
Abstract
In humans and other mammals, memories of events are encoded by neuronal ensembles (or engrams) in the hippocampus. The mnemonic information stored in these engrams can then be used to guide future behavior, including prediction- and decision-making in dynamic environments. While some hippocampal engrams may be persistently stored, others are modified over time, suggesting that the represented memories may also be transformed. How might hippocampal engrams be modified through time? Adult hippocampal neurogenesis represents one process that continuously rewires hippocampal circuitry, presumably including stored hippocampal engrams. At intermediate stages, we propose that neurogenesis-mediated rewiring of hippocampal engram circuitry induces forgetting of specific stimulus attributes, and this less precise engram allows for generalization. At more advanced stages, we propose that neurogenesis-mediated rewiring of hippocampal engram circuitry leads to silencing of hippocampal engrams, rendering them no longer accessible by natural retrieval cues.
Collapse
Affiliation(s)
- Sangyoon Y Ko
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Temerty Centre for AI Research and Education in Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Paul W Frankland
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada; Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
52
|
A Runner's High for New Neurons? Potential Role for Endorphins in Exercise Effects on Adult Neurogenesis. Biomolecules 2021; 11:biom11081077. [PMID: 34439743 PMCID: PMC8392752 DOI: 10.3390/biom11081077] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/30/2022] Open
Abstract
Physical exercise has wide-ranging benefits to cognitive functioning and mental state, effects very closely resembling enhancements to hippocampal functioning. Hippocampal neurogenesis has been implicated in many of these mental benefits of exercise. However, precise mechanisms behind these effects are not well known. Released peripherally during exercise, beta-endorphins are an intriguing candidate for moderating increases in neurogenesis and the related behavioral benefits of exercise. Although historically ignored due to their peripheral release and status as a peptide hormone, this review highlights reasons for further exploring beta-endorphin as a key mediator of hippocampal neurogenesis. This includes possible routes for beta-endorphin signaling into the hippocampus during exercise, direct effects of beta-endorphin on cell proliferation and neurogenesis, and behavioral effects of manipulating endogenous opioid signaling. Together, beta-endorphin appears to be a promising mechanism for understanding the specific ways that exercise promotes adult neurogenesis specifically and brain health broadly.
Collapse
|
53
|
Sep MSC, Vellinga M, Sarabdjitsingh RA, Joëls M. The rodent object-in-context task: A systematic review and meta-analysis of important variables. PLoS One 2021; 16:e0249102. [PMID: 34270575 PMCID: PMC8284613 DOI: 10.1371/journal.pone.0249102] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
Environmental information plays an important role in remembering events. Information about stable aspects of the environment (here referred to as 'context') and the event are combined by the hippocampal system and stored as context-dependent memory. In rodents (such as rats and mice), context-dependent memory is often investigated with the object-in-context task. However, the implementation and interpretation of this task varies considerably across studies. This variation hampers the comparison between studies and-for those who design a new experiment or carry out pilot experiments-the estimation of whether observed behavior is within the expected range. Also, it is currently unclear which of the variables critically influence the outcome of the task. To address these issues, we carried out a preregistered systematic review (PROSPERO CRD42020191340) and provide an up-to-date overview of the animal-, task-, and protocol-related variations in the object-in-context task for rodents. Using a data-driven explorative meta-analysis we next identified critical factors influencing the outcome of this task, such as sex, testbox size and the delay between the learning trials. Based on these observations we provide recommendations on sex, strain, prior arousal, context (size, walls, shape, etc.) and timing (habituation, learning, and memory phase) to create more consensus in the set-up, procedure, and interpretation of the object-in-context task for rodents. This could contribute to a more robust and evidence-based design in future animal experiments.
Collapse
Affiliation(s)
- Milou S. C. Sep
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Brain Research and Innovation Centre, Ministry of Defence, Utrecht, The Netherlands
| | - Marijn Vellinga
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - R. Angela Sarabdjitsingh
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
54
|
Che X, Bai Y, Cai J, Liu Y, Li Y, Yin M, Xu T, Wu C, Yang J. Hippocampal neurogenesis interferes with extinction and reinstatement of methamphetamine-associated reward memory in mice. Neuropharmacology 2021; 196:108717. [PMID: 34273388 DOI: 10.1016/j.neuropharm.2021.108717] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/30/2021] [Accepted: 07/11/2021] [Indexed: 12/16/2022]
Abstract
Drugs of abuse, including morphine and cocaine, can reduce hippocampal neurogenesis (HN). Whereas promotion of HN is being increasingly recognized as a promising strategy for treating morphine and cocaine addiction. The present study is focused on exploring the changes of HN during methamphetamine (METH) administration and further clarify if HN is involved in METH-associated reward memory. After successfully establishing the conditioned place preference (CPP) paradigm to simulate the METH-associated reward memory in C57BL/6 mice, we observed that HN was significantly inhibited during METH (2 mg/kg, i. p.) administration and returned to normal after the extinction of METH CPP, as indicated by the immunostaining of bromodeoxyuridine (BrdU) and doublecortin (DCX) in the hippocampus. To promote/inhibit HN levels, 7,8-dihydroxyflavone (DHF), a small tyrosine kinase receptor B (TrkB) agonist and temozolomide (TMZ), an alkylating agent, were administered intraperitoneally (i.p.), respectively. The data showed that either DHF (5 mg/kg, i. p.) or TMZ (25 mg/kg, i. p.) pre-treatment before METH administration could significantly prolong extinction and enhance reinstatement of the reward memory. Notably, DHF treatment after METH administration significantly facilitated extinction and inhibited METH reinstatement, while TMZ treatment resulted in opposite effects. The present study indicated that METH administration could induce a temporal inhibitory effect on HN. More importantly, promotion of HN after the acquisition of METH-associated reward memory, but not inhibition of HN or promotion of HN before the acquisition of reward memory, could facilitate METH extinction and inhibit METH reinstatement, indicating the beneficial effect of HN on METH addiction by erasing the according reward memory.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yijun Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yuting Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Meixue Yin
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Tianyu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|
55
|
Meng F, Li Y, Sun H, Li C, Li Q, Law PY, Loh HH, Liang L, Zheng H. Naloxone Facilitates Contextual Learning and Memory in a Receptor-Independent and Tet1-Dependent Manner. Cell Mol Neurobiol 2021; 41:1031-1038. [PMID: 32989585 PMCID: PMC11448548 DOI: 10.1007/s10571-020-00970-8] [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: 05/28/2020] [Accepted: 09/23/2020] [Indexed: 11/30/2022]
Abstract
Opioids, like morphine and naloxone, regulate the proliferation and neuronal differentiation of neural stem cells (NSCs) in a receptor-independent and ten-eleven translocation methylcytosine dioxygenase (TET1)-dependent manner in vitro. Whether naloxone regulates hippocampal NSCs and contextual learning in vivo in a similar manner was determined. Naloxone infusion increased the Ki67 and Doublecortin positive cells in subgranular zone of wild type mice, which suggested the increased proliferation and differentiation of hippocampal NSCs in vivo and was consistent with the in vitro functions of naloxone. In addition, naloxone infusion also facilitated the contextual learning and memory of wild type mice. To determine the contribution of μ-opioid receptor (OPRM1) and TET1 to these functions of naloxone, several types of knockout mice were used. Since Tet1-/- mice have high deficiency in contextual learning and memory, Tet1+/- mice were used instead. The abilities of naloxone to regulate NSCs and to facilitate contextual learning were significantly impaired in Tet1+/- mice. In addition, these abilities of naloxone were not affected in Oprm1-/- mice. Therefore, naloxone facilitates contextual learning and memory in a receptor-independent and Tet1-dependent manner, which provides new understanding on the receptor-independent functions of opioids.
Collapse
Affiliation(s)
- Fei Meng
- University of Science and Technology of China, Hefei, 230026, Anhui, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Ave., Science City, Guangzhou, 510530, China
| | - Yuan Li
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Ave., Science City, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, 510530, China
| | - Hao Sun
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Ave., Science City, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, 510530, China
| | - Changpeng Li
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Ave., Science City, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, 510530, China
| | - Qian Li
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Ave., Science City, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, 510530, China
- Guangzhou Medical University, Guangzhou, 511436, China
| | - Ping-Yee Law
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Horace H Loh
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
| | - Lining Liang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Ave., Science City, Guangzhou, 510530, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, 510530, China.
| | - Hui Zheng
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Ave., Science City, Guangzhou, 510530, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, 510530, China.
