1
|
Altamirano FG, Castro-Pascual I, Ponce IT, Coria-Lucero CD, Cargnelutti E, Ferramola ML, Delgado MS, Anzulovich AC, Lacoste MG. Late-Onset Caloric Restriction Improves Cognitive Performance and Restores Circadian Patterns of Neurotrophic, Clock, and Epigenetic Factors in the Hippocampus of Old Male Rats. J Gerontol A Biol Sci Med Sci 2024; 80:glae252. [PMID: 39447038 DOI: 10.1093/gerona/glae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Indexed: 10/26/2024] Open
Abstract
Aging is a complex multifactorial process that results in a general functional decline, including cognitive impairment. Caloric restriction (CR) can positively influence the aging processes and delay cognitive decline. There is a rhythmic variation in memory and learning processes throughout the day, indicating the involvement of the circadian clock in the regulation of these processes. Despite growing evidence on the efficacy of CR, it has not yet been fully determined whether starting this strategy at an advanced age is beneficial for improving quality of life and eventually, for protection against age-related diseases. Here, we investigated the effect of late-onset CR on the temporal organization of the molecular clock machinery, molecules related to cognitive processes and epigenetic regulation, in the hippocampus of old male rats maintained under constant darkness conditions. Our results evidenced the existence of a highly coordinated temporal organization of Bmal1, Clock, Bdnf, Trkb, Dnmts, Sirt1, and Pgc-1α in the hippocampus of young adult rats. We observed that aging led to cognitive deficits and loss of circadian oscillations of all the above variables. Interestingly, CR restored circadian rhythmicity in all cases and, in addition, improved the cognitive performance of the old animals. This work would highlight the importance of the circadian clock and its synchronization with feeding signals, as the basis of the beneficial effects of CR. Thus, lifestyle modifications, such as CR, might be a powerful intervention to preserve hippocampal circadian organization and cognitive health during aging.
Collapse
Affiliation(s)
- Fernando Gabriel Altamirano
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ivanna Castro-Pascual
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ivana Tamara Ponce
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Cinthia Daiana Coria-Lucero
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ethelina Cargnelutti
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Mariana Lucila Ferramola
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Marcela Silvia Delgado
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - Ana Cecilia Anzulovich
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| | - María Gabriela Lacoste
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research - San Luis (IMIBIO-SL), National Council for Scientific and Technical Research (CONICET), San Luis, Argentina
- Faculty of Chemistry, Biochemistry and Pharmacy (FQByF), National University of San Luis (UNSL), San Luis, Argentina
| |
Collapse
|
2
|
Zhang Y, Chen Y, Li W, Tang L, Li J, Feng X. Targeting the circadian modulation: novel therapeutic approaches in the management of ASD. Front Psychiatry 2024; 15:1451242. [PMID: 39465045 PMCID: PMC11503653 DOI: 10.3389/fpsyt.2024.1451242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024] Open
Abstract
Circadian dysfunction is prevalent in neurodevelopmental disorders, particularly in autism spectrum disorder (ASD). A plethora of empirical studies demonstrate a strong correlation between ASD and circadian disruption, suggesting that modulation of circadian rhythms and the clocks could yield satisfactory advancements. Research indicates that circadian dysfunction associated with abnormal neurodevelopmental phenotypes in ASD individuals, potentially contribute to synapse plasticity disruption. Therefore, targeting circadian rhythms may emerge as a key therapeutic approach. In this study, we did a brief review of the mammalian circadian clock, and the correlation between the circadian mechanism and the pathology of ASD at multiple levels. In addition, we highlight that circadian is the target or modulator to participate in the therapeutic approaches in the management of ASD, such as phototherapy, melatonin, modulating circadian components, natural compounds, and chronotherapies. A deep understanding of the circadian clock's regulatory role in the neurodevelopmental phenotypes in ASD may inspire novel strategies for improving ASD treatment.
Collapse
Affiliation(s)
- Yuxing Zhang
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yinan Chen
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wu Li
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liya Tang
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiangshan Li
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiang Feng
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
3
|
Pötzl L, Wolf OT, Merz CJ. The influence of time of day on memory recognition for faces. Horm Behav 2024; 165:105633. [PMID: 39244875 DOI: 10.1016/j.yhbeh.2024.105633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Time of day can alter memory performance in general. Its influence on memory recognition performance for faces, which is important for daily encounters with new persons or testimonies, has not been investigated yet. Importantly, high levels of the stress hormone cortisol impair memory recognition, in particular for emotional material. However, some studies also reported high cortisol levels to enhance memory recognition. Since cortisol levels in the morning are usually higher than in the evening, time of day might also influence recognition performance. In this pre-registered study with a two-day design, 51 healthy men encoded pictures of male and female faces with distinct emotional expressions on day one around noon. Memory for the faces was retrieved two days later at two consecutive testing times either in the morning (high and moderately increased endogenous cortisol levels) or in the evening (low endogenous cortisol levels). Additionally, alertness as well as salivary cortisol levels at the different timepoints was assessed. Cortisol levels were significantly higher in the morning compared to the evening group as expected, while both groups did not differ in alertness. Familiarity ratings for female stimuli were significantly better when participants were tested during moderately increased endogenous cortisol levels in the morning than during low endogenous cortisol levels in the evening, a pattern which was previously also observed for stressed versus non-stressed participants. In addition, cortisol levels during that time in the morning were positively correlated with the recollection of face stimuli in general. Thus, recognition memory performance may depend on the time of day and as well as on stimulus type, such as the difference of male and female faces. Most importantly, the results suggest that cortisol may be meaningful and worth investigating when studying the effects of time of day on memory performance. This research offers both, insights into daily encounters as well as legally relevant domains as for instance testimonies.
Collapse
Affiliation(s)
- Lisa Pötzl
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Germany
| | - Oliver T Wolf
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Germany
| | - Christian J Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Germany.
| |
Collapse
|
4
|
Ferro A, Arshad A, Boyd L, Stanley T, Berisha A, Vrudhula U, Gomez AM, Borniger JC, Cheadle L. The cytokine receptor Fn14 is a molecular brake on neuronal activity that mediates circadian function in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587786. [PMID: 38617238 PMCID: PMC11014623 DOI: 10.1101/2024.04.02.587786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
To survive, organisms must adapt to a staggering diversity of environmental signals, ranging from sensory information to pathogenic infection, across the lifespan. At the same time, organisms intrinsically generate biological oscillations, such as circadian rhythms, without input from the environment. While the nervous system is well-suited to integrate extrinsic and intrinsic cues, how the brain balances these influences to shape biological function system-wide is not well understood at the molecular level. Here, we demonstrate that the cytokine receptor Fn14, previously identified as a mediator of sensory experience-dependent synaptic refinement during brain development, regulates neuronal activity and function in adult mice in a time-of-day-dependent manner. We show that a subset of excitatory pyramidal (PYR) neurons in the CA1 subregion of the hippocampus increase Fn14 expression when neuronal activity is heightened. Once expressed, Fn14 constrains the activity of these same PYR neurons, suggesting that Fn14 operates as a molecular brake on neuronal activity. Strikingly, differences in PYR neuron activity between mice lacking or expressing Fn14 were most robust at daily transitions between light and dark, and genetic ablation of Fn14 caused aberrations in circadian rhythms, sleep-wake states, and sensory-cued and spatial memory. At the cellular level, microglia contacted fewer, but larger, excitatory synapses in CA1 in the absence of Fn14, suggesting that these brain-resident immune cells may dampen neuronal activity by modifying synaptic inputs onto PYR neurons. Finally, mice lacking Fn14 exhibited heightened susceptibility to chemically induced seizures, implicating Fn14 in disorders characterized by hyperexcitation, such as epilepsy. Altogether, these findings reveal that cytokine receptors that mediates inflammation in the periphery, such as Fn14, can also play major roles in healthy neurological function in the adult brain downstream of both extrinsic and intrinsic cues.
Collapse
Affiliation(s)
- Austin Ferro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Anosha Arshad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
- Department of Neurobiology and Behavior, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA
| | - Leah Boyd
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Tess Stanley
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Adrian Berisha
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Uma Vrudhula
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Adrian M. Gomez
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | | | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11740, USA
| |
Collapse
|
5
|
Hu Y, Lv Y, Long X, Yang G, Zhou J. Melatonin attenuates chronic sleep deprivation-induced cognitive deficits and HDAC3-Bmal1/clock interruption. CNS Neurosci Ther 2024; 30:e14474. [PMID: 37721401 PMCID: PMC10916425 DOI: 10.1111/cns.14474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND AND AIMS Sleep is predicted as a key modulator of cognition, but the underlying mechanisms are poorly understood. In this study, we investigated the effects of melatonin on chronic rapid eye movement sleep deprivation (CRSD)-induced cognitive impairment and circadian dysfunction in rat models. METHODS Thirty-six Sprague-Dawley male rats were divided into three groups: CRSD with saline treatment, CRSD with chronic melatonin injection (20 mg/kg/day), and non-sleep-deprived control. The cognitive behavioral tests as well as the expression of clocks and HDAC3 were evaluated in all groups. RESULTS CRSD significantly reduced recognition index in novel object location, increased escape latency and distance traveling in Morris water maze while melatonin treatment attenuated CRSD-induced hippocampal-dependent spatial learning and memory deficits. Furthermore, the mRNAs of brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1(Bmal1) and circadian locomotor output cycles kaput (Clock) were globally down-regulated by CRSD with constant intrinsic oscillation in both hippocampus and peripheral blood. The protein levels of hippocampal Bmal1, Clock, and HDAC3 were also remarkably down-regulated following CRSD. Melatonin treatment reversed CRSD-induced alterations of Bmal1/Clock and HDAC3 on both mRNA levels and protein levels. CONCLUSIONS Our data indicate that melatonin treatment attenuates CRSD-induced cognitive impairment via regulating HDAC3-Bmal1/Clock interaction. These findings explore a broader understanding of the relationship between sleep and cognition and provide a potential new therapeutic target for cognitive impairment.
Collapse
Affiliation(s)
- Yujie Hu
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
- Department of NeurologyHaikou Affiliated Hospital of Central South University Xiangya School of MedicineHaikouChina
| | - Yefan Lv
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiaoyan Long
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Guoshuai Yang
- Department of NeurologyHaikou Affiliated Hospital of Central South University Xiangya School of MedicineHaikouChina
| | - Jinxia Zhou
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
6
|
Jia X, Song Y, Li Z, Yang N, Liu T, Han D, Sun Z, Shi C, Zhou Y, Shi J, Liu Y, Guo X. Melatonin regulates the circadian rhythm to ameliorate postoperative sleep disorder and neurobehavioral abnormalities in aged mice. CNS Neurosci Ther 2024; 30:e14436. [PMID: 37736695 PMCID: PMC10916446 DOI: 10.1111/cns.14436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 06/07/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Postoperative sleep disorder (PSD) and delirium, which may be associated with surgery and inhalational anesthetics, induce adverse effects in old adults. Emerging evidence indicates that circadian rhythm contributes to various neuropathological diseases, including Alzheimer's disease. Thus, we analyzed the potential role of circadian rhythm in PSD and delirium-like behavior in aged mice and determined whether exogenous melatonin could facilitate entrainment of the circadian rhythm after laparotomy under sevoflurane anesthesia. METHODS We selected old C57BL/6J mice which receiving laparotomy/sevoflurane anesthesia as model animals. We employed buried food, open field, and Y maze test to assess delirium-like behavior, and electroencephalography/electromyography (EEG/EMG) were used to investigate sleep changes. We analyzed the transcription rhythm of clock genes in superchiasmatic nucleus (SCN) to explore the effects of surgery and melatonin pretreatment on the circadian rhythm. Then, we measured melatonin receptor levels in SCN and ERK/CREB pathway-related proteins in hippocampus and prefrontal cortex to assess their role in PSDs and delirium-like behavior. RESULTS Laparotomy under sevoflurane anesthesia had a greater influence than sevoflurane alone, leading to sleep disorder, a shift in sleep-wake rhythm, and delirium-like behavior. Bmal1, Clock, and Cry1 mRNA expression showed a peak shift, MT1 melatonin receptor expression level was increased in the SCN, and p-ERK/ERK and p-CREB/CREB were decreased in hippocampus and prefrontal cortex of aged mice 1 day after laparotomy. Melatonin showed significant efficacy in ameliorating PSD and delirium-like behavior and restoring the circadian rhythm, reversing melatonin receptor and ERK/CREB pathway expression abnormalities. In addition, most of the beneficial effect of melatonin was antagonized by luzindole, a melatonin receptor antagonist. CONCLUSIONS Melatonin receptors in SCN, circadian rhythm, and ERK/CREB signaling pathway participate in the pathophysiological processes of PSD and delirium-like behavior. Melatonin intervention could be a potential preventative approach for PSD and delirium.
