1
|
Soukhaklari R, Pirsalami F, Moezi L, Moosavi M. Curcumin ameliorates aluminum oxide nanoparticle-induced memory deficit by regulating the hippocampal p38 signaling pathway in mice. Neurol Res 2025; 47:15-22. [PMID: 39566555 DOI: 10.1080/01616412.2024.2430998] [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: 07/18/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVES Exposure to aluminum (Al) has been shown to be strongly associated with the pathogenesis of Alzheimer's disease (AD). Recent evidence indicates that the toxicity of Al nanoparticle (Al-NP) is far greater than Al itself due to its particle size. Epidemiological studies suggest that curcumin lower the prevalence of AD. MAPKs (ERK, p38 and JNK) were suggested to be involved in AD pathology and memory impairment. The present study aimed to evaluate if curcumin has the ability to protect against behavioral deficits induced by subcutaneously administered Al-NP in mice. Furthermore, the levels of phosphorylated and total hippocampal MAPKs were assessed using western blottechnique. METHODS Al-NP (10 mg/kg/s.c.) was administered to adult male NMRI mice for 10 days with or without curcumin in doses of 2.5 or 25 mg/kg/oral gavage). Memory was assessed using passive avoidance apparatus and anxiety-like behavior was evaluated using elevated plus maze. Following the behavioral tasks, western blot analysis was performed on the hippocampal tissues to detect the levels of phosphorylated and total MAPKs. RESULTS The results revealed that Al-NP deteriorated memory with no significant effect on anxiety-like behaviors. Additionally, it activated hippocampal p38 signaling pathway with no effect on ERK and JNK. Curcumin treatment at the dose of 25 mg/kg restored memory and p38 activation. DISCUSSION This study suggests that subcutaneous Al-NP administration impairs memory and hippocampal p38 signaling with no effect on ERK and JNK. Co-administration of curcumin restored Al-NP induced memory impairment and hippocampal p38 phosphorylation.
Collapse
Affiliation(s)
- Roksana Soukhaklari
- Shiraz Neuroscience Research Centre, Shiraz University of Medical sciences, Shiraz, Iran
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Fatema Pirsalami
- Department of Pharmacology, Medical School, Shiraz University of Medical sciences, Shiraz, Iran
| | - Leila Moezi
- Department of Pharmacology, Medical School, Shiraz University of Medical sciences, Shiraz, Iran
| | - Maryam Moosavi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical sciences, Shiraz, Iran
| |
Collapse
|
2
|
Geraci F, Passiatore R, Penzel N, Laudani S, Bertolino A, Blasi G, Graziano ACE, Kikidis GC, Mazza C, Parihar M, Rampino A, Sportelli L, Trevisan N, Drago F, Papaleo F, Sambataro F, Pergola G, Leggio GM. Sex dimorphism controls dysbindin-related cognitive dysfunctions in mice and humans with the contribution of COMT. Mol Psychiatry 2024; 29:2666-2677. [PMID: 38532008 PMCID: PMC11420087 DOI: 10.1038/s41380-024-02527-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024]
Abstract
Cognitive dysfunctions are core-enduring symptoms of schizophrenia, with important sex-related differences. Genetic variants of the DTBPN1 gene associated with reduced dysbindin-1 protein (Dys) expression negatively impact cognitive functions in schizophrenia through a functional epistatic interaction with Catechol-O-methyltransferase (COMT). Dys is involved in the trafficking of dopaminergic receptors, crucial for prefrontal cortex (PFC) signaling regulation. Moreover, dopamine signaling is modulated by estrogens via inhibition of COMT expression. We hypothesized a sex dimorphism in Dys-related cognitive functions dependent on COMT and estrogen levels. Our multidisciplinary approach combined behavioral-molecular findings on genetically modified mice, human postmortem Dys expression data, and in vivo fMRI during a working memory task performance. We found cognitive impairments in male mice related to genetic variants characterized by reduced Dys protein expression (pBonferroni = 0.0001), as well as in male humans through a COMT/Dys functional epistatic interaction involving PFC brain activity during working memory (t(23) = -3.21; pFDR = 0.004). Dorsolateral PFC activity was associated with lower working memory performance in males only (p = 0.04). Also, male humans showed decreased Dys expression in dorsolateral PFC during adulthood (pFDR = 0.05). Female Dys mice showed preserved cognitive performances with deficits only with a lack of estrogen tested in an ovariectomy model (pBonferroni = 0.0001), suggesting that genetic variants reducing Dys protein expression could probably become functional in females when the protective effect of estrogens is attenuated, i.e., during menopause. Overall, our results show the differential impact of functional variants of the DTBPN1 gene interacting with COMT on cognitive functions across sexes in mice and humans, underlying the importance of considering sex as a target for patient stratification and precision medicine in schizophrenia.