- Institutes for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
56
|
Sefiani A, Geoffroy CG. The Potential Role of Inflammation in Modulating Endogenous Hippocampal Neurogenesis After Spinal Cord Injury. Front Neurosci 2021; 15:682259. [PMID: 34220440 PMCID: PMC8249862 DOI: 10.3389/fnins.2021.682259] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Currently there are approximately 291,000 people suffering from a spinal cord injury (SCI) in the United States. SCI is associated with traumatic changes in mobility and neuralgia, as well as many other long-term chronic health complications, including metabolic disorders, diabetes mellitus, non-alcoholic steatohepatitis, osteoporosis, and elevated inflammatory markers. Due to medical advances, patients with SCI survive much longer than previously. This increase in life expectancy exposes them to novel neurological complications such as memory loss, cognitive decline, depression, and Alzheimer's disease. In fact, these usually age-associated disorders are more prevalent in people living with SCI. A common factor of these disorders is the reduction in hippocampal neurogenesis. Inflammation, which is elevated after SCI, plays a major role in modulating hippocampal neurogenesis. While there is no clear consensus on the mechanism of the decline in hippocampal neurogenesis and cognition after SCI, we will examine in this review how SCI-induced inflammation could modulate hippocampal neurogenesis and provoke age-associated neurological disorders. Thereafter, we will discuss possible therapeutic options which may mitigate the influence of SCI associated complications on hippocampal neurogenesis.
Collapse
|
57
|
You J, Sun L, Wang J, Sun F, Wang W, Wang D, Fan X, Liu D, Xu Z, Qiu C, Chen J, Yan H, Liu B. Role of Adiponectin-Notch pathway in cognitive dysfunction associated with depression and in the therapeutic effect of physical exercise. Aging Cell 2021; 20:e13387. [PMID: 34053165 PMCID: PMC8208781 DOI: 10.1111/acel.13387] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/22/2021] [Accepted: 05/08/2021] [Indexed: 12/17/2022] Open
Abstract
A substantial percentage of late‐life depression patients also have an cognitive impairment, which severely affects the life quality, while the co‐occurring mechanisms are still unclear. Physical exercise can ameliorate both depressive behaviors and cognitive dysfunction, but the molecular mechanisms underlying its beneficial effects remain elusive. In this study, we uncover a novel adipose tissue to hippocampus crosstalk mediated by Adiponectin‐Notch pathway, with an impact on hippocampal neurogenesis and cognitive function. Adiponectin, an adipocyte‐derived hormone, could activate Notch signaling in the hippocampus through upregulating ADAM10 and Notch1, two key molecules in the Notch signaling. Chronic stress inhibits the Adiponectin‐Notch pathway and induces impaired hippocampal neurogenesis and cognitive dysfunction, which can be rescued by AdipoRon and running. Inhibition Notch signaling by DAPT mimics the adverse effects of chronic stress on hippocampal neurogenesis and cognitive function. Adiponectin knockout mice display depressive‐like behaviors, associated with inhibited Notch signaling, impaired hippocampal neurogenesis and cognitive dysfunction. Physical exercise could activate Adiponectin‐Notch pathway, and improve hippocampal neurogenesis and cognitive function, while deleting adiponectin gene or inhibiting Notch signaling blocks its beneficial effects. Together, our data not only suggest that Adiponectin‐Notch pathway is involved in the pathogenesis of cognitive dysfunction associated with depression, but also contributes to the therapeutic effect of physical exercise. This work helps to decipher the etiology of cognitive impairment associated with depression and hence will provide a potential innovative therapeutic target for these patients.
Collapse
Affiliation(s)
- Jingjing You
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Linshan Sun
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Jiangong Wang
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Fengjiao Sun
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Wentao Wang
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Dan Wang
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Xueli Fan
- Department of Neurology Binzhou Medical University Hospital Shandong China
| | - Dunjiang Liu
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Zhicheng Xu
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Changyun Qiu
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| | - Jinbo Chen
- Department of Neurology Binzhou Medical University Hospital Shandong China
| | - Haijing Yan
- Department of Pharmacology College of Basic Medicine Binzhou Medical University Yantai China
| | - Bin Liu
- Institute for Metabolic & Neuropsychiatric Disorders Binzhou Medical University Hospital Shandong China
| |
Collapse
|
58
|
Rudolph M, Schmeer CW, Günther M, Woitke F, Kathner-Schaffert C, Karapetow L, Lindner J, Lehmann T, Jirikowski G, Witte OW, Redecker C, Keiner S. Microglia-mediated phagocytosis of apoptotic nuclei is impaired in the adult murine hippocampus after stroke. Glia 2021; 69:2006-2022. [PMID: 33942391 DOI: 10.1002/glia.24009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 01/02/2023]
Abstract
Following stroke, neuronal death takes place both in the infarct region and in brain areas distal to the lesion site including the hippocampus. The hippocampus is critically involved in learning and memory processes and continuously generates new neurons. Dysregulation of adult neurogenesis may be associated with cognitive decline after a stroke lesion. In particular, proliferation of precursor cells and the formation of new neurons are increased after lesion. Within the first week, many new precursor cells die during development. How dying precursors are removed from the hippocampus and to what extent phagocytosis takes place after stroke is still not clear. Here, we evaluated the effect of a prefrontal stroke lesion on the phagocytic activity of microglia in the dentate gyrus (DG) of the hippocampus. Three-months-old C57BL/6J mice were injected once with the proliferation marker BrdU (250 mg/kg) 6 hr after a middle cerebral artery occlusion or sham surgery. The number of apoptotic cells and the phagocytic capacity of the microglia were evaluated by means of immunohistochemistry, confocal microscopy, and 3D-reconstructions. We found a transient but significant increase in the number of apoptotic cells in the DG early after stroke, associated with impaired removal by microglia. Interestingly, phagocytosis of newly generated precursor cells was not affected. Our study shows that a prefrontal stroke lesion affects phagocytosis of apoptotic cells in the DG, a region distal to the lesion core. Whether disturbed phagocytosis might contribute to inflammatory- and maladaptive processes including cognitive impairment following stroke needs to be further investigated.
Collapse
Affiliation(s)
- Max Rudolph
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christian W Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Madlen Günther
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Florus Woitke
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - Lina Karapetow
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Julia Lindner
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Thomas Lehmann
- Institute of Medical Statistics and Computer Science, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Gustav Jirikowski
- Health and Medical University Potsdam, University Potsdam, Potsdam, Germany
| | - Otto W Witte
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christoph Redecker
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Department of Neurology, Lippe General Hospital, Lemgo, Germany
| | - Silke Keiner
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
59
|
Lauretta G, Ravalli S, Maugeri G, D'Agata V, Rosa MD, Musumeci G. The impact of physical exercise on hippocampus, in physiological condition and ageing-related decline: current evidence from animal and human studies. Curr Pharm Biotechnol 2021; 23:180-189. [PMID: 33820516 DOI: 10.2174/1389201022666210405142611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/19/2021] [Accepted: 02/14/2021] [Indexed: 11/22/2022]
Abstract
Physical exercise (PE), notoriously, promotes a state of general well-being, throughout the entire human lifespan. Moreover, maintaining an adequate and regular PE habit results to be a powerful preventive factor towards many diseases and may also help in managing existing pathological conditions. PE induces structural and functional changes in various districts of the body, determining biological and psychological benefits. Additionally, in elderly, PE might represent a remarkable tool reducing cognitive impairments related to the normal aging processes and it has also been found to have an impact in neurodegenerative diseases such as Alzheimer's disease. The present review aims to provide an overview about PE effects on hippocampus, since it is one of the brain regions most susceptible to aging and, therefore, involved in diseases characterized by cognitive impairment.