Collapse
Affiliation(s)
- Xixi Jia
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Yanan Song
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Zhengqian Li
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Ning Yang
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Taotao Liu
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Dengyang Han
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Zhuonan Sun
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Chengmei Shi
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Yang Zhou
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug DependencePeking UniversityBeijingChina
| | - Yajie Liu
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Xiangyang Guo
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| |
Collapse
|
7
|
Yáñez-Gómez F, Gálvez-Melero L, Ledesma-Corvi S, Bis-Humbert C, Hernández-Hernández E, Salort G, García-Cabrerizo R, García-Fuster MJ. Evaluating the daily modulation of FADD and related molecular markers in different brain regions in male rats. J Neurosci Res 2024; 102:e25296. [PMID: 38361411 DOI: 10.1002/jnr.25296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 02/17/2024]
Abstract
Fas-Associated protein with Death Domain (FADD), a key molecule controlling cell fate by balancing apoptotic versus non-apoptotic functions, is dysregulated in post-mortem brains of subjects with psychopathologies, in animal models capturing certain aspects of these disorders, and by several pharmacological agents. Since persistent disruptions in normal functioning of daily rhythms are linked with these conditions, oscillations over time of key biomarkers, such as FADD, could play a crucial role in balancing the clinical outcome. Therefore, we characterized the 24-h regulation of FADD (and linked molecular partners: p-ERK/t-ERK ratio, Cdk-5, p35/p25, cell proliferation) in key brain regions for FADD regulation (prefrontal cortex, striatum, hippocampus). Samples were collected during Zeitgeber time (ZT) 2, ZT5, ZT8, ZT11, ZT14, ZT17, ZT20, and ZT23 (ZT0, lights-on or inactive period; ZT12, lights-off or active period). FADD showed similar daily fluctuations in all regions analyzed, with higher values during lights off, and opposite to p-ERK/t-ERK ratios regulation. Both Cdk-5 and p35 remained stable and did not change across ZT. However, p25 increased during lights off, but exclusively in striatum. Finally, no 24-h modulation was observed for hippocampal cell proliferation, although higher values were present during lights off. These results demonstrated a clear daily modulation of FADD in several key brain regions, with a more prominent regulation during the active time of rats, and suggested a key role for FADD, and molecular partners, in the normal physiological functioning of the brain's daily rhythmicity, which if disrupted might participate in the development of certain pathologies.
Collapse
Affiliation(s)
- Fernando Yáñez-Gómez
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Medicine, University of the Balearic Islands, Palma, Spain
| | - Laura Gálvez-Melero
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Cristian Bis-Humbert
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Elena Hernández-Hernández
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Glòria Salort
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Rubén García-Cabrerizo
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Medicine, University of the Balearic Islands, Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Medicine, University of the Balearic Islands, Palma, Spain
| |
Collapse
|
8
|
Pracucci E, Graham RT, Alberio L, Nardi G, Cozzolino O, Pillai V, Pasquini G, Saieva L, Walsh D, Landi S, Zhang J, Trevelyan AJ, Ratto GM. Daily rhythm in cortical chloride homeostasis underpins functional changes in visual cortex excitability. Nat Commun 2023; 14:7108. [PMID: 37925453 PMCID: PMC10625537 DOI: 10.1038/s41467-023-42711-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 10/19/2023] [Indexed: 11/06/2023] Open
Abstract
Cortical activity patterns are strongly modulated by fast synaptic inhibition mediated through ionotropic, chloride-conducting receptors. Consequently, chloride homeostasis is ideally placed to regulate activity. We therefore investigated the stability of baseline [Cl-]i in adult mouse neocortex, using in vivo two-photon imaging. We found a two-fold increase in baseline [Cl-]i in layer 2/3 pyramidal neurons, from day to night, with marked effects upon both physiological cortical processing and seizure susceptibility. Importantly, the night-time activity can be converted to the day-time pattern by local inhibition of NKCC1, while inhibition of KCC2 converts day-time [Cl-]i towards night-time levels. Changes in the surface expression and phosphorylation of the cation-chloride cotransporters, NKCC1 and KCC2, matched these pharmacological effects. When we extended the dark period by 4 h, mice remained active, but [Cl-]i was modulated as for animals in normal light cycles. Our data thus demonstrate a daily [Cl-]i modulation with complex effects on cortical excitability.
Collapse
Affiliation(s)
- Enrico Pracucci
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Robert T Graham
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Laura Alberio
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Gabriele Nardi
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Olga Cozzolino
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Vinoshene Pillai
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Giacomo Pasquini
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Luciano Saieva
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Darren Walsh
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Silvia Landi
- Institute of Neuroscience CNR, Pisa, Italy
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Institute of Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
- State Key Laboratory of Chemical Biology. Research Center of Chemical Kinomics, Shangai. Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Andrew J Trevelyan
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| | - Gian-Michele Ratto
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy.
- Institute of Neuroscience CNR, Pisa, Italy.
- Padova Neuroscience Center, Padova, Italy.
| |
Collapse
|
9
|
Bering T, Gadgaard C, Vorum H, Honoré B, Rath MF. Diurnal proteome profile of the mouse cerebral cortex: Conditional deletion of the Bmal1 circadian clock gene elevates astrocyte protein levels and cell abundance in the neocortex and hippocampus. Glia 2023; 71:2623-2641. [PMID: 37470358 DOI: 10.1002/glia.24443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/21/2023]
Abstract
Circadian oscillators, defined by cellular 24 h clock gene rhythms, are found throughout the brain. Cerebral cortex-specific conditional knockout of the clock gene Bmal1 (Bmal1 CKO) leads to depressive-like behavior, but the molecular link from clock gene to altered behavior is unknown. Further, diurnal proteomic data on the cerebral cortex are currently unavailable. With the aim of determining the diurnal proteome profile and downstream targets of the cortical circadian clock, we here performed a proteomic analysis of the mouse cerebral cortex. Proteomics identified approximately 2700 proteins in both the neocortex and the hippocampus. In the neocortex, 15 proteins were differentially expressed (>2-fold) between day and night, mainly mitochondrial and neuronal plasticity proteins. Only three hippocampal proteins were differentially expressed, suggesting that daily protein oscillations are more prominent in the neocortex. The number of differentially expressed proteins was reduced in the Bmal1 CKO, suggesting that daily rhythms in the cerebral cortex are primarily driven by local clocks. The proteome of the Bmal1 CKO cerebral cortex was dominated by upregulated proteins expressed in astrocytes, including GFAP (4-fold) and FABP7 (>20-fold), in both the neocortex and hippocampus. These findings were confirmed at the transcript level. Cellular analyses of astrocyte components revealed an increased number of GFAP-positive cells in the Bmal1 CKO cerebral cortex. Further, BMAL1 was found to be expressed in both GFAP- and FABP7-positive astrocytes of control animals. Our data show that Bmal1 is required for proper cellular composition of the cerebral cortex, suggesting that increased cortical astrocyte activity may induce behavioral changes.
Collapse
Affiliation(s)
- Tenna Bering
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Gadgaard
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Vorum
- Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Bent Honoré
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Martin Fredensborg Rath
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Barone I, Gilette NM, Hawks-Mayer H, Handy J, Zhang KJ, Chifamba FF, Mostafa E, Johnson-Venkatesh EM, Sun Y, Gibson JM, Rotenberg A, Umemori H, Tsai PT, Lipton JO. Synaptic BMAL1 phosphorylation controls circadian hippocampal plasticity. SCIENCE ADVANCES 2023; 9:eadj1010. [PMID: 37878694 PMCID: PMC10599629 DOI: 10.1126/sciadv.adj1010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/25/2023] [Indexed: 10/27/2023]
Abstract
The time of day strongly influences adaptive behaviors like long-term memory, but the correlating synaptic and molecular mechanisms remain unclear. The circadian clock comprises a canonical transcription-translation feedback loop (TTFL) strictly dependent on the BMAL1 transcription factor. We report that BMAL1 rhythmically localizes to hippocampal synapses in a manner dependent on its phosphorylation at Ser42 [pBMAL1(S42)]. pBMAL1(S42) regulates the autophosphorylation of synaptic CaMKIIα and circadian rhythms of CaMKIIα-dependent molecular interactions and LTP but not global rest/activity behavior. Therefore, our results suggest a model in which repurposing of the clock protein BMAL1 to synapses locally gates the circadian timing of plasticity.
Collapse
Affiliation(s)
- Ilaria Barone
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Nicole M. Gilette
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hannah Hawks-Mayer
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jonathan Handy
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Kevin J. Zhang
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Fortunate F. Chifamba
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Engie Mostafa
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Erin M. Johnson-Venkatesh
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Yan Sun
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jennifer M. Gibson
- Departments of Neurology, Neuroscience, Pediatrics, and Psychiatry, University of Texas at Southwestern, Dallas, TX 75390, USA
| | - Alexander Rotenberg
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hisashi Umemori
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Peter T. Tsai
- Departments of Neurology, Neuroscience, Pediatrics, and Psychiatry, University of Texas at Southwestern, Dallas, TX 75390, USA
| | - Jonathan O. Lipton
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurology and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Lee J, Chen S, Monfared RV, Derdeyn P, Leong K, Chang T, Beier K, Baldi P, Alachkar A. Reanalysis of primate brain circadian transcriptomics reveals connectivity-related oscillations. iScience 2023; 26:107810. [PMID: 37752952 PMCID: PMC10518731 DOI: 10.1016/j.isci.2023.107810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/22/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
Research shows that brain circuits controlling vital physiological processes are closely linked with endogenous time-keeping systems. In this study, we aimed to examine oscillatory gene expression patterns of well-characterized neuronal circuits by reanalyzing publicly available transcriptomic data from a spatiotemporal gene expression atlas of a non-human primate. Unexpectedly, brain structures known for regulating circadian processes (e.g., hypothalamic nuclei) did not exhibit robust cycling expression. In contrast, basal ganglia nuclei, not typically associated with circadian physiology, displayed the most dynamic cycling behavior of its genes marked by sharp temporally defined expression peaks. Intriguingly, the mammillary bodies, considered hypothalamic nuclei, exhibited gene expression patterns resembling the basal ganglia, prompting reevaluation of their classification. Our results emphasize the potential for high throughput circadian gene expression analysis to deepen our understanding of the functional synchronization across brain structures that influence physiological processes and resulting complex behaviors.
Collapse
Affiliation(s)
- Justine Lee
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Siwei Chen
- Department of Computer Science, School of Information and Computer Sciences, University of California, Irvine, Irvine, CA, USA
| | - Roudabeh Vakil Monfared
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Pieter Derdeyn
- Mathematical, Computational, and Systems Biology Program, University of California, Irvine, Irvine, CA, USA
| | - Kenneth Leong
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Tiffany Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Kevin Beier
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
- Department of Physiology and Biophysics, School of medicine, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4560, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697-4560, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
| | - Pierre Baldi
- Department of Computer Science, School of Information and Computer Sciences, University of California, Irvine, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, Irvine, CA, USA
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
12
|
Malhan D, Schoenrock B, Yalçin M, Blottner D, Relόgio A. Circadian regulation in aging: Implications for spaceflight and life on earth. Aging Cell 2023; 22:e13935. [PMID: 37493006 PMCID: PMC10497835 DOI: 10.1111/acel.13935] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023] Open
Abstract
Alterations in the circadian system are characteristic of aging on Earth. With the decline in physiological processes due to aging, several health concerns including vision loss, cardiovascular disorders, cognitive impairments, and muscle mass loss arise in elderly populations. Similar health risks are reported as "red flag" risks among astronauts during and after a long-term Space exploration journey. However, little is known about the common molecular alterations underlying terrestrial aging and space-related aging in astronauts, and controversial conclusions have been recently reported. In light of the regulatory role of the circadian clock in the maintenance of human health, we review here the overlapping role of the circadian clock both on aging on Earth and spaceflight with a focus on the four most affected systems: visual, cardiovascular, central nervous, and musculoskeletal systems. In this review, we briefly introduce the regulatory role of the circadian clock in specific cellular processes followed by alterations in those processes due to aging. We next summarize the known molecular alterations associated with spaceflight, highlighting involved clock-regulated genes in space flown Drosophila, nematodes, small mammals, and astronauts. Finally, we discuss common genes that are altered in terms of their expression due to aging on Earth and spaceflight. Altogether, the data elaborated in this review strengthen our hypothesis regarding the timely need to include circadian dysregulation as an emerging hallmark of aging on Earth and beyond.