Collapse
Affiliation(s)
- Federica Geraci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Roberta Passiatore
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 21205, Baltimore, MD, USA
| | - Nora Penzel
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Samuele Laudani
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Alessandro Bertolino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- Psychiatric Unit - University Hospital, 70124, Bari, Italy
| | - Giuseppe Blasi
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- Psychiatric Unit - University Hospital, 70124, Bari, Italy
| | - Adriana C E Graziano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Gianluca C Kikidis
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 21205, Baltimore, MD, USA
| | - Ciro Mazza
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Madhur Parihar
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 21205, Baltimore, MD, USA
| | - Antonio Rampino
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- Psychiatric Unit - University Hospital, 70124, Bari, Italy
| | - Leonardo Sportelli
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 21205, Baltimore, MD, USA
- Department of Human Genetics, Radboud University Nijmegen, 6525 GD, Nijmegen, The Netherlands
| | - Nicolò Trevisan
- Department of Neuroscience (DNS), University of Padova, 35121, Padova, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience area, Istituto Italiano di Tecnologia, Genova, Italy
| | - Fabio Sambataro
- Department of Neuroscience (DNS), University of Padova, 35121, Padova, Italy
| | - Giulio Pergola
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 21205, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 21205, Baltimore, MD, USA
| | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123, Catania, Italy.
| |
Collapse
|
3
|
Belozertseva IV, Merkulovs DD, Kaiser H, Rozhdestvensky TS, Skryabin BV. Advancing 3Rs: The Mouse Estrus Detector (MED) as a Low-Stress, Painless, and Efficient Tool for Estrus Determination in Mice. Int J Mol Sci 2024; 25:9429. [PMID: 39273375 PMCID: PMC11395264 DOI: 10.3390/ijms25179429] [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: 08/12/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Determining the estrous cycle stages in mice is essential for optimizing breeding strategies, synchronizing experimental timelines, and facilitating studies in behavior, drug testing, and genetics. It is critical for reducing the production of genetically unmodified offspring in the generation and investigation of genetically modified animal models. An accurate detection of the estrus cycle is particularly relevant in the context of the 3Rs-Replacement, Reduction, and Refinement. The estrous cycle, encompassing the reproductive phases of mice, is key to refining experimental designs and addressing ethical issues related to the use of animals in research. This study presents results from two independent laboratories on the efficacy of the Mouse Estrus Detector (MED) from ELMI Ltd. (Latvia) for the accurate determination of the estrus phase. The female mice of five strains/stocks (CD1, FVB/N, C57Bl6/J, B6D2F1, and Swiss) were used. The results showed that the MEDProTM is a low-traumatic, simple, rapid, and painless method of estrus detection that supports the principles of the 3Rs. The use of the MEDProTM for estrus detection in mice caused minimal stress, enhanced mating efficiency, facilitated an increase in the number of embryos for in vitro fertilization, and allowed the production of the desired number of foster animals.