Collapse
Affiliation(s)
- Giovanni Lauretta
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Silvia Ravalli
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Velia D'Agata
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| |
Collapse
|
60
|
Tracy JI, Chaudhary K, Modi S, Crow A, Kumar A, Weinstein D, Sperling MR. Computational support, not primacy, distinguishes compensatory memory reorganization in epilepsy. Brain Commun 2021; 3:fcab025. [PMID: 34222865 PMCID: PMC8244645 DOI: 10.1093/braincomms/fcab025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/12/2020] [Accepted: 01/04/2021] [Indexed: 02/03/2023] Open
Abstract
Temporal lobe epilepsy is associated with impairment in episodic memory. A substantial subgroup, however, is able to maintain adequate memory despite temporal lobe pathology. Missing from prior work in cognitive reorganization is a direct comparison of temporal lobe epilepsy patients with intact status with those who are memory impaired. Little is known about the regional activations, functional connectivities and/or network reconfigurations that implement changes in primary computations or support functions that drive adaptive plasticity and compensated memory. We utilized task functional MRI on 54 unilateral temporal lobe epilepsy patients and 24 matched healthy controls during the performance of a paired-associate memory task to address three questions: (i) which regions implement paired-associate memory in temporal lobe epilepsy, and do they vary as a function of good versus poor performance, (ii) is there unique functional connectivity present during memory encoding that accounts for intact status by preservation of primary memory computations or the supportive computations that allow for intact memory responses and (iii) what features during memory encoding are most distinctive: is it the magnitude and location of regional activations, or the presence of enhanced functional connections to key structures such as the hippocampus? The study revealed non-dominant hemisphere regions (right posterior temporal regions) involving both increased regional activity and increased modulatory communication with the hippocampi as most important to intact memory in left temporal lobe epilepsy compared to impaired status. The profile involved areas that are neither contralateral homologues to left hemisphere memory areas, nor regions traditionally considered computationally primary for episodic memory. None of these areas of increased activation or functional connectivity were associated with advantaged memory in healthy controls. Our emphasis on different performance levels yielded insight into two forms of cognitive reorganization: computational primacy, where left temporal lobe epilepsy showed little change relative to healthy controls, and computational support where intact left temporal lobe epilepsy patients showed adaptive abnormalities. The analyses isolated the unique regional activations and mediating functional connectivity that implements truly compensatory reorganization in left temporal lobe epilepsy. The results provided a new perspective on memory deficits by making clear that they arise not just from the knockout of a functional hub, but from the failure to instantiate a complex set of reorganization responses. Such responses provided the computational support to ensure successful memory. We demonstrated that by keeping track of performance levels, we can increase understanding of adaptive brain responses and neuroplasticity in epilepsy.
Collapse
Affiliation(s)
- Joseph I Tracy
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA,Correspondence to: Joseph I. Tracy, Department of Neurology, Thomas Jefferson University, 901 Walnut Street, Health Sciences Building, Suite 447, Philadelphia, PA 19107, USA. E-mail:
| | - Kapil Chaudhary
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shilpi Modi
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew Crow
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ashith Kumar
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - David Weinstein
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Michael R Sperling
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
61
|
Stojanovic T, Benes H, Awad A, Bormann D, Monje FJ. Nicotine abolishes memory-related synaptic strengthening and promotes synaptic depression in the neurogenic dentate gyrus of miR-132/212 knockout mice. Addict Biol 2021; 26:e12905. [PMID: 32293776 PMCID: PMC7988623 DOI: 10.1111/adb.12905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 12/25/2022]
Abstract
Micro-RNAs (miRNAs) are highly evolutionarily conserved short-length/noncoding RNA molecules that modulate a wide range of cellular functions in many cell types by regulating the expression of a variety of targeted genes. miRNAs have also recently emerged as key regulators of neuronal genes mediating the effects of psychostimulant drugs and memory-related neuroplasticity processes. Smoking is a predominant addictive behaviour associated with millions of deaths worldwide, and nicotine is a potent natural psychoactive agonist of cholinergic receptors, highly abundant in cigarettes. The influence of miRNAs modulation on cholinergic signalling in the nervous system remains however poorly explored. Using miRNA knockout mice and biochemical, electrophysiological and pharmacological approaches, we examined the effects of miR-132/212 gene disruption on the levels of hippocampal nicotinic acetylcholine receptors, total ERK and phosphorylated ERK (pERK) and MeCP2 protein levels, and studied the impact of nicotine stimulation on hippocampal synaptic transmission and synaptic depression and strengthening. miR-132/212 deletion significantly altered α7-nAChR and pERK protein levels, but not total ERK or MeCP2, and resulted in both exacerbated synaptic depression and virtually abolished memory-related synaptic strengthening upon nicotine stimulation. These observations reveal a functional miRNAs/nicotinergic signalling interplay critical for nicotinic-receptor expression and neuroplasticity in brain structures relevant for drug addiction and learning and memory functions.
Collapse
Affiliation(s)
- Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Hannah Benes
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Amena Awad
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Daniel Bormann
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and NeuropharmacologyMedical University of ViennaViennaAustria
| |
Collapse
|
62
|
Reset of hippocampal-prefrontal circuitry facilitates learning. Nature 2021; 591:615-619. [PMID: 33627872 PMCID: PMC7990705 DOI: 10.1038/s41586-021-03272-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 01/20/2021] [Indexed: 01/31/2023]
Abstract
The ability to rapidly adapt to novel situations is essential for survival, and this flexibility is impaired in many neuropsychiatric disorders1. Thus, understanding whether and how novelty prepares, or primes, brain circuitry to facilitate cognitive flexibility has important translational relevance. Exposure to novelty recruits the hippocampus and medial prefrontal cortex (mPFC)2 and may prime hippocampal-prefrontal circuitry for subsequent learning-associated plasticity. Here we show that novelty resets the neural circuits that link the ventral hippocampus (vHPC) and the mPFC, facilitating the ability to overcome an established strategy. Exposing mice to novelty disrupted a previously encoded strategy by reorganizing vHPC activity to local theta (4-12 Hz) oscillations and weakening existing vHPC-mPFC connectivity. As mice subsequently adapted to a new task, vHPC neurons developed new task-associated activity, vHPC-mPFC connectivity was strengthened, and mPFC neurons updated to encode the new rules. Without novelty, however, mice adhered to their established strategy. Blocking dopamine D1 receptors (D1Rs) or inhibiting novelty-tagged cells that express D1Rs in the vHPC prevented these behavioural and physiological effects of novelty. Furthermore, activation of D1Rs mimicked the effects of novelty. These results suggest that novelty promotes adaptive learning by D1R-mediated resetting of vHPC-mPFC circuitry, thereby enabling subsequent learning-associated circuit plasticity.
Collapse
|
63
|
Otsuka T, Hori H, Yoshida F, Itoh M, Lin M, Niwa M, Ino K, Imai R, Ogawa S, Matsui M, Kamo T, Kunugi H, Kim Y. Association of CRP genetic variation with symptomatology, cognitive function, and circulating proinflammatory markers in civilian women with PTSD. J Affect Disord 2021; 279:640-649. [PMID: 33190115 DOI: 10.1016/j.jad.2020.10.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/08/2020] [Accepted: 10/25/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Posttraumatic stress disorder (PTSD) has been associated with increased inflammation. C-reactive protein (CRP) is a marker of systemic inflammation, and recently, single nucleotide polymorphisms (SNPs) in the CRP gene have been associated with increased blood CRP protein levels and illness severity in PTSD patients. However, the mechanism by which the CRP SNPs are involved in PTSD remains unclear. Here we investigated the association of CRP genetic variation with blood proinflammatory protein levels, symptomatology, and cognitive function, and further explored the moderating effect of childhood maltreatment history, in adult patients with PTSD. METHODS Fifty-seven Japanese civilian women with PTSD and 73 healthy control women were enrolled. Three SNPs in the CRP gene, namely rs2794520, rs1130864, and rs3093059, were genotyped, and analyses focused on rs2794520 (T/C). Serum levels of high-sensitivity CRP (hsCRP), high-sensitivity tumor necrosis factor-α (hsTNF-α), and interleukin-6 were measured. PTSD symptoms were evaluated by the Posttraumatic Diagnostic Scale. Cognitive function was assessed by the Repeatable Battery for the Assessment of Neuropsychological Status. Childhood maltreatment history was assessed by the Childhood Trauma Questionnaire. RESULTS Patients with the rs2794520 CC/CT genotype, compared to those with the TT genotype, showed significantly higher levels of hsCRP (p=0.009) and hsTNF-α (p=0.001), more severe PTSD symptoms (p=0.036), and poorer cognitive function (p=0.018). A two-way analysis of variance revealed a significant genotype-by-maltreatment interaction for more severe PTSD avoidance symptom (p=0.012). LIMITATIONS The relatively small sample size limited our findings. CONCLUSIONS These findings may provide an insight into the etiology of PTSD from the inflammatory perspective.