Collapse
Affiliation(s)
- Deeksha Malhan
- Institute for Systems Medicine and Faculty of Human MedicineMSH Medical School HamburgHamburgGermany
| | - Britt Schoenrock
- Institute of Integrative NeuroanatomyCharité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Müge Yalçin
- Institute for Systems Medicine and Faculty of Human MedicineMSH Medical School HamburgHamburgGermany
- Institute for Theoretical Biology (ITB)Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Dieter Blottner
- Institute of Integrative NeuroanatomyCharité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Neuromuscular System and Neuromuscular SignalingBerlin Center of Space Medicine & Extreme EnvironmentsBerlinGermany
| | - Angela Relόgio
- Institute for Systems Medicine and Faculty of Human MedicineMSH Medical School HamburgHamburgGermany
- Institute for Theoretical Biology (ITB)Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| |
Collapse
|
13
|
Nam H, Kim B, Lee Y, Choe HK, Yu SW. Presenilin 2 N141I Mutation Induces Hyperimmunity by Immune Cell-specific Suppression of REV-ERBα without Altering Central Circadian Rhythm. Exp Neurobiol 2023; 32:259-270. [PMID: 37749927 PMCID: PMC10569138 DOI: 10.5607/en23012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/21/2023] [Accepted: 07/07/2023] [Indexed: 09/27/2023] Open
Abstract
Circadian rhythm is a 24-hour cycle of behavioral and physiological changes. Disrupted sleep-wake patterns and circadian dysfunction are common in patients of Alzheimer Disease (AD) and are closely related with neuroinflammation. However, it is not well known how circadian rhythm of immune cells is altered during the progress of AD. Previously, we found presenilin 2 (Psen2) N141I mutation, one of familial AD (FAD) risk genes, induces hyperimmunity through the epigenetic repression of REV-ERBα expression in microglia and bone marrow-derived macrophage (BMDM) cells. Here, we investigated whether repression of REV-ERBα is associated with dysfunction of immune cell-endogenous or central circadian rhythm by analyses of clock genes expression and cytokine secretion, bioluminescence recording of rhythmic PER2::LUC expression, and monitoring of animal behavioral rhythm. Psen2 N141I mutation down-regulated REV-ERBα and induced selective over-production of IL-6 (a well-known clock-dependent cytokine) following the treatment of toll-like receptor (TLR) ligands in microglia, astrocytes, and BMDM. Psen2 N141I mutation also lowered amplitude of intrinsic daily oscillation in these immune cells representatives of brain and periphery. Of interest, however, the period of daily rhythm remained intact in immune cells. Furthermore, analyses of the central clock and animal behavioral rhythms revealed that central clock remained normal without down-regulation of REV-ERBα. These results suggest that Psen2 N141I mutation induces hyperimmunity mainly through the suppression of REV-ERBα in immune cells, which have lowered amplitude but normal period of rhythmic oscillation. Furthermore, our data reveal that central circadian clock is not affected by Psen2 N141I mutation.
Collapse
Affiliation(s)
- Hyeri Nam
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Boil Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Younghwan Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Han Kyoung Choe
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Seong-Woon Yu
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
14
|
Bering T, Blancas-Velazquez AS, Rath MF. Circadian Clock Genes Are Regulated by Rhythmic Corticosterone at Physiological Levels in the Rat Hippocampus. Neuroendocrinology 2023; 113:1076-1090. [PMID: 37517388 PMCID: PMC10614510 DOI: 10.1159/000533151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/10/2023] [Indexed: 08/01/2023]
Abstract
INTRODUCTION In the hippocampus, clock gene expression is important for memory and mood; however, the signaling mechanism controlling clock gene expression in the hippocampus is unknown. Recent findings suggest that circadian glucocorticoid rhythms driven by the suprachiasmatic nucleus (SCN) control rhythmic clock gene expression in neurons; in addition, dexamethasone modulates hippocampal clock gene expression. We therefore hypothesized that oscillations of clock genes in the hippocampus could be driven by SCN-controlled circadian rhythms in glucocorticoids. METHODS Temporal profiles of hippocampal clock gene expression were established by quantitative reverse-transcription real-time PCR on rat hippocampi, while cellular distribution was established by in situ hybridization. To determine the effect of rhythmic glucocorticoids on hippocampal clock gene expression, the SCN was lesioned, adrenal glands removed and a 24 h exogenous corticosterone rhythm at physiological levels was reestablished by use of a programmable infusion pump. RESULTS Daily rhythms were detected for Per1, Per2, Bmal1, Nr1d1, and Dbp, while clock gene products were confirmed in both the hippocampus proper and the dentate gyrus. In sham controls, differential hippocampal expression of Per1 and Dbp between ZT3 and ZT15 was detectable. This rhythm was abolished by SCN lesion; however, reestablishing the natural rhythm in corticosterone restored differential rhythmic expression of both Per1 and Dbp. Further, a 6 h phase delay in the corticosterone profile caused a predictable shift in expression of Nr1d1. CONCLUSION Our data show that rhythmic corticosterone can drive hippocampal clock gene rhythms suggesting that the SCN regulates the circadian oscillator of the hippocampus by controlling the circadian rhythm in circulating glucocorticoids.
Collapse
Affiliation(s)
- Tenna Bering
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Aurea Susana Blancas-Velazquez
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Martin Fredensborg Rath
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| |
Collapse
|
15
|
Barrio-Alonso E, Lituma PJ, Notaras MJ, Albero R, Bouchekioua Y, Wayland N, Stankovic IN, Jain T, Gao S, Calderon DP, Castillo PE, Colak D. Circadian protein TIMELESS regulates synaptic function and memory by modulating cAMP signaling. Cell Rep 2023; 42:112375. [PMID: 37043347 PMCID: PMC10564971 DOI: 10.1016/j.celrep.2023.112375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
The regulation of neurons by circadian clock genes is thought to contribute to the maintenance of neuronal functions that ultimately underlie animal behavior. However, the impact of specific circadian genes on cellular and molecular mechanisms controlling synaptic plasticity and cognitive function remains elusive. Here, we show that the expression of the circadian protein TIMELESS displays circadian rhythmicity in the mammalian hippocampus. We identify TIMELESS as a chromatin-bound protein that targets synaptic-plasticity-related genes such as phosphodiesterase 4B (Pde4b). By promoting Pde4b transcription, TIMELESS negatively regulates cAMP signaling to modulate AMPA receptor GluA1 function and influence synaptic plasticity. Conditional deletion of Timeless in the adult forebrain impairs working and contextual fear memory in mice. These cognitive phenotypes were accompanied by attenuation of hippocampal Schaffer-collateral synapse long-term potentiation. Together, these data establish a neuron-specific function of mammalian TIMELESS by defining a mechanism that regulates synaptic plasticity and cognitive function.
Collapse
Affiliation(s)
- Estibaliz Barrio-Alonso
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Pablo J Lituma
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Michael J Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Robert Albero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Youcef Bouchekioua
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | - Natalie Wayland
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Isidora N Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Tanya Jain
- Program of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Sijia Gao
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA; Gale & Ira Drukier Institute for Children's Health, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| |
Collapse
|
16
|
Spurny-Dworak B, Reed MB, Handschuh P, Vanicek T, Spies M, Bogner W, Lanzenberger R. The influence of season on glutamate and GABA levels in the healthy human brain investigated by magnetic resonance spectroscopy imaging. Hum Brain Mapp 2023; 44:2654-2663. [PMID: 36840505 PMCID: PMC10028653 DOI: 10.1002/hbm.26236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/22/2023] [Accepted: 02/02/2023] [Indexed: 02/26/2023] Open
Abstract
Seasonal changes in neurotransmitter systems have been demonstrated in imaging studies and are especially noticeable in diseased states such as seasonal affective disorder (SAD). These modulatory neurotransmitters, such as serotonin, are influencing glutamatergic and GABAergic neurotransmission. Furthermore, central components of the circadian pacemaker are regulated by GABA (the suprachiasmatic nucleus) or glutamate (e.g., the retinohypothalamic tract). Therefore, we explored seasonal differences in the GABAergic and glutamatergic system in 159 healthy individuals using magnetic resonance spectroscopy imaging with a GABA-edited 3D-MEGA-LASER sequence at 3T. We quantified GABA+/tCr, GABA+/Glx, and Glx/tCr ratios (GABA+, GABA+ macromolecules; Glx, glutamate + glutamine; tCr, total creatine) in five different subcortical brain regions. Differences between time periods throughout the year, seasonal patterns, and stationarity were tested using ANCOVA models, curve fitting approaches, and unit root and stationarity tests, respectively. Finally, Spearman correlation analyses between neurotransmitter ratios within each brain region and cumulated daylight and global radiation were performed. No seasonal or monthly differences, seasonal patterns, nor significant correlations could be shown in any region or ratio. Unit root and stationarity tests showed stable patterns of GABA+/tCr, GABA+/Glx, and Glx/tCr levels throughout the year, except for hippocampal Glx/tCr. Our results indicate that neurotransmitter levels of glutamate and GABA in healthy individuals are stable throughout the year. Hence, despite the important correction for age and gender in the analyses of MRS derived GABA and glutamate, a correction for seasonality in future studies does not seem necessary. Future investigations in SAD and other psychiatric patients will be of high interest.
Collapse
Affiliation(s)
- B Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - M B Reed
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - P Handschuh
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - T Vanicek
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - M Spies
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - W Bogner
- Department of Biomedical Imaging and Image-Guided Therapy, High Field MR Centre, Medical University of Vienna, Vienna, Austria
| | - R Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Birnie M, Claydon M, Troy O, Flynn B, Yoshimura M, Kershaw Y, Zhao Z, Demski-Allen R, Barker G, Warburton E, Bortolotto Z, Lightman S, Conway-Campbell B. Circadian regulation of hippocampal function is disrupted with corticosteroid treatment. Proc Natl Acad Sci U S A 2023; 120:e2211996120. [PMID: 37023133 PMCID: PMC10104554 DOI: 10.1073/pnas.2211996120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/24/2023] [Indexed: 04/07/2023] Open
Abstract
Disrupted circadian activity is associated with many neuropsychiatric disorders. A major coordinator of circadian biological systems is adrenal glucocorticoid secretion which exhibits a pronounced preawakening peak that regulates metabolic, immune, and cardiovascular processes, as well as mood and cognitive function. Loss of this circadian rhythm during corticosteroid therapy is often associated with memory impairment. Surprisingly, the mechanisms that underlie this deficit are not understood. In this study, in rats, we report that circadian regulation of the hippocampal transcriptome integrates crucial functional networks that link corticosteroid-inducible gene regulation to synaptic plasticity processes via an intrahippocampal circadian transcriptional clock. Further, these circadian hippocampal functions were significantly impacted by corticosteroid treatment delivered in a 5-d oral dosing treatment protocol. Rhythmic expression of the hippocampal transcriptome, as well as the circadian regulation of synaptic plasticity, was misaligned with the natural light/dark circadian-entraining cues, resulting in memory impairment in hippocampal-dependent behavior. These findings provide mechanistic insights into how the transcriptional clock machinery within the hippocampus is influenced by corticosteroid exposure, leading to adverse effects on critical hippocampal functions, as well as identifying a molecular basis for memory deficits in patients treated with long-acting synthetic corticosteroids.
Collapse
Affiliation(s)
- Matthew T. Birnie
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Matthew D. B. Claydon
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Oliver Troy
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Benjamin P. Flynn
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Mitsuhiro Yoshimura
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Yvonne M. Kershaw
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Zidong Zhao
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Rebecca C. R. Demski-Allen
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Gareth R. I. Barker
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - E. Clea Warburton
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Zuner A. Bortolotto
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Stafford L. Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Becky L. Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| |
Collapse
|
18
|
Lodovichi C, Ratto GM. Control of circadian rhythm on cortical excitability and synaptic plasticity. Front Neural Circuits 2023; 17:1099598. [PMID: 37063387 PMCID: PMC10098176 DOI: 10.3389/fncir.2023.1099598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/09/2023] [Indexed: 04/18/2023] Open
Abstract
Living organisms navigate through a cyclic world: activity, feeding, social interactions are all organized along the periodic succession of night and day. At the cellular level, periodic activity is controlled by the molecular machinery driving the circadian regulation of cellular homeostasis. This mechanism adapts cell function to the external environment and its crucial importance is underlined by its robustness and redundancy. The cell autonomous clock regulates cell function by the circadian modulation of mTOR, a master controller of protein synthesis. Importantly, mTOR integrates the circadian modulation with synaptic activity and extracellular signals through a complex signaling network that includes the RAS-ERK pathway. The relationship between mTOR and the circadian clock is bidirectional, since mTOR can feedback on the cellular clock to shift the cycle to maintain the alignment with the environmental conditions. The mTOR and ERK pathways are crucial determinants of synaptic plasticity and function and thus it is not surprising that alterations of the circadian clock cause defective responses to environmental challenges, as witnessed by the bi-directional relationship between brain disorders and impaired circadian regulation. In physiological conditions, the feedback between the intrinsic clock and the mTOR pathway suggests that also synaptic plasticity should undergo circadian regulation.