Collapse
Affiliation(s)
- Irina V Belozertseva
- Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, L'va Tolstogo str. 6-8, St. Petersburg 197022, Russia
| | | | - Helena Kaiser
- Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), Medical Faculty, University of Münster, von-Esmarch str. 56, D-48149 Münster, Germany
| | - Timofey S Rozhdestvensky
- Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), Medical Faculty, University of Münster, von-Esmarch str. 56, D-48149 Münster, Germany
| | - Boris V Skryabin
- Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), Medical Faculty, University of Münster, von-Esmarch str. 56, D-48149 Münster, Germany
| |
Collapse
|
4
|
Che J, Sun Y, Deng Y, Zhang J. Blood-brain barrier disruption: a culprit of cognitive decline? Fluids Barriers CNS 2024; 21:63. [PMID: 39113115 PMCID: PMC11305076 DOI: 10.1186/s12987-024-00563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Cognitive decline covers a broad spectrum of disorders, not only resulting from brain diseases but also from systemic diseases, which seriously influence the quality of life and life expectancy of patients. As a highly selective anatomical and functional interface between the brain and systemic circulation, the blood-brain barrier (BBB) plays a pivotal role in maintaining brain homeostasis and normal function. The pathogenesis underlying cognitive decline may vary, nevertheless, accumulating evidences support the role of BBB disruption as the most prevalent contributing factor. This may mainly be attributed to inflammation, metabolic dysfunction, cell senescence, oxidative/nitrosative stress and excitotoxicity. However, direct evidence showing that BBB disruption causes cognitive decline is scarce, and interestingly, manipulation of the BBB opening alone may exert beneficial or detrimental neurological effects. A broad overview of the present literature shows a close relationship between BBB disruption and cognitive decline, the risk factors of BBB disruption, as well as the cellular and molecular mechanisms underlying BBB disruption. Additionally, we discussed the possible causes leading to cognitive decline by BBB disruption and potential therapeutic strategies to prevent BBB disruption or enhance BBB repair. This review aims to foster more investigations on early diagnosis, effective therapeutics, and rapid restoration against BBB disruption, which would yield better cognitive outcomes in patients with dysregulated BBB function, although their causative relationship has not yet been completely established.
Collapse
Affiliation(s)
- Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yinying Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yixu Deng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
5
|
Huffman DM, Ajwad AA, Agarwal A, Lhamon ME, Donohue K, O'Hara BF, Sunderam S. Selective REM sleep restriction in mice using a device designed for tunable somatosensory stimulation. J Neurosci Methods 2024; 404:110063. [PMID: 38301833 PMCID: PMC10922658 DOI: 10.1016/j.jneumeth.2024.110063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 01/18/2024] [Accepted: 01/27/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Sleep perturbation is widely used to investigate the physiological mechanisms that mediate sleep-wake dynamics, and to isolate the specific roles of sleep in health and disease. However, state-of-the-art methods to accomplish sleep perturbation in preclinical models are limited in their throughput, flexibility, and specificity. NEW METHOD A system was developed to deliver vibro-tactile somatosensory stimulation aimed at controlled, selective sleep perturbation. The frequency and intensity of stimulation can be tuned to target a variety of experimental applications, from sudden arousal to sub-threshold transitions between light and deep stages of NREM sleep. This device was activated in closed-loop to selectively interrupt REM sleep in mice. RESULTS Vibro-tactile stimulation effectively and selectively interrupted REM sleep - significantly reducing the average REM bout duration relative to matched, unstimulated baseline recordings. As REM sleep was repeatedly interrupted, homeostatic mechanisms prompted a progressively quicker return to REM sleep. These effects were dependent on the parameters of stimulation applied. COMPARISON WITH EXISTING METHODS Existing sleep perturbation systems often require moving parts within the cage and/or restrictive housing. The system presented is unique in that it interrupts sleep without invading the animal's space. The ability to vary stimulation parameters is a great advantage over existing methods, as it allows for adaptation in response to habituation and/or circadian/homeostatic changes in arousal threshold. CONCLUSIONS The proposed method of stimulation demonstrates feasibility in affecting mouse sleep within a standard home cage environment, thus limiting environmental stress. Furthermore, the ability to tune frequency and intensity of stimulation allows for graded control over the extent of sleep perturbation, which potentially expands the utility of this technology beyond applications related to sleep.