Collapse
Affiliation(s)
- Takeshi Otsuka
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroaki Hori
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan.
| | - Fuyuko Yoshida
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Mariko Itoh
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Mingming Lin
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Madoka Niwa
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Keiko Ino
- Department of Psychiatry and Cognitive-Behavioral Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Risa Imai
- Department of Psychiatry and Cognitive-Behavioral Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sei Ogawa
- Department of Psychiatry and Cognitive-Behavioral Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Mie Matsui
- Department of Clinical Cognitive Neuroscience, Institute of Liberal Arts and Science, Kanazawa University, Kanazawa, Japan
| | - Toshiko Kamo
- Wakamatsu-cho Mental and Skin Clinic, Tokyo, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoshiharu Kim
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
64
|
Ishimoto T, Kato Y. Regulation of Neurogenesis by Organic Cation Transporters: Potential Therapeutic Implications. Handb Exp Pharmacol 2021; 266:281-300. [PMID: 33782772 DOI: 10.1007/164_2021_445] [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] [Indexed: 02/03/2023]
Abstract
Neurogenesis is the process by which new neurons are generated from neural stem cells (NSCs), which are cells that have the ability to proliferate and differentiate into neurons, astrocytes, and oligodendrocytes. The process is essential for homeostatic tissue regeneration and the coordination of neural plasticity throughout life, as neurons cannot regenerate once injured. Therefore, defects in neurogenesis are related to the onset and exacerbation of several neuropsychiatric disorders, and therefore, the regulation of neurogenesis is considered to be a novel strategy for treatment. Neurogenesis is regulated not only by NSCs themselves, but also by the functional microenvironment surrounding the NSCs, known as the "neurogenic niche." The neurogenic niche consists of several types of neural cells, including neurons, glial cells, and vascular cells. To allow communication with these cells, transporters may be involved in the secretion and uptake of substrates that are essential for signal transduction. This chapter will focus on the involvement of polyspecific solute carriers transporting organic cations in the possible regulation of neurogenesis by controlling the concentration of several organic cation substrates in NSCs and the neurogenic niche. The potential therapeutic implications of neurogenesis regulation by these transporters will also be discussed.
Collapse
Affiliation(s)
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
65
|
Blackmore DG, Turpin F, Palliyaguru T, Evans HT, Chicoteau A, Lee W, Pelekanos M, Nguyen N, Song J, Sullivan RKP, Sah P, Bartlett PF, Götz J. Low-intensity ultrasound restores long-term potentiation and memory in senescent mice through pleiotropic mechanisms including NMDAR signaling. Mol Psychiatry 2021; 26:6975-6991. [PMID: 34040151 PMCID: PMC8760044 DOI: 10.1038/s41380-021-01129-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/31/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022]
Abstract
Advanced physiological aging is associated with impaired cognitive performance and the inability to induce long-term potentiation (LTP), an electrophysiological correlate of memory. Here, we demonstrate in the physiologically aged, senescent mouse brain that scanning ultrasound combined with microbubbles (SUS+MB), by transiently opening the blood-brain barrier, fully restores LTP induction in the dentate gyrus of the hippocampus. Intriguingly, SUS treatment without microbubbles (SUSonly), i.e., without the uptake of blood-borne factors, proved even more effective, not only restoring LTP, but also ameliorating the spatial learning deficits of the aged mice. This functional improvement is accompanied by an altered milieu of the aged hippocampus, including a lower density of perineuronal nets, increased neurogenesis, and synaptic signaling, which collectively results in improved spatial learning. We therefore conclude that therapeutic ultrasound is a non-invasive, pleiotropic modality that may enhance cognition in elderly humans.
Collapse
Affiliation(s)
- Daniel G. Blackmore
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Fabrice Turpin
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Tishila Palliyaguru
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Harrison T. Evans
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Antony Chicoteau
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Wendy Lee
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Matthew Pelekanos
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Nghia Nguyen
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Jae Song
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Robert K. P. Sullivan
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Pankaj Sah
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.263817.90000 0004 1773 1790Joint Center for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province P. R. China
| | - Perry F. Bartlett
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.263817.90000 0004 1773 1790Joint Center for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province P. R. China
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
66
|
Lam J, Lee J, Liu CY, Lozano AM, Lee DJ. Deep Brain Stimulation for Alzheimer's Disease: Tackling Circuit Dysfunction. Neuromodulation 2020; 24:171-186. [PMID: 33377280 DOI: 10.1111/ner.13305] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/07/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Treatments for Alzheimer's disease are urgently needed given its enormous human and economic costs and disappointing results of clinical trials targeting the primary amyloid and tau pathology. On the other hand, deep brain stimulation (DBS) has demonstrated success in other neurological and psychiatric disorders leading to great interest in DBS as a treatment for Alzheimer's disease. MATERIALS AND METHODS We review the literature on 1) circuit dysfunction in Alzheimer's disease and 2) DBS for Alzheimer's disease. Human and animal studies are reviewed individually. RESULTS There is accumulating evidence of neural circuit dysfunction at the structural, functional, electrophysiological, and neurotransmitter level. Recent evidence from humans and animals indicate that DBS has the potential to restore circuit dysfunction in Alzheimer's disease, similarly to other movement and psychiatric disorders, and may even slow or reverse the underlying disease pathophysiology. CONCLUSIONS DBS is an intriguing potential treatment for Alzheimer's disease, targeting circuit dysfunction as a novel therapeutic target. However, further exploration of the basic disease pathology and underlying mechanisms of DBS is necessary to better understand how circuit dysfunction can be restored. Additionally, robust clinical data in the form of ongoing phase III clinical trials are needed to validate the efficacy of DBS as a viable treatment.
Collapse
Affiliation(s)
- Jordan Lam
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| | - Justin Lee
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| | - Charles Y Liu
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| | - Andres M Lozano
- Division of Neurological Surgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Darrin J Lee
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA.,Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, 90033, USA
| |
Collapse
|
67
|
Mau W, Hasselmo ME, Cai DJ. The brain in motion: How ensemble fluidity drives memory-updating and flexibility. eLife 2020; 9:e63550. [PMID: 33372892 PMCID: PMC7771967 DOI: 10.7554/elife.63550] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/12/2020] [Indexed: 12/18/2022] Open
Abstract
While memories are often thought of as flashbacks to a previous experience, they do not simply conserve veridical representations of the past but must continually integrate new information to ensure survival in dynamic environments. Therefore, 'drift' in neural firing patterns, typically construed as disruptive 'instability' or an undesirable consequence of noise, may actually be useful for updating memories. In our view, continual modifications in memory representations reconcile classical theories of stable memory traces with neural drift. Here we review how memory representations are updated through dynamic recruitment of neuronal ensembles on the basis of excitability and functional connectivity at the time of learning. Overall, we emphasize the importance of considering memories not as static entities, but instead as flexible network states that reactivate and evolve across time and experience.
Collapse
Affiliation(s)
- William Mau
- Neuroscience Department, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | | | - Denise J Cai
- Neuroscience Department, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| |
Collapse
|
68
|
Ji S, Wu H, Ding X, Chen Q, Jin X, Yu J, Yang M. Increased hippocampal TrkA expression ameliorates cranial radiation‑induced neurogenesis impairment and cognitive deficit via PI3K/AKT signaling. Oncol Rep 2020; 44:2527-2536. [PMID: 33125501 PMCID: PMC7640353 DOI: 10.3892/or.2020.7782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 09/01/2020] [Indexed: 01/13/2023] Open
Abstract
Cognitive deficit is one of the most serious complications of cranial radiotherapy of head and neck cancers. However, the underlying mechanism of this cognitive impairment remains unclear. In the present study, the role of tropomyosin receptor kinase A (TrkA) and its ligand neurotrophin nerve growth factor (NGF) were investigated following whole‑brain irradiation (WBI). Young male Sprague‑Dawley rats underwent WBI at a single dose of 10 Gy. WBI was determined to result in notable memory decline and substantial neurogenesis impairment in the hippocampus 3 months post‑irradiation. Compared with the control group, TrkA protein expression was greater in irradiated rats 1 week after WBI, which then decreased significantly by the 3‑month time‑point. However, no difference in NGF expression was observed from 1 day to 3 months post‑WBI. Overexpression of hippocampal TrkA in rats using adeno‑associated virus ameliorated memory decline induced by irradiation. Additionally, upregulating TrkA expression rescued irradiation‑induced hippocampal precursor cell proliferation and promoted neurogenesis. PI3K, Akt and ERK1/2 phosphorylation were also revealed to be significantly inhibited by WBI, which was ameliorated by TrkA overexpression. Findings of the present study indicated that the TrkA‑dependent signaling pathway may serve a critical role in radiotherapy‑induced cognitive deficit and impairments in neurogenesis.