Collapse
Affiliation(s)
- Claudia Lodovichi
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Padova Neuroscience Center, Universitá degli Studi di Padova, Padova, Italy
| | - Gian Michele Ratto
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
- Padova Neuroscience Center, Universitá degli Studi di Padova, Padova, Italy
- National Enterprise for NanoScience and NanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
19
|
Shimizu K, Inoue KI, Oishi T, Takada M, Fukada Y, Imai H. Diurnal variation in declarative memory and the involvement of SCOP in cognitive functions in nonhuman primates. Mol Brain 2023; 16:31. [PMID: 36966302 PMCID: PMC10039603 DOI: 10.1186/s13041-023-01022-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023] Open
Abstract
Cognitive functions depend on the time of day in various organisms. Previously, we found that 24-h recognition memory performance of nocturnal mice changes diurnally through SCOP protein-dependent regulation. It remains unknown whether diurnal change and SCOP-dependent regulation of memory performance are conserved across species with diurnal/nocturnal habits. We tested whether the memory performance of diurnal Japanese macaques depends on the time of day. The memory association between bitter taste of drinking water and the nozzle color of the water bottle was established during the light period of the day to evaluate of memory performance for macaques. Here we found diurnal variation of declarative memory in Japanese macaques. The middle of the daytime is the most effective time for memory performance during the light period. To assess whether SCOP is involved in declarative memory performance, we interfered with SCOP expression by using lentiviral vector expressing shRNA against Scop in the hippocampus of Japanese macaques. Scop knockdown in the hippocampus abrogated the memory performance in the middle of the daytime. Our results implicate that SCOP in the hippocampus is necessary for the diurnal rhythm of the memory system and that the SCOP-dependent memory regulation system may be conserved in mammals.
Collapse
Affiliation(s)
- Kimiko Shimizu
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan.
- Laboratory of Animal Resources, Graduate School of Medicine, Center for Disease Biology and Integrative Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Takao Oishi
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Yoshitaka Fukada
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.
- Laboratory of Animal Resources, Graduate School of Medicine, Center for Disease Biology and Integrative Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Hiroo Imai
- Molecular Biology Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan.
| |
Collapse
|
20
|
Gonzalez JC, Lee H, Vincent AM, Hill AL, Goode LK, King GD, Gamble KL, Wadiche JI, Overstreet-Wadiche L. Circadian regulation of dentate gyrus excitability mediated by G-protein signaling. Cell Rep 2023; 42:112039. [PMID: 36749664 PMCID: PMC10404305 DOI: 10.1016/j.celrep.2023.112039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/27/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023] Open
Abstract
The central circadian regulator within the suprachiasmatic nucleus transmits time of day information by a diurnal spiking rhythm driven by molecular clock genes controlling membrane excitability. Most brain regions, including the hippocampus, harbor similar intrinsic circadian transcriptional machinery, but whether these molecular programs generate oscillations of membrane properties is unclear. Here, we show that intrinsic excitability of mouse dentate granule neurons exhibits a 24-h oscillation that controls spiking probability. Diurnal changes in excitability are mediated by antiphase G-protein regulation of potassium and sodium currents that reduce excitability during the Light phase. Disruption of the circadian transcriptional machinery by conditional deletion of Bmal1 enhances excitability selectively during the Light phase by removing G-protein regulation. These results reveal that circadian transcriptional machinery regulates intrinsic excitability by coordinated regulation of ion channels by G-protein signaling, highlighting a potential novel mechanism of cell-autonomous oscillations.
Collapse
Affiliation(s)
- Jose Carlos Gonzalez
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Haeun Lee
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Angela M Vincent
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Angela L Hill
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gwendalyn D King
- Department of Biology, Creighton University, Omaha, NE 68178, USA
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jacques I Wadiche
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Linda Overstreet-Wadiche
- Department of Neurobiology and McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
21
|
Yin JCP, Cui E, Hardin PE, Zhou H. Circadian disruption of memory consolidation in Drosophila. Front Syst Neurosci 2023; 17:1129152. [PMID: 37034015 PMCID: PMC10073699 DOI: 10.3389/fnsys.2023.1129152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
The role of the circadian system in memory formation is an important question in neurobiology. Despite this hypothesis being intuitively appealing, the existing data is confusing. Recent work in Drosophila has helped to clarify certain aspects of the problem, but the emerging sense is that the likely mechanisms are more complex than originally conceptualized. In this report, we identify a post-training window of time (during consolidation) when the circadian clock and its components are involved in memory formation. In the broader context, our data suggest that circadian biology might have multiple roles during memory formation. Testing for its roles at multiple timepoints, and in different cells, will be necessary to resolve some of the conflicting data.
Collapse
Affiliation(s)
- Jerry C. P. Yin
- Laboratory of Genetics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- Neurology Department, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
- *Correspondence: Jerry C. P. Yin
| | - Ethan Cui
- Laboratory of Genetics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| | - Paul E. Hardin
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, College Station, TX, United States
| | - Hong Zhou
- Laboratory of Genetics, School of Medicine and Public Health, University of Wisconsin—Madison, Madison, WI, United States
| |
Collapse
|
22
|
Goode LK, Fusilier AR, Remiszewski N, Reeves JM, Abiraman K, Defenderfer M, Paul JR, McMahon LL, Gamble KL. Examination of Diurnal Variation and Sex Differences in Hippocampal Neurophysiology and Spatial Memory. eNeuro 2022; 9:ENEURO.0124-22.2022. [PMID: 36265903 PMCID: PMC9668349 DOI: 10.1523/eneuro.0124-22.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
Circadian rhythms are biological processes that cycle across 24 h and regulate many facets of neurophysiology, including learning and memory. Circadian variation in spatial memory task performance is well documented; however, the effect of sex across circadian time (CT) remains unclear. Additionally, little is known regarding the impact of time-of-day on hippocampal neuronal physiology. Here, we investigated the influence of both sex and time-of-day on hippocampal neurophysiology and memory in mice. Performance on the object location memory (OLM) task depended on both circadian time and sex, with memory enhanced at night in males but during the day in females. Long-term synaptic potentiation (LTP) magnitude at CA3-CA1 synapses was greater at night compared with day in both sexes. Next, we measured spontaneous synaptic excitation and inhibition onto CA1 pyramidal neurons. Frequency and amplitude of inhibition was greater during the day compared with night, regardless of sex. Frequency and amplitude of excitation was larger in females, compared with males, independent of time-of-day, although both time-of-day and sex influenced presynaptic release probability. At night, CA1 pyramidal neurons showed enhanced excitability (action potential firing and/or baseline potential) that was dependent on synaptic excitation and inhibition, regardless of sex. This study emphasizes the importance of sex and time-of-day in hippocampal physiology, especially given that many neurologic disorders impacting the hippocampus are linked to circadian disruption and present differently in men and women. Knowledge about how sex and circadian rhythms affect hippocampal physiology can improve the translational relevancy of therapeutics and inform the appropriate timing of existing treatments.
Collapse
Affiliation(s)
- Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham 35233, AL
| | - Allison R Fusilier
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham 35233, AL
| | - Natalie Remiszewski
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham 35233, AL
| | - Jacob M Reeves
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham 35233, AL
| | | | - Matthew Defenderfer
- Research Computing, Information Technology, University of Alabama at Birmingham, Birmingham 35233, AL
| | - Jodi R Paul
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham 35233, AL
| | - Lori L McMahon
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham 35233, AL
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham 35233, AL
| |
Collapse
|
23
|
Deibel SH, Higdon S, Cassell TTS, House-Denine ML, Giberson E, Webb IC, Thorpe CM. Impaired Morris water task retention following T21 light dark cycle exposure is not due to reduced hippocampal c-FOS expression. Front Behav Neurosci 2022; 16:1025388. [PMID: 36311860 PMCID: PMC9596763 DOI: 10.3389/fnbeh.2022.1025388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/09/2022] [Indexed: 11/21/2022] Open
Abstract
Circadian rhythms influence virtually all aspects of physiology and behavior. This is problematic when circadian rhythms no longer reliably predict time. Circadian rhythm disruption can impair memory, yet we don’t know how this fully works at the systems and molecular level. When trying to determine the root of a memory impairment, assessing neuronal activation with c-FOS is useful. This has yet to be assessed in the hippocampi of circadian rhythm disrupted rats in a hippocampal gold standard task. Rats were trained on the Morris water task (MWT), then received 6 days of a 21-h day (T21), 13 days of a normal light dark cycle, probe trial, and tissue extraction an hour later. Despite having impaired memory in the probe trial, compared to controls there were no differences in c-FOS expression in hippocampal sub regions: CA1; CA3; Dentate gyrus. These data confirm others in hamsters demonstrating that arrhythmicity which produces an impairment in spontaneous alternation does not affect c-FOS in the dentate gyrus. The current study indicates that the memory impairment induced by a lighting manipulation is likely not due to attenuated neuronal activation. Determining how the master clock in the brain communicates with the hippocampus is needed to untangle the relationship between circadian rhythms and memory.
Collapse
Affiliation(s)
- Scott H. Deibel
- Department of Psychology, University of New Brunswick, Fredericton, NB, Canada
- *Correspondence: Scott H. Deibel,
| | - S. Higdon
- Department of Psychology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - T. T. S. Cassell
- Department of Psychology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - M. L. House-Denine
- Department of Psychology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - E. Giberson
- Department of Psychology, University of New Brunswick, Fredericton, NB, Canada
| | - I. C. Webb
- Department of Psychology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - C. M. Thorpe
- Department of Psychology, Memorial University of Newfoundland, St. John’s, NL, Canada
| |
Collapse
|
24
|
Valeri J, O’Donovan SM, Wang W, Sinclair D, Bollavarapu R, Gisabella B, Platt D, Stockmeier C, Pantazopoulos H. Altered expression of somatostatin signaling molecules and clock genes in the hippocampus of subjects with substance use disorder. Front Neurosci 2022; 16:903941. [PMID: 36161151 PMCID: PMC9489843 DOI: 10.3389/fnins.2022.903941] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Substance use disorders are a debilitating group of psychiatric disorders with a high degree of comorbidity with major depressive disorder. Sleep and circadian rhythm disturbances are commonly reported in people with substance use disorder and major depression and associated with increased risk of relapse. Hippocampal somatostatin signaling is involved in encoding and consolidation of contextual memories which contribute to relapse in substance use disorder. Somatostatin and clock genes also have been implicated in depression, suggesting that these molecules may represent key converging pathways involved in contextual memory processing in substance use and major depression. We used hippocampal tissue from a cohort of subjects with substance use disorder (n = 20), subjects with major depression (n = 20), subjects with comorbid substance use disorder and major depression (n = 24) and psychiatrically normal control subjects (n = 20) to test the hypothesis that expression of genes involved in somatostatin signaling and clock genes is altered in subjects with substance use disorder. We identified decreased expression of somatostatin in subjects with substance use disorder and in subjects with major depression. We also observed increased somatostatin receptor 2 expression in subjects with substance use disorder with alcohol in the blood at death and decreased expression in subjects with major depression. Expression of the clock genes Arntl, Nr1d1, Per2 and Cry2 was increased in subjects with substance use disorder. Arntl and Nr1d1 expression in comparison was decreased in subjects with major depression. We observed decreased expression of Gsk3β in subjects with substance use disorder. Subjects with comorbid substance use disorder and major depression displayed minimal changes across all outcome measures. Furthermore, we observed a significant increase in history of sleep disturbances in subjects with substance use disorder. Our findings represent the first evidence for altered somatostatin and clock gene expression in the hippocampus of subjects with substance use disorder and subjects with major depression. Altered expression of these molecules may impact memory consolidation and contribute to relapse risk.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Sinead M. O’Donovan
- Department of Neuroscience, University of Toledo Medical Center, Toledo, OH, United States
| | - Wei Wang
- Department of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, United States
| | - David Sinclair
- Department of Neuroscience, University of Toledo Medical Center, Toledo, OH, United States
| | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Donna Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Craig Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
- *Correspondence: Harry Pantazopoulos,
| |
Collapse
|
25
|
Deibel SH, Lewis LM, Cleary J, Cassell TTS, Skinner DM, Thorpe CM. Unpredictable mealtimes rather than social jetlag affects acquisition and retention of hippocampal dependent memory. Behav Processes 2022; 201:104704. [PMID: 35842197 DOI: 10.1016/j.beproc.2022.104704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/15/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022]
Abstract
Some degree of circadian rhythm disruption is hard to avoid in today's society. Along, with many other deleterious effects, circadian rhythm disruption impairs memory. One way to study this is to expose rats to daylengths that are outside the range of entrainment. As a result, circadian processes and behaviours occur during phases of the light dark cycle in which they typically would not. Even brief exposures to these day lengths can impair hippocampal dependent memory. In a recent report, we created an unentrainable light dark cycle that was intended to resemble aspects of social jetlag. As predictable mealtime impacts circadian entrainment, in that report, we also created an unpredictable meal schedule with the idea that failure to entrain to a meal might afford a disadvantage in some instances. Both of these manipulations impaired retention in a spatial water plus-maze task. Using the same manipulations, the present study investigated their effects on acquisition in distributed and massed spatial water plus-maze paradigms. As in other reports with unentrainable daylengths, acquisition was not affected by our lighting manipulation. Conversely, in accordance with our past report, unpredictable mealtimes had a harmful effect on hippocampal dependent memory. Notably, impaired acquisition in the distributed version, and impaired retention in the massed version. In tandem, these data suggest that failure to consolidate or retrieve the information is the likely culprit. The unpredictable mealtime manipulation offers a unique opportunity to study the effects of circadian entrainment on memory.