Collapse
Affiliation(s)
- Shengjun Ji
- Clinical College, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
- Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Haohao Wu
- Department of Radiotherapy and Oncology, Yancheng No. 1 People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Xin Ding
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Qingqing Chen
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Xing Jin
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Jinming Yu
- Clinical College, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ming Yang
- Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
69
|
Sep MSC, Joëls M, Geuze E. Individual differences in the encoding of contextual details following acute stress: An explorative study. Eur J Neurosci 2020; 55:2714-2738. [PMID: 33249674 PMCID: PMC9291333 DOI: 10.1111/ejn.15067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/05/2020] [Accepted: 11/21/2020] [Indexed: 12/19/2022]
Abstract
Information processing under stressful circumstances depends on many experimental conditions, like the information valence or the point in time at which brain function is probed. This also holds true for memorizing contextual details (or ‘memory contextualization’). Moreover, large interindividual differences appear to exist in (context‐dependent) memory formation after stress, but it is mostly unknown which individual characteristics are essential. Various characteristics were explored from a theory‐driven and data‐driven perspective, in 120 healthy men. In the theory‐driven model, we postulated that life adversity and trait anxiety shape the stress response, which impacts memory contextualization following acute stress. This was indeed largely supported by linear regression analyses, showing significant interactions depending on valence and time point after stress. Thus, during the acutephase of the stress response, reduced neutral memory contextualization was related to salivary cortisol level; moreover, certain individual characteristics correlated with memory contextualization of negatively valenced material: (a) life adversity, (b) α‐amylase reactivity in those with low life adversity and (c) cortisol reactivity in those with low trait anxiety. Better neutral memory contextualization during the recoveryphase of the stress response was associated with (a) cortisol in individuals with low life adversity and (b) α‐amylase in individuals with high life adversity. The data‐driven Random Forest‐based variable selection also pointed to (early) life adversity—during the acutephase—and (moderate) α‐amylase reactivity—during the recoveryphase—as individual characteristics related to better memory contextualization. Newly identified characteristics sparked novel hypotheses about non‐anxious personality traits, age, mood and states during retrieval of context‐related information.
Collapse
Affiliation(s)
- Milou S C Sep
- Brain Research and Innovation Centre, Ministry of Defence, Utrecht, The Netherlands.,Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elbert Geuze
- Brain Research and Innovation Centre, Ministry of Defence, Utrecht, The Netherlands.,Department of Psychiatry, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
70
|
Jung HY, Kim W, Kwon HJ, Yoo DY, Nam SM, Hahn KR, Yi SS, Choi JH, Kim DW, Yoon YS, Hwang IK. Physical Stress Induced Reduction of Proliferating Cells and Differentiated Neuroblasts Is Ameliorated by Fermented Laminaria japonica Extract Treatment. Mar Drugs 2020; 18:E587. [PMID: 33255381 PMCID: PMC7760277 DOI: 10.3390/md18120587] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Laminaria japonica is widely cultivated in East Asia, including South Korea. Fucoidan, a main component of L. japonica, protects neurons from neurological disorders such as ischemia and traumatic brain injury. In the present study, we examined the effects of extract from fermented L. japonica on the reduction of proliferating cells and neuroblasts in mice that were physically (with electric food shock) or psychologically (with visual, auditory and olfactory sensation) stressed with the help of a communication box. Vehicle (distilled water) or fermented L. japonica extract (50 mg/kg) were orally administered to the mice once a day for 21 days. On the 19th day of the treatment, physical and psychological stress was induced by foot shock using a communication box and thereafter for three days. Plasma corticosterone levels were significantly increased after exposure to physical stress and decreased Ki67 positive proliferating cells and doublecortin immunoreactive neuroblasts. In addition, western blot analysis demonstrated that physical stress as well as psychological stress decreased the expression levels of brain-derived neurotrophic factor (BDNF) and the number of phosphorylated cAMP response element binding protein (pCREB) positive nuclei in the dentate gyrus. Fermentation of L. japonica extract significantly increased the contents of reduced sugar and phenolic compounds. Supplementation with fermented L. japonica extract significantly ameliorated the increases of plasma corticosterone revels and decline in the proliferating cells, neuroblasts, and expression of BDNF and pCREB in the physically stressed mice. These results indicate that fermented L. japonica extract has positive effects in ameliorating the physical stress induced reduction in neurogenesis by modulating BDNF and pCREB expression in the dentate gyrus.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Dae Young Yoo
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea;
| | - Sung Min Nam
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Iksan 54538, Korea;
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Sun Shin Yi
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea;
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| |
Collapse
|
71
|
Maharjan R, Diaz Bustamante L, Ghattas KN, Ilyas S, Al-Refai R, Khan S. Role of Lifestyle in Neuroplasticity and Neurogenesis in an Aging Brain. Cureus 2020; 12:e10639. [PMID: 33133809 PMCID: PMC7586385 DOI: 10.7759/cureus.10639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 09/24/2020] [Indexed: 11/21/2022] Open
Abstract
Neuroplasticity is the brain's ability to transform its shape, adapt, and develop a new neuronal connection provided with a new stimulus. The stronger the electrical stimulation, the robust is the transformation. Neurogenesis is a complex process when the new neuronal blast cells present in the dentate gyrus divide in the hippocampus. We collected articles from the past 11 years for review, using the Medical Subject Headings (MeSH) strategy from PubMed. Quality appraisal was done for each research article using various assessment tools. A total of 24 articles were chosen, applying all the mentioned inclusion and exclusion criteria and reviewed. The reviewed studies emphasized that modifiable lifestyle factors such as diet and exercise should be implemented as an intervention in the elderly for healthy aging of the brain, as the world's aging population is going to be increased, leading to the expansion of health care and cost. Multiple studies have publicized the relation of diet and exercise with cognition function in aging people. A diet consisting of curcumin in its food has its anti-oxidative property, which prevents rapid aging of the brain, other diet patterns such as a caloric restriction diet can influence brain plasticity and preclude the decline of memory. Exercise can increase brain-derived growth factor (BDGF), vascular endothelial growth factor (VEGF), synapsin one, and tyrosine kinase activity that can expand the size of the brain, enhance the plasticity and neurogenesis. This review aimed at exploring lifestyle factors that contribute to neuroplasticity and neurogenesis. Thus, providing a new path for clinicians and researchers to map out the future possible significant benefits for optimal brain aging in a healthy fashion.
Collapse
Affiliation(s)
- Reeju Maharjan
- Neurology, V.N. Karazin Kharkiv National University, Kharkiv, UKR
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Liliana Diaz Bustamante
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kyrillos N Ghattas
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shahbakht Ilyas
- Medicine and Surgery, CMH Lahore Medical College and Institute of Dentistry, Lahore, PAK
- Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Reham Al-Refai
- Pathology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
72
|
Ghaddar B, Veeren B, Rondeau P, Bringart M, Lefebvre d'Hellencourt C, Meilhac O, Bascands JL, Diotel N. Impaired brain homeostasis and neurogenesis in diet-induced overweight zebrafish: a preventive role from A. borbonica extract. Sci Rep 2020; 10:14496. [PMID: 32879342 PMCID: PMC7468118 DOI: 10.1038/s41598-020-71402-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Overweight and obesity are worldwide health concerns leading to many physiological disorders. Recent data highlighted their deleterious effects on brain homeostasis and plasticity, but the mechanisms underlying such disruptions are still not well understood. In this study, we developed and characterized a fast and reliable diet-induced overweight (DIO) model in zebrafish, for (1) studying the effects of overfeeding on brain homeostasis and for (2) testing different preventive and/or therapeutic strategies. By overfeeding zebrafish for 4 weeks, we report the disruption of many metabolic parameters reproducing human overweight features including increased body weight, body mass index, fasting blood glucose levels and liver steatosis. Furthermore, DIO fish displayed blood–brain barrier leakage, cerebral oxidative stress, neuroinflammation and decreased neurogenesis. Finally, we investigated the preventive beneficial effects of A. borbonica, an endogenous plant from Reunion Island. Overnight treatment with A. borbonica aqueous extract during the 4 weeks of overfeeding limited some detrimental central effects of DIO. In conclusion, we established a relevant DIO model in zebrafish demonstrating that overfeeding impairs peripheral and central homeostasis. This work also highlights the preventive protective effects of A. borbonica aqueous extracts in DIO, and opens a way to easily screen drugs aiming at limiting overweight and associated neurological disorders.
Collapse
Affiliation(s)
- Batoul Ghaddar
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France
| | - Bryan Veeren
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France
| | - Philippe Rondeau
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France
| | - Matthieu Bringart
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France
| | - Christian Lefebvre d'Hellencourt
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France
| | - Olivier Meilhac
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France.,CHU de La Réunion, Saint-Denis, France
| | - Jean-Loup Bascands
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France
| | - Nicolas Diotel
- INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Denis de La Réunion, France.
| |
Collapse
|
73
|
Berger T, Lee H, Young AH, Aarsland D, Thuret S. Adult Hippocampal Neurogenesis in Major Depressive Disorder and Alzheimer's Disease. Trends Mol Med 2020; 26:803-818. [PMID: 32418723 DOI: 10.1016/j.molmed.2020.03.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/16/2020] [Accepted: 03/31/2020] [Indexed: 12/25/2022]
Abstract
Depression and dementia are major public health problems. Major depressive disorder (MDD) and Alzheimer's disease (AD) reciprocally elevate the risk for one another. No effective drug is available to treat AD and about one-third of depressive patients show treatment resistance. The biological connection between MDD and AD is still unclear. Uncovering this link might open novel ways of treatment and prevention to improve patient healthcare. Here, we discuss recent studies specifically on the role of human adult hippocampal neurogenesis (AHN) in MDD and AD. We compare diverse approaches to analyse the effect of MDD and AD on human AHN and analyse different studies implicating the role of human AHN as a potential converging mechanism in MDD and AD.
Collapse
Affiliation(s)
- Thomas Berger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Hyunah Lee
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
74
|
Harvey AR. Links Between the Neurobiology of Oxytocin and Human Musicality. Front Hum Neurosci 2020; 14:350. [PMID: 33005139 PMCID: PMC7479205 DOI: 10.3389/fnhum.2020.00350] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/04/2020] [Indexed: 12/22/2022] Open
Abstract
The human species possesses two complementary, yet distinct, universal communication systems—language and music. Functional imaging studies have revealed that some core elements of these two systems are processed in closely related brain regions, but there are also clear differences in brain circuitry that likely underlie differences in functionality. Music affects many aspects of human behavior, especially in encouraging prosocial interactions and promoting trust and cooperation within groups of culturally compatible but not necessarily genetically related individuals. Music, presumably via its impact on the limbic system, is also rewarding and motivating, and music can facilitate aspects of learning and memory. In this review these special characteristics of music are considered in light of recent research on the neuroscience of the peptide oxytocin, a hormone that has both peripheral and central actions, that plays a role in many complex human behaviors, and whose expression has recently been reported to be affected by music-related activities. I will first briefly discuss what is currently known about the peptide’s physiological actions on neurons and its interactions with other neuromodulator systems, then summarize recent advances in our knowledge of the distribution of oxytocin and its receptor (OXTR) in the human brain. Next, the complex links between oxytocin and various social behaviors in humans are considered. First, how endogenous oxytocin levels relate to individual personality traits, and then how exogenous, intranasal application of oxytocin affects behaviors such as trust, empathy, reciprocity, group conformity, anxiety, and overall social decision making under different environmental conditions. It is argued that many of these characteristics of oxytocin biology closely mirror the diverse effects that music has on human cognition and emotion, providing a link to the important role music has played throughout human evolutionary history and helping to explain why music remains a special prosocial human asset. Finally, it is suggested that there is a potential synergy in combining oxytocin- and music-based strategies to improve general health and aid in the treatment of various neurological dysfunctions.
Collapse
Affiliation(s)
- Alan R Harvey
- School of Human Sciences, The University of Western Australia, Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| |
Collapse
|
75
|
Kumar D, Koyanagi I, Carrier-Ruiz A, Vergara P, Srinivasan S, Sugaya Y, Kasuya M, Yu TS, Vogt KE, Muratani M, Ohnishi T, Singh S, Teixeira CM, Chérasse Y, Naoi T, Wang SH, Nondhalee P, Osman BAH, Kaneko N, Sawamoto K, Kernie SG, Sakurai T, McHugh TJ, Kano M, Yanagisawa M, Sakaguchi M. Sparse Activity of Hippocampal Adult-Born Neurons during REM Sleep Is Necessary for Memory Consolidation. Neuron 2020; 107:552-565.e10. [PMID: 32502462 DOI: 10.1016/j.neuron.2020.05.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 03/21/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022]
Abstract
The occurrence of dreaming during rapid eye movement (REM) sleep prompts interest in the role of REM sleep in hippocampal-dependent episodic memory. Within the mammalian hippocampus, the dentate gyrus (DG) has the unique characteristic of exhibiting neurogenesis persisting into adulthood. Despite their small numbers and sparse activity, adult-born neurons (ABNs) in the DG play critical roles in memory; however, their memory function during sleep is unknown. Here, we investigate whether young ABN activity contributes to memory consolidation during sleep using Ca2+ imaging in freely moving mice. We found that contextual fear learning recruits a population of young ABNs that are reactivated during subsequent REM sleep against a backdrop of overall reduced ABN activity. Optogenetic silencing of this sparse ABN activity during REM sleep alters the structural remodeling of spines on ABN dendrites and impairs memory consolidation. These findings provide a causal link between ABN activity during REM sleep and memory consolidation.
Collapse
Affiliation(s)
- Deependra Kumar
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Iyo Koyanagi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Alvaro Carrier-Ruiz
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo 113-0033, Japan
| | - Pablo Vergara
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Sakthivel Srinivasan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Yuki Sugaya
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo 113-0033, Japan
| | - Masatoshi Kasuya
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Tzong-Shiue Yu
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Kaspar E Vogt
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Takaaki Ohnishi
- Graduate School of Information Science and Technology, The University of Tokyo, Tokyo 113-8656, Japan
| | - Sima Singh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Catia M Teixeira
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, NY 10962, USA
| | - Yoan Chérasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Toshie Naoi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Szu-Han Wang
- Centre for Clinical Brain Sciences, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Pimpimon Nondhalee
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Boran A H Osman
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Biology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan; Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Steven G Kernie
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Thomas J McHugh
- RIKEN Center for Brain Science, Wako, Saitama 351-0106, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo 113-0033, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan.
| |
Collapse
|
76
|
Endonuclease VIII-like 1 deficiency impairs survival of newly generated hippocampal neurons and memory performance in young-adult male mice. Life Sci 2020; 254:117755. [PMID: 32437792 DOI: 10.1016/j.lfs.2020.117755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 11/20/2022]
Abstract
AIMS Efficient memory formation in rodents depends on adult neurogenesis in the subgranular zone of the hippocampus, and mounting evidence suggests that deficiencies in initiating repair of oxidatively induced DNA damage may impair neurogenesis. Hence, we aimed to determine whether loss of the DNA glycosylase, endonuclease VIII-like 1 (Neil1), affects hippocampal neurogenesis and memory performance in young-adult mice. MAIN METHODS Eight-week-old male wild-type and Neil1-deficient (Neil1-/-) mice were treated with bromodeoxyuridine to track neuronal proliferation and differentiation. A neurosphere formation assay was further used to measure neuroprogenitor proliferative capacity. Hippocampus-related memory functions were assessed with Y-maze spontaneous alternation and novel object recognition tests. KEY FINDINGS Young-adult male Neil1-/- mice exhibited diminished adult hippocampal neurogenesis in the dentate gyrus, probably as a result of poor survival of newly proliferated neurons. Furthermore, the Y-maze spontaneous alternation and novel object recognition tests respectively revealed that Neil1 deficiency impairs spatial and non-spatial hippocampus-related memory functions. We also found that expression of p53, a central regulator of apoptosis, was upregulated in the dentate gyrus of Neil1-/- mice, while the level of β-catenin, a key cell survival molecule, was downregulated. SIGNIFICANCE The DNA glycosylase, Neil1, promotes successful hippocampal neurogenesis and learning and memory in young-adult mice.
Collapse
|
77
|
Fotuhi SN, Khalaj-Kondori M, Feizi MAH, Talebi M. Memory-related process in physiological status and alzheimer's disease. Mol Biol Rep 2020; 47:4651-4657. [PMID: 32279208 DOI: 10.1007/s11033-020-05438-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/03/2019] [Indexed: 12/27/2022]
Abstract
Rejecting central dogma around static status of adult mammalian brain, CNS has the nascent neurons generated in subgranular zone of dentate gyrus in hippocampus which develop to novel glutamatergic granule cells, with the innate feature of transmuting to memory disks. Structural plasticity proceeds with synaptic plasticity to process all the developing stages required to successful maturation and functional integration, whereby the memory context is ready to leave the hippocampus toward cortex network through consolidation process, for being installed and run the memory disk forever. However, in Alzheimer's disease, brain deal with subtle deadly progressive loss of synapsis, neuronal dysfunction and ultimately network failure, resulting in memory decay and cognitive decline-concluding that AD destroys memory formation related-pathways.