Collapse
Affiliation(s)
- Scott H Deibel
- Department of Psychology, University of New Brunswick, Canada.
| | - Leanna M Lewis
- Department of Psychology, Memorial University of Newfoundland, Canada
| | - Jillian Cleary
- Department of Psychology, Memorial University of Newfoundland, Canada
| | | | - Darlene M Skinner
- Department of Psychology, Memorial University of Newfoundland, Canada
| | | |
Collapse
|
26
|
Gessner N, Shinbashi M, Chuluun B, Heller C, Pittaras E. Handling, task complexity, time-of-day, and sleep deprivation as dynamic modulators of recognition memory in mice. Physiol Behav 2022; 251:113803. [PMID: 35398333 DOI: 10.1016/j.physbeh.2022.113803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/25/2022]
Abstract
Sleep is essential for optimal cognitive functioning. Although we lack a complete understanding of the role of sleep in memory consolidation, we know that various factors that disturb sleep or sleep quality have consequences for cognitive performance. Such factors can be unintended components of behavioral experiments on rodents and other experimental animals that generate differing results from different labs. These experimental variables include habituation to handling, intended or unintended sleep deprivation, task complexity, time of testing, and environmental features. We have examined how these variables impact recognition memory in C57BL/6 mice. Handled mice outperformed their non-handled counterparts across different combinations of delay phase duration and lighting conditions. Results also suggest that simple task recall is more resistant to diurnal variation and the impairing effects of sleep deprivation than is complex task recall. This study underscores the role of protocol and environmental factors in recognition memory and in conflicting results from different laboratories.
Collapse
Affiliation(s)
- Nicholas Gessner
- Stanford University Department of Biology, 371 Jane Stanford Way, Stanford, CA 94305-5020
| | - Meagan Shinbashi
- Stanford University Department of Biology, 371 Jane Stanford Way, Stanford, CA 94305-5020
| | - Bayarsaikhan Chuluun
- Stanford University Department of Biology, 371 Jane Stanford Way, Stanford, CA 94305-5020
| | - Craig Heller
- Stanford University Department of Biology, 371 Jane Stanford Way, Stanford, CA 94305-5020
| | - Elsa Pittaras
- Stanford University Department of Biology, 371 Jane Stanford Way, Stanford, CA 94305-5020.
| |
Collapse
|
27
|
Circadian regulation of memory under stress: Endocannabinoids matter. Neurosci Biobehav Rev 2022; 138:104712. [PMID: 35643119 DOI: 10.1016/j.neubiorev.2022.104712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/27/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022]
Abstract
Organisms ranging from plants to higher mammals have developed 24-hour oscillation rhythms to optimize physiology to environmental changes and regulate a plethora of neuroendocrine and behavioral processes, including neurotransmitter and hormone regulation, stress response and learning and memory function. Compelling evidence indicates that a wide array of memory processes is strongly influenced by stress- and emotional arousal-activated neurobiological systems, including the endocannabinoid system which has been extensively shown to play an integral role in mediating stress effects on memory. Here, we review findings showing how circadian rhythms and time-of-day influence stress systems and memory performance. We report evidence of circadian regulation of memory under stress, focusing on the role of the endocannabinoid system and highlighting its circadian rhythmicity. Our discussion illustrates how the endocannabinoid system mediates stress effects on memory in a circadian-dependent fashion. We suggest that endocannabinoids might regulate molecular mechanisms that control memory function under circadian and stress influence, with potential important clinical implications for both neurodevelopmental disorders and psychiatric conditions involving memory impairments.
Collapse
|
28
|
Hoyt KR, Obrietan K. Circadian clocks, cognition, and Alzheimer's disease: synaptic mechanisms, signaling effectors, and chronotherapeutics. Mol Neurodegener 2022; 17:35. [PMID: 35525980 PMCID: PMC9078023 DOI: 10.1186/s13024-022-00537-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/08/2022] [Indexed: 12/20/2022] Open
Abstract
Modulation of basic biochemical and physiological processes by the circadian timing system is now recognized as a fundamental feature of all mammalian organ systems. Within the central nervous system, these clock-modulating effects are reflected in some of the most complex behavioral states including learning, memory, and mood. How the clock shapes these behavioral processes is only now beginning to be realized. In this review we describe recent findings regarding the complex set of cellular signaling events, including kinase pathways, gene networks, and synaptic circuits that are under the influence of the clock timing system and how this, in turn, shapes cognitive capacity over the circadian cycle. Further, we discuss the functional roles of the master circadian clock located in the suprachiasmatic nucleus, and peripheral oscillator populations within cortical and limbic circuits, in the gating of synaptic plasticity and memory over the circadian cycle. These findings are then used as the basis to discuss the connection between clock dysregulation and cognitive impairments resulting from Alzheimer's disease (AD). In addition, we discuss the conceptually novel idea that in AD, there is a selective disruption of circadian timing within cortical and limbic circuits, and that it is the disruption/desynchronization of these regions from the phase-entraining effects of the SCN that underlies aspects of the early- and mid-stage cognitive deficits in AD. Further, we discuss the prospect that the disruption of circadian timing in AD could produce a self-reinforcing feedback loop, where disruption of timing accelerates AD pathogenesis (e.g., amyloid deposition, oxidative stress and cell death) that in turn leads to a further disruption of the circadian timing system. Lastly, we address potential therapeutic approaches that could be used to strengthen cellular timing networks and, in turn, how these approaches could be used to improve cognitive capacity in Alzheimer's patients.
Collapse
Affiliation(s)
- Kari R Hoyt
- Division of Pharmaceutics and Pharmacology, Ohio State University, 412 Riffe Building, 12th Ave, Columbus, OH, 43210, USA.
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Graves Hall, 333 W. 10th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
29
|
Diurnal rhythm regulates the frequency of carbachol-induced beta oscillation via inhibitory neural system in rat hippocampus. Cogn Neurodyn 2021; 16:507-518. [DOI: 10.1007/s11571-021-09736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/08/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022] Open
|
30
|
Urban MW, Lo C, Bodinayake KK, Brunswick CA, Murakami S, Heimann AC, Kwapis JL. The circadian clock gene Per1 modulates context fear memory formation within the retrosplenial cortex in a sex-specific manner. Neurobiol Learn Mem 2021; 185:107535. [PMID: 34624524 PMCID: PMC8595856 DOI: 10.1016/j.nlm.2021.107535] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 01/18/2023]
Abstract
Context memory formation is a complex process that requires transcription in many subregions of the brain including the dorsal hippocampus and retrosplenial cortex. One critical gene necessary for memory formation is the circadian gene Period1 (Per1), which has been shown to function in the dorsal hippocampus to modulate spatial memory in addition to its well-documented role in regulating the diurnal clock within the suprachiasmatic nucleus (SCN). We recently found that alterations in Per1 expression in the dorsal hippocampus can modulate spatial memory formation, with reduced hippocampal Per1 impairing memory and overexpression of Per1 ameliorating age-related impairments in spatial memory. Whether Per1 similarly functions within other memory-relevant brain regions is currently unknown. Here, to test whether Per1 is a general mechanism that modulates memory across the brain, we tested the role of Per1 in the retrosplenial cortex (RSC), a brain region necessary for context memory formation. First, we demonstrate that context fear conditioning drives a transient increase in Per1 mRNA expression within the anterior RSC that peaks 60 m after training. Next, using HSV-CRISPRi-mediated knockdown of Per1, we show that reducing Per1 within the anterior RSC before context fear acquisition impairs memory in both male and female mice. In contrast, overexpressing Per1 with either HSV-CRISPRa or HSV-Per1 before context fear acquisition drives a sex-specific memory impairment; males show impaired context fear memory whereas females are not affected by Per1 overexpression. Finally, as Per1 levels are known to rhythmically oscillate across the day/night cycle, we tested the possibility that Per1 overexpression might have different effects on memory depending on the time of day. In contrast to the impairment in memory we observed during the daytime, Per1 overexpression has no effect on context fear memory during the night in either male or female mice. Together, our results indicate that Per1 modulates memory in the anterior retrosplenial cortex in addition to its documented role in regulating memory within the dorsal hippocampus, although this role may differ between males and females.
Collapse
Affiliation(s)
- Mark W Urban
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Chenyu Lo
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Kasuni K Bodinayake
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Chad A Brunswick
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Shoko Murakami
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Ashley C Heimann
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
31
|
Asadian N, Parsaie H, Vafaei AA, Dadkhah M, Omoumi S, Sedaghat K. Chronic light deprivation induces different effects on spatial and fear memory and hippocampal BDNF/TRKB expression during light and dark phases of rat diurnal rhythm. Behav Brain Res 2021; 418:113638. [PMID: 34695541 DOI: 10.1016/j.bbr.2021.113638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 09/11/2021] [Accepted: 10/18/2021] [Indexed: 11/02/2022]
Abstract
Disruptions in light/dark cycle have been associated with an altered ability to form and retrieve memory in human and animals. Animal studies have shown that chronic light deprivation disrupts the light/dark cycle and alters the neural connections that mediate hippocampal memory formation. In order to better understand how light deprivation affects the formation and retrieval of memory in adult rats, we examined the effect of total darkness on spatial and auditory fear learning and memory formation and BDNF/TRKB protein levels during the light and dark phases of the rat circadian cycle. Male Wistar rats (n = 60), were randomly divided into two main groups: normal rearing (NR, 12 h light/dark cycle for 3 weeks) and dark rearing (DR, kept in constant darkness for 3 weeks); and each of these groups had a "light (day)" and "dark (night)" sub-group. After 3 weeks, the Morris Water maze and auditory fear conditioning were used to assess spatial and fear memory acquisition and retrieval, respectively. BDNF and TRKB protein levels in the hippocampus of rats from the four sub-groups were measured by Western blot, at the completion of the 3 week constant darkness exposure and after the behavioral experiments. These studies revealed that DR for 3 weeks impaired spatial memory retrieval and enhanced extinction of auditory fear memory specifically during the light (day) phase. DR also eliminated the normal fluctuations in BDNF/TRKB levels observed in the hippocampus across the light/dark cycle.
Collapse
Affiliation(s)
- Nader Asadian
- Department of Biophysics, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Houman Parsaie
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Masoumeh Dadkhah
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Samira Omoumi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Katayoun Sedaghat
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
32
|
Spatio-temporal heterogeneity in hippocampal metabolism in control and epilepsy conditions. Proc Natl Acad Sci U S A 2021; 118:2013972118. [PMID: 33692123 DOI: 10.1073/pnas.2013972118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The hippocampus's dorsal and ventral parts are involved in different operative circuits, the functions of which vary in time during the night and day cycle. These functions are altered in epilepsy. Since energy production is tailored to function, we hypothesized that energy production would be space- and time-dependent in the hippocampus and that such an organizing principle would be modified in epilepsy. Using metabolic imaging and metabolite sensing ex vivo, we show that the ventral hippocampus favors aerobic glycolysis over oxidative phosphorylation as compared to the dorsal part in the morning in control mice. In the afternoon, aerobic glycolysis is decreased and oxidative phosphorylation increased. In the dorsal hippocampus, the metabolic activity varies less between these two times but is weaker than in the ventral. Thus, the energy metabolism is different along the dorsoventral axis and changes as a function of time in control mice. In an experimental model of epilepsy, we find a large alteration of such spatiotemporal organization. In addition to a general hypometabolic state, the dorsoventral difference disappears in the morning, when seizure probability is low. In the afternoon, when seizure probability is high, the aerobic glycolysis is enhanced in both parts, the increase being stronger in the ventral area. We suggest that energy metabolism is tailored to the functions performed by brain networks, which vary over time. In pathological conditions, the alterations of these general rules may contribute to network dysfunctions.