Collapse
Affiliation(s)
- Seyedeh Nahid Fotuhi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | | | - Mahnaz Talebi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
78
|
Keshavarzi M, Khoshnoud MJ, Ghaffarian Bahraman A, Mohammadi-Bardbori A. An Endogenous Ligand of Aryl Hydrocarbon Receptor 6-Formylindolo[3,2-b]Carbazole (FICZ) Is a Signaling Molecule in Neurogenesis of Adult Hippocampal Neurons. J Mol Neurosci 2020; 70:806-817. [DOI: 10.1007/s12031-020-01506-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
|
79
|
Spritzer MD, Roy EA. Testosterone and Adult Neurogenesis. Biomolecules 2020; 10:biom10020225. [PMID: 32028656 PMCID: PMC7072323 DOI: 10.3390/biom10020225] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
It is now well established that neurogenesis occurs throughout adulthood in select brain regions, but the functional significance of adult neurogenesis remains unclear. There is considerable evidence that steroid hormones modulate various stages of adult neurogenesis, and this review provides a focused summary of the effects of testosterone on adult neurogenesis. Initial evidence came from field studies with birds and wild rodent populations. Subsequent experiments with laboratory rodents have tested the effects of testosterone and its steroid metabolites upon adult neurogenesis, as well as the functional consequences of induced changes in neurogenesis. These experiments have provided clear evidence that testosterone increases adult neurogenesis within the dentate gyrus region of the hippocampus through an androgen-dependent pathway. Most evidence indicates that androgens selectively enhance the survival of newly generated neurons, while having little effect on cell proliferation. Whether this is a result of androgens acting directly on receptors of new neurons remains unclear, and indirect routes involving brain-derived neurotrophic factor (BDNF) and glucocorticoids may be involved. In vitro experiments suggest that testosterone has broad-ranging neuroprotective effects, which will be briefly reviewed. A better understanding of the effects of testosterone upon adult neurogenesis could shed light on neurological diseases that show sex differences.
Collapse
Affiliation(s)
- Mark D. Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA
- Correspondence: ; Tel.: 802-443-5676
| | - Ethan A. Roy
- Graduate School of Education, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
80
|
Wang ZJ, Han YF, Zhao F, Yang GZ, Yuan L, Cai HY, Yang JT, Holscher C, Qi JS, Wu MN. A dual GLP-1 and Gcg receptor agonist rescues spatial memory and synaptic plasticity in APP/PS1 transgenic mice. Horm Behav 2020; 118:104640. [PMID: 31765661 DOI: 10.1016/j.yhbeh.2019.104640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 11/16/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that severely affects the health and lifespan of the elderly worldwide. Recently, the correlation between AD and type 2 diabetes mellitus (T2DM) has received intensive attention, and a promising new anti-AD strategy is the use of anti-diabetic drugs. Oxyntomodulin (Oxm) is a peptide hormone and growth factor that acts on neurons in the hypothalamus. OXM activates glucagon-like peptide 1 (GLP-1) and glucagon (Gcg) receptors, facilitates insulin signaling and has neuroprotective effects against Aβ1-42-induced cytotoxicity in primary hippocampal neurons. Here, we tested the effects of the protease-resistant analogue (D-Ser2)Oxm on spatial memory and synaptic plasticity and the underlying molecular mechanisms in the APP/PS1 transgenic mouse model of AD. The results showed that (D-Ser2)Oxm not only alleviated the impairments of working memory and long-term spatial memory, but also reduced the number of Aβ plaques in the hippocampus, and reversed the suppression of hippocampal synaptic long-term potentiation (LTP). Moreover, (D-Ser2)Oxm administration significantly increased p-PI3K/p-AKT1 expression and decreased p-GSK3β levels in the hippocampus. These results are the first to show an in vivo neuroprotective role of (D-Ser2)Oxm in APP/PS1 mice, and this role involves the improvement of synaptic plasticity, clearance of Aβ and normalization of PI3K/AKT/GSK3β cell signaling in the hippocampus. This study suggests that (D-Ser2)Oxm holds promise for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, PR China
| | - Yu-Fei Han
- Guangzhou Kingmed Diagnostics, Guangzhou, PR China
| | - Fang Zhao
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, PR China
| | - Guang-Zhao Yang
- Department of Cardiovascular Medicine, The First Hospital of Shanxi Medical University, Taiyuan, PR China
| | - Li Yuan
- Department of Physiology, Changzhi Medical College, Changzhi, PR China
| | - Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, PR China
| | - Jun-Ting Yang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, PR China
| | - Christian Holscher
- Neuroscience research group, Henan university of Chinese medicine, Zhengzhou, PR China
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, PR China.
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, PR China.
| |
Collapse
|
81
|
Cicvaric A, Sachernegg HM, Stojanovic T, Symmank D, Smani T, Moeslinger T, Uhrin P, Monje FJ. Podoplanin Gene Disruption in Mice Promotes in vivo Neural Progenitor Cells Proliferation, Selectively Impairs Dentate Gyrus Synaptic Depression and Induces Anxiety-Like Behaviors. Front Cell Neurosci 2020; 13:561. [PMID: 32009902 PMCID: PMC6974453 DOI: 10.3389/fncel.2019.00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Podoplanin (Pdpn), a brain-tumor-related glycoprotein identified in humans and animals, is endogenously expressed in several organs critical for life support such as kidney, lung, heart and brain. In the brain, Pdpn has been identified in proliferative nestin-positive adult neural progenitor cells and in neurons of the neurogenic hippocampal dentate gyrus (DG), a structure associated to anxiety, critical for learning and memory functions and severely damaged in people with Alzheimer's Disease (AD). The in vivo role of Pdpn in adult neurogenesis and anxiety-like behavior remained however unexplored. Using mice with disrupted Pdpn gene as a model organism and applying combined behavioral, molecular biological and electrophysiological assays, we here show that the absence of Pdpn selectively impairs long-term synaptic depression in the neurogenic DG without affecting the CA3-Schaffer's collateral-CA1 synapses. Pdpn deletion also enhanced the proliferative capacity of DG neural progenitor cells and diminished survival of differentiated neuronal cells in vitro. In addition, mice with podoplanin gene disruption showed increased anxiety-like behaviors in experimentally validated behavioral tests as compared to wild type littermate controls. Together, these findings broaden our knowledge on the molecular mechanisms influencing hippocampal synaptic plasticity and neurogenesis in vivo and reveal Pdpn as a novel molecular target for future studies addressing general anxiety disorder and synaptic depression-related memory dysfunctions.
Collapse
Affiliation(s)
- Ana Cicvaric
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannah M. Sachernegg
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Dörte Symmank
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville (IBiS)/University of Seville/CIBERCV, Seville, Spain
| | - Thomas Moeslinger
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
82
|
Excitation/inhibition imbalance and impaired neurogenesis in neurodevelopmental and neurodegenerative disorders. Rev Neurosci 2019; 30:807-820. [DOI: 10.1515/revneuro-2019-0014] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022]
Abstract
AbstractThe excitation/inhibition (E/I) balance controls the synaptic inputs to prevent the inappropriate responses of neurons to input strength, and is required to restore the initial pattern of network activity. Various neurotransmitters affect synaptic plasticity within neural networks via the modulation of neuronal E/I balance in the developing and adult brain. Less is known about the role of E/I balance in the control of the development of the neural stem and progenitor cells in the course of neurogenesis and gliogenesis. Recent findings suggest that neural stem and progenitor cells appear to be the target for the action of GABA within the neurogenic or oligovascular niches. The same might be true for the role of neuropeptides (i.e. oxytocin) in neurogenic niches. This review covers current understanding of the role of E/I balance in the regulation of neuroplasticity associated with social behavior in normal brain, and in neurodevelopmental and neurodegenerative diseases. Further studies are required to decipher the GABA-mediated regulation of postnatal neurogenesis and synaptic integration of newly-born neurons as a potential target for the treatment of brain diseases.