Collapse
|
33
|
Salehinejad MA, Wischnewski M, Ghanavati E, Mosayebi-Samani M, Kuo MF, Nitsche MA. Cognitive functions and underlying parameters of human brain physiology are associated with chronotype. Nat Commun 2021; 12:4672. [PMID: 34344864 PMCID: PMC8333420 DOI: 10.1038/s41467-021-24885-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/08/2021] [Indexed: 01/03/2023] Open
Abstract
Circadian rhythms have natural relative variations among humans known as chronotype. Chronotype or being a morning or evening person, has a specific physiological, behavioural, and also genetic manifestation. Whether and how chronotype modulates human brain physiology and cognition is, however, not well understood. Here we examine how cortical excitability, neuroplasticity, and cognition are associated with chronotype in early and late chronotype individuals. We monitor motor cortical excitability, brain stimulation-induced neuroplasticity, and examine motor learning and cognitive functions at circadian-preferred and non-preferred times of day in 32 individuals. Motor learning and cognitive performance (working memory, and attention) along with their electrophysiological components are significantly enhanced at the circadian-preferred, compared to the non-preferred time. This outperformance is associated with enhanced cortical excitability (prominent cortical facilitation, diminished cortical inhibition), and long-term potentiation/depression-like plasticity. Our data show convergent findings of how chronotype can modulate human brain functions from basic physiological mechanisms to behaviour and higher-order cognition.
Collapse
Affiliation(s)
- Mohammad Ali Salehinejad
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Miles Wischnewski
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Elham Ghanavati
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Department of Psychology, Ruhr-University Bochum, Bochum, Germany
| | - Mohsen Mosayebi-Samani
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Min-Fang Kuo
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Michael A Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany.
- Department of Neurology, University Medical Hospital Bergmannsheil, Bochum, Germany.
| |
Collapse
|
34
|
Tandoc MC, Bayda M, Poskanzer C, Cho E, Cox R, Stickgold R, Schapiro AC. Examining the effects of time of day and sleep on generalization. PLoS One 2021; 16:e0255423. [PMID: 34339459 PMCID: PMC8328323 DOI: 10.1371/journal.pone.0255423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/16/2021] [Indexed: 12/26/2022] Open
Abstract
Extracting shared structure across our experiences allows us to generalize our knowledge to novel contexts. How do different brain states influence this ability to generalize? Using a novel category learning paradigm, we assess the effect of both sleep and time of day on generalization that depends on the flexible integration of recent information. Counter to our expectations, we found no evidence that this form of generalization is better after a night of sleep relative to a day awake. Instead, we observed an effect of time of day, with better generalization in the morning than the evening. This effect also manifested as increased false memory for generalized information. In a nap experiment, we found that generalization did not benefit from having slept recently, suggesting a role for time of day apart from sleep. In follow-up experiments, we were unable to replicate the time of day effect for reasons that may relate to changes in category structure and task engagement. Despite this lack of consistency, we found a morning benefit for generalization when analyzing all the data from experiments with matched protocols (n = 136). We suggest that a state of lowered inhibition in the morning may facilitate spreading activation between otherwise separate memories, promoting this form of generalization.
Collapse
Affiliation(s)
- Marlie C. Tandoc
- Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mollie Bayda
- Department of Psychiatry, Beth Israel Deaconess Medical Center / Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Psychology, University of California-Los Angeles, Los Angeles, California, United States of America
| | - Craig Poskanzer
- Department of Psychiatry, Beth Israel Deaconess Medical Center / Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Eileen Cho
- Department of Psychiatry, Beth Israel Deaconess Medical Center / Harvard Medical School, Boston, Massachusetts, United States of America
| | - Roy Cox
- Department of Psychiatry, Beth Israel Deaconess Medical Center / Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Robert Stickgold
- Department of Psychiatry, Beth Israel Deaconess Medical Center / Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anna C. Schapiro
- Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Psychiatry, Beth Israel Deaconess Medical Center / Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
35
|
Fusilier AR, Davis JA, Paul JR, Yates SD, McMeekin LJ, Goode LK, Mokashi MV, Remiszewski N, van Groen T, Cowell RM, McMahon LL, Roberson ED, Gamble KL. Dysregulated clock gene expression and abnormal diurnal regulation of hippocampal inhibitory transmission and spatial memory in amyloid precursor protein transgenic mice. Neurobiol Dis 2021; 158:105454. [PMID: 34333153 PMCID: PMC8477442 DOI: 10.1016/j.nbd.2021.105454] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 11/27/2022] Open
Abstract
Patients with Alzheimer's disease (AD) often have fragmentation of sleep/wake cycles and disrupted 24-h (circadian) activity. Despite this, little work has investigated the potential underlying day/night disruptions in cognition and neuronal physiology in the hippocampus. The molecular clock, an intrinsic transcription-translation feedback loop that regulates circadian behavior, may also regulate hippocampal neurophysiological activity. We hypothesized that disrupted diurnal variation in clock gene expression in the hippocampus corresponds with loss of normal day/night differences in membrane excitability, synaptic physiology, and cognition. We previously reported disrupted circadian locomotor rhythms and neurophysiological output of the suprachiasmatic nucleus (the primary circadian clock) in Tg-SwDI mice with human amyloid-beta precursor protein mutations. Here, we report that Tg-SwDI mice failed to show day/night differences in a spatial working memory task, unlike wild-type controls that exhibited enhanced spatial working memory at night. Moreover, Tg-SwDI mice had lower levels of Per2, one of the core components of the molecular clock, at both mRNA and protein levels when compared to age-matched controls. Interestingly, we discovered neurophysiological impairments in area CA1 of the Tg-SwDI hippocampus. In controls, spontaneous inhibitory post-synaptic currents (sIPSCs) in pyramidal cells showed greater amplitude and lower inter-event interval during the day than the night. However, the normal day/night differences in sIPSCs were absent (amplitude) or reversed (inter-event interval) in pyramidal cells from Tg-SwDI mice. In control mice, current injection into CA1 pyramidal cells produced more firing during the night than during the day, but no day/night difference in excitability was observed in Tg-SwDI mice. The normal day/night difference in excitability in controls was blocked by GABA receptor inhibition. Together, these results demonstrate that the normal diurnal regulation of inhibitory transmission in the hippocampus is diminished in a mouse model of AD, leading to decreased daytime inhibition onto hippocampal CA1 pyramidal cells. Uncovering disrupted day/night differences in circadian gene regulation, hippocampal physiology, and memory in AD mouse models may provide insight into possible chronotherapeutic strategies to ameliorate Alzheimer's disease symptoms or delay pathological onset.
Collapse
Affiliation(s)
- Allison R Fusilier
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennifer A Davis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jodi R Paul
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefani D Yates
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Laura J McMeekin
- Department of Cell, Developmental, & Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neuroscience, Southern Research, Birmingham, AL 35205, USA
| | - Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mugdha V Mokashi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Natalie Remiszewski
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Thomas van Groen
- Department of Cell, Developmental, & Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rita M Cowell
- Department of Cell, Developmental, & Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neuroscience, Southern Research, Birmingham, AL 35205, USA
| | - Lori L McMahon
- Department of Cell, Developmental, & Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
36
|
Zheng Y, Zhang YM, Tang ZS, Du JK, Guo DW, Xu YJ, Sheng H, Lu JQ, Ni X. Spatial learning and memory deficits induced by prenatal glucocorticoid exposure depend on hippocampal CRHR1 and CXCL5 signaling in rats. J Neuroinflammation 2021; 18:85. [PMID: 33810797 PMCID: PMC8019183 DOI: 10.1186/s12974-021-02129-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background Prenatal synthetic glucocorticoid (sGC) exposure increases the susceptibility to cognitive and affective disorders in postnatal life. We previously demonstrated that prenatal sGC exposure results in an increase in corticotropin-releasing hormone (CRH) receptor type 1 (CRHR1) expression in the hippocampus of rats, and CRHR1 is involved in synapse formation via regulation of C-X-C chemokine ligand 5 (CXCL5) in hippocampus. We sought to investigate that the roles of CRHR1 and CXCL5 in learning and memory impairment caused by prenatal sGC exposure. Methods Pregnant rats were administered with saline or dexamethasone (DEX) from gestational day (GD) 14 to GD21. DEX offspring at 2-day old were treated with saline and CRHR1 antagonists (antalarmin and CP154526) for 7 days. Some DEX offspring received intra-hippocampal injection of AAV9 carrying CXCL5 gene. Spatial learning and memory was assessed by Morris water maze test. Immunofluorescence analysis was applied to show synapsin I and PSD95 signals in hippocampus. Synapsin I and PSD95 protein level and CXCL5 concentration were determined by western blotting and ELISA, respectively. Organotypic hippocampal slice cultures were used to investigate the effect of DEX on CXCL5 production in vitro. Results Both male and female DEX offspring displayed impairment of spatial learning and memory in adulthood. Synapsin I and PSD95 signals and CXCL5 levels were decreased in DEX offspring. DEX offspring with antalarmin and CP154526 treatment showed improved spatial learning and memory. Antalarmin and CP154526 treatment increased synapsin I and PSD95 signals and CXCL5 concentration in hippocampus. Bilaterally hippocampal injection of AAV9 carrying CXCL5 gene improved the spatial learning and memory and increased CXCL5 concentration and synapsin I and PSD95 levels in hippocampus. DEX dose-dependently suppressed CXCL5 production in cultured hippocammpal slices, which was prevented by antalarmin treatment. Conclusion CRHR1 and CXCL5 signaling in the hippocampus are involved in spatial learning and memory deficits caused by prenatal DEX exposure. CRHR1 activation contributes to decreased CXCL5 production in hippocampus induced by prenatal DEX treatment. Our study provides a molecular basis of prenatal GC exposure programming spatial learning and memory.
Collapse
Affiliation(s)
- You Zheng
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China.,Department of Physiology, Navy Medical University, Shanghai, 200433, China.,Laboratory of Cell Engineering, Institute of Biotechnology, Beijing, 100071, China
| | - Yan-Min Zhang
- Department of Physiology, Navy Medical University, Shanghai, 200433, China.,School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Zheng-Shan Tang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Jian-Kui Du
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China
| | - De-Wei Guo
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Yong-Jun Xu
- Department of Physiology, Navy Medical University, Shanghai, 200433, China
| | - Hui Sheng
- Department of Physiology, Navy Medical University, Shanghai, 200433, China
| | - Jian-Qiang Lu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Xin Ni
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China. .,Department of Physiology, Navy Medical University, Shanghai, 200433, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China.
| |
Collapse
|
37
|
Buijink MR, Michel S. A multi-level assessment of the bidirectional relationship between aging and the circadian clock. J Neurochem 2021; 157:73-94. [PMID: 33370457 PMCID: PMC8048448 DOI: 10.1111/jnc.15286] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
The daily temporal order of physiological processes and behavior contribute to the wellbeing of many organisms including humans. The central circadian clock, which coordinates the timing within our body, is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Like in other parts of the brain, aging impairs the SCN function, which in turn promotes the development and progression of aging-related diseases. We here review the impact of aging on the different levels of the circadian clock machinery-from molecules to organs-with a focus on the role of the SCN. We find that the molecular clock is less effected by aging compared to other cellular components of the clock. Proper rhythmic regulation of intracellular signaling, ion channels and neuronal excitability of SCN neurons are greatly disturbed in aging. This suggests a disconnection between the molecular clock and the electrophysiology of these cells. The neuronal network of the SCN is able to compensate for some of these cellular deficits. However, it still results in a clear reduction in the amplitude of the SCN electrical rhythm, suggesting a weakening of the output timing signal. Consequently, other brain areas and organs not only show aging-related deficits in their own local clocks, but also receive a weaker systemic timing signal. The negative spiral completes with the weakening of positive feedback from the periphery to the SCN. Consequently, chronotherapeutic interventions should aim at strengthening overall synchrony in the circadian system using life-style and/or pharmacological approaches.
Collapse
Affiliation(s)
- M. Renate Buijink
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Stephan Michel
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
38
|
Lehr AB, Kumar A, Tetzlaff C, Hafting T, Fyhn M, Stöber TM. CA2 beyond social memory: Evidence for a fundamental role in hippocampal information processing. Neurosci Biobehav Rev 2021; 126:398-412. [PMID: 33775693 DOI: 10.1016/j.neubiorev.2021.03.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 01/16/2023]
Abstract
Hippocampal region CA2 has received increased attention due to its importance in social recognition memory. While its specific function remains to be identified, there are indications that CA2 plays a major role in a variety of situations, widely extending beyond social memory. In this targeted review, we highlight lines of research which have begun to converge on a more fundamental role for CA2 in hippocampus-dependent memory processing. We discuss recent proposals that speak to the computations CA2 may perform within the hippocampal circuit.
Collapse
Affiliation(s)
- Andrew B Lehr
- Department of Computational Neuroscience, University of Göttingen, Germany; Bernstein Center for Computational Neuroscience, University of Göttingen, Germany; Department of Computational Physiology, Simula Research Laboratory, Lysaker, Norway; Centre for Integrative Neuroplasticity, University of Oslo, Norway.
| | - Arvind Kumar
- Department of Computational Science and Technology, KTH Royal Institute of Technology, Sweden
| | - Christian Tetzlaff
- Department of Computational Neuroscience, University of Göttingen, Germany; Bernstein Center for Computational Neuroscience, University of Göttingen, Germany
| | - Torkel Hafting
- Centre for Integrative Neuroplasticity, University of Oslo, Norway; Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Marianne Fyhn
- Centre for Integrative Neuroplasticity, University of Oslo, Norway; Department of Biosciences, University of Oslo, Norway
| | - Tristan M Stöber
- Department of Computational Physiology, Simula Research Laboratory, Lysaker, Norway; Centre for Integrative Neuroplasticity, University of Oslo, Norway; Department of Informatics, University of Oslo, Norway.
| |
Collapse
|
39
|
Zhang Y, Zhou Y, Zhang X, Wang L, Zhong Y. Clock neurons gate memory extinction in Drosophila. Curr Biol 2021; 31:1337-1343.e4. [PMID: 33545046 DOI: 10.1016/j.cub.2021.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/15/2020] [Accepted: 01/06/2021] [Indexed: 11/25/2022]
Abstract
Memory forms when a previously neutral stimulus (CS+) becomes competent to predict a biologically potent stimulus (US). However, if the CS+ is repeatedly presented without the US after the memory formation, this memory will be suppressed by newly formed extinction memory.1,2 The striking feature of extinction learning is that it requires repeated trials to robustly form extinction. Extended repetition only yields memories that remain transient in nature,3 thus imposing challenges in understanding the underlying mechanisms of extinction learning. Here, we took advantage of the versatile genetic tools4 and the well-characterized circadian system of Drosophila5,6 to link these unique features to clock neurons. We report that inhibiting the activity of clock neurons blocks the formation of extinction memory. Further investigation attributes this role to a subset of cryptochrome-positive dorsal neurons 1 (DN1s) and their downstream SIFamide neurons. The requirement of clock neurons from a gating mechanism of extinction for a single extinction learning trial robustly causes typical extinction when coupled with acute activation of DN1s, as marked by the initially enhanced but eventually diminished memory suppression. Accordingly, we detected specific neural responses to extinction training in a few DN1s via calcium imaging fulfilled by the TRIC tool,7 but not in dorsal neurons 2 or dorsolateral neurons. Based on these findings, we propose that in extinction of appetitive long-term memory, multiple trials of extinction learning robustly activate DN1 clock neurons to open the gate of extinction, which may contribute to the transient nature of extinction memory.
Collapse
Affiliation(s)
- Yunchuan Zhang
- School of Life Science, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yinzhong Zhou
- School of Life Science, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xuchen Zhang
- School of Life Science, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Lingling Wang
- School of Life Science, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yi Zhong
- School of Life Science, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
40
|
Stehle JH, Zemmar A, Hausmann L. How to time the time - A preface to the special issue Circadian Rhythms in the Brain. J Neurochem 2021; 157:6-10. [PMID: 33724468 DOI: 10.1111/jnc.15311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 01/01/2023]
Abstract
In this Preface to the Journal of Neurochemistry special issue "Circadian Rhythms in the Brain", we summarize recent insights into connections between circadian rhythms and societal concerns related to aging and food intake, with consequences for healthy or aberrant metabolic homeostasis. The articles in this special issue were written by leading authors who presented their research at the 2019 Congress of the European Biological Rhythm Society, and are thus reflective of a broad variety of state-of-the-art research on all levels of chronobiology, from circadian rhythm generators in various tissues (including astrocytes) and the molecular mechanisms they base on, such as GABAergic regulation or ubiquitination, to the systems and behavioral level effects of chrono-nutrition and aging. Cover Image for this issue: https://doi.org/10.1111/jnc.15058.
Collapse
Affiliation(s)
- Jörg H Stehle
- Department of Neurosurgery, People's Hospital of Zhengzhou University, Henan Provincial People´s Hospital, Henan University People's Hospital, Henan University School of Medicine, Henan, China.,Institute of Cellular and Molecular Anatomy, Goethe-University, Frankfurt, Germany
| | - Ajmal Zemmar
- Department of Neurosurgery, People's Hospital of Zhengzhou University, Henan Provincial People´s Hospital, Henan University People's Hospital, Henan University School of Medicine, Henan, China.,Brain Research Institute, University of Zurich, Zurich, Switzerland.,Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Department of Neurosurgery, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Laura Hausmann
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
41
|
Soni SK, Basu P, Singaravel M, Sharma R, Pandi-Perumal SR, Cardinali DP, Reiter RJ. Sirtuins and the circadian clock interplay in cardioprotection: focus on sirtuin 1. Cell Mol Life Sci 2021; 78:2503-2515. [PMID: 33388853 PMCID: PMC11073088 DOI: 10.1007/s00018-020-03713-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/09/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Chronic disruption of circadian rhythms which include intricate molecular transcription-translation feedback loops of evolutionarily conserved clock genes has serious health consequences and negatively affects cardiovascular physiology. Sirtuins (SIRTs) are nuclear, cytoplasmic and mitochondrial histone deacetylases that influence the circadian clock with clock-controlled oscillatory protein, NAMPT, and its metabolite NAD+. Sirtuins are linked to the multi-organ protective role of melatonin, particularly in acute kidney injury and in cardiovascular diseases, where melatonin, via upregulation of SIRT1 expression, inhibits the apoptotic pathway. This review focuses on SIRT1, an NAD+-dependent class III histone deacetylase which counterbalances the intrinsic histone acetyltransferase activity of one of the clock genes, CLOCK. SIRT1 is involved in the development of cardiomyocytes, regulation of voltage-gated cardiac sodium ion channels via deacetylation, prevention of atherosclerotic plaque formation in the cardiovascular system, protection against oxidative damage and anti-thrombotic actions. Overall, SIRT1 has a see-saw effect on cardioprotection, with low levels being cardioprotective and higher levels leading to cardiac hypertrophy.
Collapse
Affiliation(s)
- Sanjeev Kumar Soni
- Chronobiology Laboratory, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Priyoneel Basu
- Chronobiology Laboratory, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Muniyandi Singaravel
- Chronobiology Laboratory, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | | | - Daniel P Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
42
|
Mahoney H, Peterson E, Justin H, Gonzalez D, Cardona C, Stevanovic K, Faulkner J, Yunus A, Portugues A, Henriksen A, Burns C, McNeill C, Gamsby J, Gulick D. Inhibition of casein kinase 1 δ/ε improves cognitive performance in adult C57BL/6J mice. Sci Rep 2021; 11:4746. [PMID: 33637777 PMCID: PMC7910436 DOI: 10.1038/s41598-021-83957-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/14/2021] [Indexed: 01/31/2023] Open
Abstract
Time-of-day effects have been noted in a wide variety of cognitive behavioral tests, and perturbation of the circadian system, either at the level of the master clock in the SCN or downstream, impairs hippocampus-dependent learning and memory. A number of kinases, including the serine-threonine casein kinase 1 (CK1) isoforms CK1δ/ε, regulate the timing of the circadian period through post-translational modification of clock proteins. Modulation of these circadian kinases presents a novel treatment direction for cognitive deficits through circadian modulation. Here, we tested the potential for PF-670462, a small molecule inhibitor of CK1δ/ε, to improve cognitive performance in C57BL/6J mice in an array of behavioral tests. Compared to vehicle-treated mice tested at the same time of the circadian day, mice treated with PF-670462 displayed better recall of contextual fear conditioning, made fewer working memory errors in the radial arm water maze, and trained more efficiently in the Morris Water Maze. These benefits were accompanied by increased expression of activity-regulated cytoskeleton-associated protein (Arc) in the amygdala in response to an acute learning paradigm. Our results suggest the potential utility of CK1δ/ε inhibition in improving time-of-day cognitive performance.
Collapse
Affiliation(s)
- Heather Mahoney
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Emily Peterson
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Hannah Justin
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - David Gonzalez
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Christopher Cardona
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Korey Stevanovic
- National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, USA
| | - John Faulkner
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Amara Yunus
- College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Alexandra Portugues
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Amy Henriksen
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Camden Burns
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Cameron McNeill
- USF Health Informatics Institute, University of South Florida Health, Tampa, FL, USA
| | - Joshua Gamsby
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Danielle Gulick
- Byrd Alzheimer's Institute, University of South Florida Health, Tampa, FL, USA.
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
43
|
Diurnal changes in perineuronal nets and parvalbumin neurons in the rat medial prefrontal cortex. Brain Struct Funct 2021; 226:1135-1153. [PMID: 33585984 DOI: 10.1007/s00429-021-02229-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/21/2021] [Indexed: 01/08/2023]
Abstract
Perineuronal nets (PNNs) surrounding fast-spiking, parvalbumin (PV) interneurons provide excitatory:inhibitory balance, which is impaired in several disorders associated with altered diurnal rhythms, yet few studies have examined diurnal rhythms of PNNs or PV cells. We measured the intensity and number of PV cells and PNNs labeled with Wisteria floribunda agglutinin (WFA) and also the oxidative stress marker 8-oxo-deoxyguanosine (8-oxo-dG) in rat prelimbic medial prefrontal cortex (mPFC) at Zeitgeber times (ZT) ZT0 (lights-on, inactive phase), ZT6 (mid-inactive phase), ZT12 (lights-off, active phase), and ZT18 (mid-active phase). Relative to ZT0, the intensities of PNN and PV labeling were increased in the dark (active) phase compared with the light (inactive) phase. The intensity of 8-oxo-dG was decreased from ZT0 at all times (ZT6,12,18). We also measured GAD 65/67 and vGLUT1 puncta apposed to PV cells with and without PNNs. There were more excitatory puncta on PV cells with PNNs at ZT18 vs. ZT6, but no changes in PV cells without PNNs and no changes in inhibitory puncta. Whole-cell slice recordings in fast-spiking (PV) cells with PNNs showed an increased ratio of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor:N-methyl-D-aspartate receptor (AMPA: NMDA) at ZT18 vs. ZT6. The number of PV cells and PV/PNN cells containing orthodenticle homeobox 2 (OTX2), which maintains PNNs, showed a strong trend toward an increase from ZT6 to ZT18. Diurnal fluctuations in PNNs and PV cells are expected to alter cortical excitatory:inhibitory balance and provide new insights into treatments for diseases impacted by disturbances in sleep and circadian rhythms.
Collapse
|
44
|
Reshetnikov VV, Kisaretova PE, Ershov NI, Shulyupova AS, Oshchepkov DY, Klimova NV, Ivanchihina AV, Merkulova TI, Bondar NP. Genes associated with cognitive performance in the Morris water maze: an RNA-seq study. Sci Rep 2020; 10:22078. [PMID: 33328525 PMCID: PMC7744575 DOI: 10.1038/s41598-020-78997-6] [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: 06/26/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Learning and memory are among higher-order cognitive functions that are based on numerous molecular processes including changes in the expression of genes. To identify genes associated with learning and memory formation, here, we used the RNA-seq (high-throughput mRNA sequencing) technology to compare hippocampal transcriptomes between mice with high and low Morris water maze (MWM) cognitive performance. We identified 88 differentially expressed genes (DEGs) and 24 differentially alternatively spliced transcripts between the high- and low-MWM-performance mice. Although the sets of DEGs and differentially alternatively spliced transcripts did not overlap, both were found to be enriched with genes related to the same type of biological processes: trans-synaptic signaling, cognition, and glutamatergic transmission. These findings were supported by the results of weighted-gene co-expression network analysis (WGCNA) revealing the enrichment of MWM-cognitive-performance-correlating gene modules with very similar Gene Ontology terms. High-MWM-performance mice manifested mostly higher expression of the genes associated with glutamatergic transmission and long-term potentiation implementation, which are processes necessary for memory acquisition and consolidation. In this set, there were genes participating in the regulation of trans-synaptic signaling, primarily AMPA receptor signaling (Nrn1, Nptx1, Homer3, Prkce, Napa, Camk2b, Syt7, and Nrgn) and calcium turnover (Hpca, Caln1, Orai2, Cpne4, and Cpne9). In high-MWM-performance mice, we also demonstrated significant upregulation of the “flip” splice variant of Gria1 and Gria2 transcripts encoding subunits of AMPA receptor. Altogether, our data helped to identify specific genes in the hippocampus that are associated with learning and long-term memory. We hypothesized that the differences in MWM cognitive performance between the mouse groups are linked with increased long-term potentiation, which is mainly mediated by increased glutamatergic transmission, primarily AMPA receptor signaling.