Collapse
|
83
|
Terranova JI, Ogawa SK, Kitamura T. Adult hippocampal neurogenesis for systems consolidation of memory. Behav Brain Res 2019; 372:112035. [DOI: 10.1016/j.bbr.2019.112035] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022]
|
84
|
High-intensity interval training prevents cognitive-motor impairment and serum BDNF level reduction in parkinson mice model. SPORT SCIENCES FOR HEALTH 2019. [DOI: 10.1007/s11332-019-00586-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
85
|
Barry DN, Maguire EA. Consolidating the Case for Transient Hippocampal Memory Traces. Trends Cogn Sci 2019; 23:635-636. [PMID: 31270021 DOI: 10.1016/j.tics.2019.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Daniel N Barry
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, 12 Queen Square, London, WC1N 3AR, UK
| | - Eleanor A Maguire
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, 12 Queen Square, London, WC1N 3AR, UK.
| |
Collapse
|
86
|
Pregnancy Promotes Maternal Hippocampal Neurogenesis in Guinea Pigs. Neural Plast 2019; 2019:5765284. [PMID: 31097956 PMCID: PMC6487096 DOI: 10.1155/2019/5765284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/08/2019] [Accepted: 02/21/2019] [Indexed: 11/26/2022] Open
Abstract
Adult neurogenesis in the hippocampal dentate gyrus (DG) modulates cognition and behavior in mammals, while motherhood is associated with cognitive and behavioral changes essential for the care of the young. In mice and rats, hippocampal neurogenesis is reported to be reduced or unchanged during pregnancy, with few data available from other species. In guinea pigs, pregnancy lasts ~9 weeks; we set to explore if hippocampal neurogenesis is altered in these animals, relative to gestational stages. Time-pregnant primigravidas (3-5 months old) and age-matched nonpregnant females were examined, with neurogenic potential evaluated via immunolabeling of Ki67, Sp8, doublecortin (DCX), and neuron-specific nuclear antigen (NeuN) combined with bromodeoxyuridine (BrdU) birth-dating. Relative to control, subgranular Ki67, Sp8, and DCX-immunoreactive (+) cells tended to increase from early gestation to postpartum and peaked at the late gestational stage. In BrdU pulse-chasing experiments in nonpregnant females surviving for different time points (2-120 days), BrdU+ cells in the DG colocalized with DCX partially from 2 to 42 days (most frequently at 14-30 days) and with NeuN increasingly from 14 to 120 days. In pregnant females that received BrdU at early, middle, and late gestational stages and survived for 42 days, the density of BrdU+ cells in the DG was mostly high in the late gestational group. The rates of BrdU/DCX and BrdU/NeuN colocalization were similar among these groups and comparable to those among the corresponding control group. Together, the findings suggest that pregnancy promotes maternal hippocampal neurogenesis in guinea pigs, at least among primigravidas.
Collapse
|
87
|
Augusto-Oliveira M, Arrifano GPF, Malva JO, Crespo-Lopez ME. Adult Hippocampal Neurogenesis in Different Taxonomic Groups: Possible Functional Similarities and Striking Controversies. Cells 2019; 8:cells8020125. [PMID: 30764477 PMCID: PMC6406791 DOI: 10.3390/cells8020125] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 12/13/2022] Open
Abstract
Adult neurogenesis occurs in many species, from fish to mammals, with an apparent reduction in the number of both neurogenic zones and new neurons inserted into established circuits with increasing brain complexity. Although the absolute number of new neurons is high in some species, the ratio of these cells to those already existing in the circuit is low. Continuous replacement/addition plays a role in spatial navigation (migration) and other cognitive processes in birds and rodents, but none of the literature relates adult neurogenesis to spatial navigation and memory in primates and humans. Some models developed by computational neuroscience attribute a high weight to hippocampal adult neurogenesis in learning and memory processes, with greater relevance to pattern separation. In contrast to theories involving neurogenesis in cognitive processes, absence/rarity of neurogenesis in the hippocampus of primates and adult humans was recently suggested and is under intense debate. Although the learning process is supported by plasticity, the retention of memories requires a certain degree of consolidated circuitry structures, otherwise the consolidation process would be hampered. Here, we compare and discuss hippocampal adult neurogenesis in different species and the inherent paradoxical aspects.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil.
- Laboratory of Research on Neurodegeneration and Infection, University Hospital João de Barros Barreto, Federal University of Pará, Belém 66073-005, Brazil.
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK.
| | - Gabriela P F Arrifano
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil.
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK.
| | - João O Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), and Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences (CNC.IBILI), Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil.
| |
Collapse
|
88
|
Oishi N, Nomoto M, Ohkawa N, Saitoh Y, Sano Y, Tsujimura S, Nishizono H, Matsuo M, Muramatsu SI, Inokuchi K. Artificial association of memory events by optogenetic stimulation of hippocampal CA3 cell ensembles. Mol Brain 2019; 12:2. [PMID: 30621738 PMCID: PMC6323779 DOI: 10.1186/s13041-018-0424-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/26/2018] [Indexed: 11/10/2022] Open
Abstract
Previous gain-of-function studies using an optogenetic technique showed that manipulation of the hippocampal dentate gyrus or CA1 cell ensembles is important for memory reactivation and to generate synthetic or false memory. However, gain-of-function study manipulating CA3 cell ensembles has not been reported. The CA3 area of the hippocampus comprises a recurrent excitatory circuit, which is thought to be important for the generation of associations among the stored information within one brain region. We investigated whether the coincident firing of cell ensembles in one brain region, hippocampal CA3, associates distinct events. CA3 cell ensembles responding to context exploration and during contextual fear conditioning were labeled with channelrhodopsin-2 (ChR2)-mCherry. The synchronous activation of these ensembles induced freezing behavior in mice in a neutral context, in which a foot shock had never been delivered. The recall of this artificial associative fear memory was context specific. In vivo electrophysiological recordings showed that 20-Hz optical stimulation of ChR2-mCherry-expressing CA3 neurons, which is the same stimulation protocol used in behavioral experiment, induced long-term potentiation at CA3-CA3 synapses. Altogether, these results demonstrate that the synchronous activation of ensembles in one brain region, CA3 of the hippocampus, is sufficient for the association of distinct events. The results of our electrophysiology potentially suggest that this artificial association of memory events might be induced by the strengthening of synaptic efficacy between CA3 ensembles via recurrent circuit.
Collapse
Affiliation(s)
- Naoya Oishi
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan
| | - Masanori Nomoto
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan
| | - Noriaki Ohkawa
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), JST, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshito Saitoh
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan
| | - Yoshitake Sano
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan.,Present address: Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Shuhei Tsujimura
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan
| | - Hirofumi Nishizono
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan.,Division of Animal Experimental Laboratory, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Mina Matsuo
- Division of Animal Experimental Laboratory, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan.,Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Kaoru Inokuchi
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan. .,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
89
|
Bacopa monnieri extract improves novel object recognition, cell proliferation, neuroblast differentiation, brain-derived neurotrophic factor, and phosphorylation of cAMP response element-binding protein in the dentate gyrus. Lab Anim Res 2018; 34:239-247. [PMID: 30671111 PMCID: PMC6333610 DOI: 10.5625/lar.2018.34.4.239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 01/22/2023] Open
Abstract
Bacopa monnieri is a medicinal plant with a long history of use in Ayurveda, especially in the treatment of poor memory and cognitive deficits. In the present study, we hypothesized that Bacopa monnieri extract (BME) can improve memory via increased cell proliferation and neuroblast differentiation in the dentate gyrus. BME was administered to 7-week-old mice once a day for 4 weeks and a novel object recognition memory test was performed. Thereafter, the mice were euthanized followed by immunohistochemistry analysis for Ki67, doublecortin (DCX), and phosphorylated cAMP response element-binding protein (CREB), and western blot analysis of brain-derived neurotrophic factor (BDNF). BME-treated mice showed moderate increases in the exploration of new objects when compared with that of familiar objects, leading to a significant higher discrimination index compared with vehicle-treated mice. Ki67 and DCX immunohistochemistry showed a facilitation of cell proliferation and neuroblast differentiation following the administration of BME in the dentate gyrus. In addition, administration of BME significantly elevated the BDNF protein expression in the hippocampal dentate gyrus, and increased CREB phosphorylation in the dentate gyrus. These data suggest that BME improves novel object recognition by increasing the cell proliferation and neuroblast differentiation in the dentate gyrus, and this may be closely related to elevated levels of BDNF and CREB phosphorylation in the dentate gyrus.
Collapse
|