Collapse
Affiliation(s)
- Vasiliy V Reshetnikov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Polina E Kisaretova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Nikita I Ershov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Anastasia S Shulyupova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Dmitry Yu Oshchepkov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Natalia V Klimova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | | | - Tatiana I Merkulova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Natalia P Bondar
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia. .,Novosibirsk State University, Novosibirsk, Russia.
| |
Collapse
|
45
|
Aten S, Kalidindi A, Yoon H, Rumbaugh G, Hoyt KR, Obrietan K. SynGAP is expressed in the murine suprachiasmatic nucleus and regulates circadian-gated locomotor activity and light-entrainment capacity. Eur J Neurosci 2020; 53:732-749. [PMID: 33174316 DOI: 10.1111/ejn.15043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/15/2022]
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus functions as the master circadian clock. The phasing of the SCN oscillator is locked to the daily solar cycle, and an intracellular signaling cassette from the small GTPase Ras to the p44/42 mitogen-activated protein kinase (ERK/MAPK) pathway is central to this entrainment process. Here, we analyzed the expression and function of SynGAP-a GTPase-activating protein that serves as a negative regulator of Ras signaling-within the murine SCN. Using a combination of immunohistochemical and Western blotting approaches, we show that SynGAP is broadly expressed throughout the SCN. In addition, temporal profiling assays revealed that SynGAP expression is regulated over the circadian cycle, with peak expression occurring during the circadian night. Further, time-of-day-gated expression of SynGAP was not observed in clock arrhythmic BMAL1 null mice, indicating that the daily oscillation in SynGAP is driven by the inherent circadian timing mechanism. We also show that SynGAP phosphorylation at serine 1138-an event that has been found to modulate its functional efficacy-is regulated by clock time and is responsive to photic input. Finally, circadian phenotypic analysis of Syngap1 heterozygous mice revealed enhanced locomotor activity, increased sensitivity to light-evoked clock entrainment, and elevated levels of light-evoked MAPK activity, which is consistent with the role of SynGAP as a negative regulator of MAPK signaling. These findings reveal that SynGAP functions as a modulator of SCN clock entrainment, an effect that may contribute to sleep and circadian abnormalities observed in patients with SYNGAP1 gene mutations.
Collapse
Affiliation(s)
- Sydney Aten
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Anisha Kalidindi
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Hyojung Yoon
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Gavin Rumbaugh
- Scripps Research, Department of Neuroscience, Jupiter, FL, USA.,Scripps Research, Department of Molecular Medicine, Jupiter, FL, USA
| | - Kari R Hoyt
- Division of Pharmaceutics and Pharmacology, Ohio State University, Columbus, OH, USA
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| |
Collapse
|
46
|
Song N, Du J, Gao Y, Yang S. Epitranscriptome of the ventral tegmental area in a deep brain-stimulated chronic unpredictable mild stress mouse model. Transl Neurosci 2020; 11:402-418. [PMID: 33343932 PMCID: PMC7724003 DOI: 10.1515/tnsci-2020-0146] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/08/2020] [Accepted: 10/02/2020] [Indexed: 12/20/2022] Open
Abstract
Deep brain stimulation (DBS) applied to the nucleus accumbens (NAc) alleviates the depressive symptoms of major depressive disorders. We investigated the mechanism of this effect by assessing gene expression and RNA methylation changes in the ventral tegmental area (VTA) following NAc-DBS in a chronic unpredictable mild stress (CUMS) mouse model of depression. Gene expression and N 6-methyladenosine (m6A) levels in the VTA were measured in mice subjected to CUMS and then DBS, and transcriptome-wide m6A changes were profiled using immunoprecipitated methylated RNAs with microarrays, prior to gene ontology analysis. The expression levels of genes linked to neurotransmitter receptors, transporters, transcription factors, neuronal activities, synaptic functions, and mitogen-activated protein kinase and dopamine signaling were upregulated in the VTA upon NAc-DBS. Furthermore, m6A modifications included both hypermethylation and hypomethylation, and changes were positively correlated with the upregulation of some genes. Moreover, the effects of CUMS on gene expression and m6A-mRNA modification were reversed by DBS for some genes. Interestingly, while the expression of certain genes was not changed by DBS, long-term stimulation did alter their m6A modifications. NAc-DBS-induced modifications are correlated largely with upregulation but sometimes downregulation of genes in CUMS mice. Our findings improve the current understanding of the molecular mechanisms underlying DBS effects on depression.
Collapse
Affiliation(s)
- Nan Song
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| | - Jun Du
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| | - Yan Gao
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| | - Shenglian Yang
- Center of Military Brain Science, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences (AMMS), The Academy of Military Sciences, No. 27 Taiping Road, Haidian District, Beijing, China, 100850
| |
Collapse
|
47
|
Yamakawa G, Brady R, Sun M, McDonald S, Shultz S, Mychasiuk R. The interaction of the circadian and immune system: Desynchrony as a pathological outcome to traumatic brain injury. Neurobiol Sleep Circadian Rhythms 2020; 9:100058. [PMID: 33364525 PMCID: PMC7752723 DOI: 10.1016/j.nbscr.2020.100058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/11/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) is a complex and costly worldwide phenomenon that can lead to many negative health outcomes including disrupted circadian function. There is a bidirectional relationship between the immune system and the circadian system, with mammalian coordination of physiological activities being controlled by the primary circadian pacemaker in the suprachiasmatic nucleus (SCN) of the hypothalamus. The SCN receives light information from the external environment and in turn synchronizes rhythms throughout the brain and body. The SCN is capable of endogenous self-sustained oscillatory activity through an intricate clock gene negative feedback loop. Following TBI, the response of the immune system can become prolonged and pathophysiological. This detrimental response not only occurs in the brain, but also within the periphery, where a leaky blood brain barrier can permit further infiltration of immune and inflammatory factors. The prolonged and pathological immune response that follows TBI can have deleterious effects on clock gene cycling and circadian function not only in the SCN, but also in other rhythmic areas throughout the body. This could bring about a state of circadian desynchrony where different rhythmic structures are no longer working together to promote optimal physiological function. There are many parallels between the negative symptomology associated with circadian desynchrony and TBI. This review discusses the significant contributions of an immune-disrupted circadian system on the negative symptomology following TBI. The implications of TBI symptomology as a disorder of circadian desynchrony are discussed.
Collapse
Affiliation(s)
- G.R. Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - R.D. Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
| | - M. Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - S.J. McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Australia
| | - S.R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
| | - R. Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
48
|
Chatterjee S, Angelakos CC, Bahl E, Hawk JD, Gaine ME, Poplawski SG, Schneider-Anthony A, Yadav M, Porcari GS, Cassel JC, Giese KP, Michaelson JJ, Lyons LC, Boutillier AL, Abel T. The CBP KIX domain regulates long-term memory and circadian activity. BMC Biol 2020; 18:155. [PMID: 33121486 PMCID: PMC7597000 DOI: 10.1186/s12915-020-00886-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/01/2020] [Indexed: 12/23/2022] Open
Abstract
Background CREB-dependent transcription necessary for long-term memory is driven by interactions with CREB-binding protein (CBP), a multi-domain protein that binds numerous transcription factors potentially affecting expression of thousands of genes. Identifying specific domain functions for multi-domain proteins is essential to understand processes such as cognitive function and circadian clocks. We investigated the function of the CBP KIX domain in hippocampal memory and gene expression using CBPKIX/KIX mice with mutations that prevent phospho-CREB (Ser133) binding. Results We found that CBPKIX/KIX mice were impaired in long-term memory, but not learning acquisition or short-term memory for the Morris water maze. Using an unbiased analysis of gene expression in the dorsal hippocampus after training in the Morris water maze or contextual fear conditioning, we discovered dysregulation of CREB, CLOCK, and BMAL1 target genes and downregulation of circadian genes in CBPKIX/KIX mice. Given our finding that the CBP KIX domain was important for transcription of circadian genes, we profiled circadian activity and phase resetting in CBPKIX/KIX mice. CBPKIX/KIX mice exhibited delayed activity peaks after light offset and longer free-running periods in constant dark. Interestingly, CBPKIX/KIX mice displayed phase delays and advances in response to photic stimulation comparable to wildtype littermates. Thus, this work delineates site-specific regulation of the circadian clock by a multi-domain protein. Conclusions These studies provide insight into the significance of the CBP KIX domain by defining targets of CBP transcriptional co-activation in memory and the role of the CBP KIX domain in vivo on circadian rhythms. Graphical abstract ![]()
Collapse
Affiliation(s)
- Snehajyoti Chatterjee
- Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Université de Strasbourg, Strasbourg, France.,LNCA, CNRS UMR 7364, Strasbourg, France.,Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Christopher C Angelakos
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, USA.,Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ethan Bahl
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Joshua D Hawk
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, USA.,Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie E Gaine
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Shane G Poplawski
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, USA.,Department of Biology, University of Pennsylvania, Philadelphia, PA, USA.,Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, USA
| | - Anne Schneider-Anthony
- Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Université de Strasbourg, Strasbourg, France.,LNCA, CNRS UMR 7364, Strasbourg, France
| | - Manish Yadav
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Giulia S Porcari
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean-Christophe Cassel
- Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Université de Strasbourg, Strasbourg, France
| | - K Peter Giese
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | - Jacob J Michaelson
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, USA.,Department of Communication Sciences and Disorders, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, USA.,Iowa Institute of Human Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Lisa C Lyons
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Anne-Laurence Boutillier
- Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Université de Strasbourg, Strasbourg, France. .,LNCA, CNRS UMR 7364, Strasbourg, France.
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.
| |
Collapse
|
49
|
Hozer C, Pifferi F. Physiological and cognitive consequences of a daily 26 h photoperiod in a primate : exploring the underlying mechanisms of the circadian resonance theory. Proc Biol Sci 2020; 287:20201079. [PMID: 32693726 PMCID: PMC7423648 DOI: 10.1098/rspb.2020.1079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
The biological clock expresses circadian rhythms, whose endogenous period (tau) is close to 24 h. Daily resetting of the circadian clock to the 24 h natural photoperiod might induce marginal costs that would accumulate over time and forward affect fitness. It was proposed as the circadian resonance theory. For the first time, we aimed to evaluate these physiological and cognitive costs that would partially explain the mechanisms of the circadian resonance hypothesis. We evaluated the potential costs of imposing a 26 h photoperiodic regimen compared to the classical 24 h entrainment measuring several physiological and cognitive parameters (body temperature, energetic expenditure, oxidative stress, cognitive performances) in males of a non-human primate (Microcebus murinus), a nocturnal species whose endogenous period is about 23.5 h. We found significant higher resting body temperature and energy expenditure and lower cognitive performances when the photoperiodic cycle length was 26 h. Together these results suggest that a great deviation of external cycles from tau leads to daily greater energetic expenditure, and lower cognitive capacities. To our knowledge, this study is the first to highlight potential mechanisms of circadian resonance theory.
Collapse
Affiliation(s)
| | - Fabien Pifferi
- UMR CNRS MNHN 7179 MECADEV, 1 Avenue du Petit Château 91800 Brunoy, France
| |
Collapse
|
50
|
Zhang B, Zhao J, Wang Z, Xu L, Liu A, Du G. DL0410 attenuates oxidative stress and neuroinflammation via BDNF/TrkB/ERK/CREB and Nrf2/HO-1 activation. Int Immunopharmacol 2020; 86:106729. [PMID: 32645628 DOI: 10.1016/j.intimp.2020.106729] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/02/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022]
Abstract
Oxidative stress and neuroinflammation have been deeply associated with Alzheimer's disease. DL0410 is a novel acetylcholinesterase inhibitor with potential anti-oxidative effects in AD-related animal models, while the specific mechanism has not been fully clarified. In this study, DL0410 was predicted to be related to the modification of cell apoptosis, oxidation-reduction process, inflammatory response and ERK1/ERK2 cascade by in silico target fishing and GO enrichment analysis. Then the possible protective effects of DL0410 were evaluated by hydrogen peroxide (H2O2)-induced oxidative stress model and lipopolysaccharides (LPS)-induced neuroinflammation model H2O2 decreased the viability of SH-SY5Y cells, induced malondialdehyde (MDA) accumulation, mitochondrial membrane potential (Δψm) loss and cell apoptosis, which could be reversed by DL0410 dose-dependently, indicating that DL0410 protected SH-SY5Y cells against H2O2-mediated oxidative stress. Western blot analysis showed that DL0410 increased the H2O2-triggered down-regulated TrkB, ERK and CREB phosphorylation and the expression of BDNF. In addition, TrkB inhibitor ANA-12, ERK inhibitor SCH772984 and CREB inhibitor 666-15 eliminated the inhibition of DL0410 on MDA accumulation and Δψm loss. Furthermore, DL0410 attenuates inflammatory responses and ROS production in LPS-treated BV2 cells, which is responsible for Nrf2 and HO-1 up-regulation. The present study demonstrates that DL0410 is a potential activator of the BDNF/TrkB/ERK/CREB and Nrf2/HO-1 pathway and may be a potential candidate for regulating oxidative stress and neuroinflammatory response in the brain. Together, the results showed that DL0410 is a promising drug candidate for treating AD and possibly other nervous system diseases associated with oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Baoyue Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jun Zhao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhe Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lvjie Xu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ailin Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Guanhua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|