Collapse
Affiliation(s)
- Dillon M Huffman
- F. Joseph Halcomb, III M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| | - Asma'a A Ajwad
- F. Joseph Halcomb, III M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| | | | | | | | | | - Sridhar Sunderam
- F. Joseph Halcomb, III M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
6
|
de Villiers O, Elliot-Wilson C, Thomas KGF, Semple PL, Naiker T, Henry M, Ross IL. Sleep and cognition in South African patients with non-functioning pituitary adenomas. PLoS One 2024; 19:e0296387. [PMID: 38236816 PMCID: PMC10796019 DOI: 10.1371/journal.pone.0296387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Strong lines of evidence in the neuroscience literature indicate that (a) healthy sleep facilitates cognitive processing, and (b) sleep disruption is associated with cognitive dysfunction. Despite the fact that patients with pituitary disease often display both disrupted sleep and cognitive dysfunction, few previous studies investigate whether these clinical characteristics in these patients might be related. Hence, we explored whether sleep disruption in patients with pituitary disease mediates their cognitive dysfunction. We recruited 18 patients with non-functioning pituitary adenomas (NFPA) and 19 sociodemographically matched healthy controls. They completed the Global Sleep Assessment Questionnaire (thus providing self-report data regarding sleep disruption) and were administered the Brief Test of Adult Cognition by Telephone, which assesses cognitive functioning in the domains of processing speed, working memory, episodic memory, inhibition, and reasoning. We found no significant differences in cognition between patients and controls. Furthermore, spectra of sleep disturbance did not differ significantly between patients and controls. Our data suggest that NFPA patients' cognition and sleep quality is relatively intact, and that sleep disruption does not mediate cognitive dysfunction. Larger studies should characterize sleep and cognition in patients with NFPA (and other pituitary diseases) to confirm whether disruption of the former mediates impairment in the latter.
Collapse
Affiliation(s)
- Olivia de Villiers
- ACSENT Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Claudia Elliot-Wilson
- ACSENT Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Kevin G. F. Thomas
- ACSENT Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Patrick L. Semple
- Division of Neurosurgery, University of Cape Town, Cape Town, South Africa
| | - Thurandrie Naiker
- Department of Radiation Oncology, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Michelle Henry
- Numeracy Centre, University of Cape Town, Cape Town, South Africa
| | - Ian L. Ross
- Division of Endocrinology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
7
|
Park J, Kim DY, Hwang GS, Han IO. Repeated sleep deprivation decreases the flux into hexosamine biosynthetic pathway/O-GlcNAc cycling and aggravates Alzheimer's disease neuropathology in adult zebrafish. J Neuroinflammation 2023; 20:257. [PMID: 37946213 PMCID: PMC10634120 DOI: 10.1186/s12974-023-02944-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
This study investigated chronic and repeated sleep deprivation (RSD)-induced neuronal changes in hexosamine biosynthetic pathway/O-linked N-acetylglucosamine (HBP/O-GlcNAc) cycling of glucose metabolism and further explored the role of altered O-GlcNAc cycling in promoting neurodegeneration using an adult zebrafish model. RSD-triggered degenerative changes in the brain led to impairment of memory, neuroinflammation and amyloid beta (Aβ) accumulation. Metabolite profiling of RSD zebrafish brain revealed a significant decrease in glucose, indicating a potential association between RSD-induced neurodegeneration and dysregulated glucose metabolism. While RSD had no impact on overall O-GlcNAcylation levels in the hippocampus region, changes were observed in two O-GlcNAcylation-regulating enzymes, specifically, a decrease in O-GlcNAc transferase (OGT) and an increase in O-GlcNAcase (OGA). Glucosamine (GlcN) treatment induced an increase in O-GlcNAcylation and recovery of the OGT level that was decreased in the RSD group. In addition, GlcN reversed cognitive impairment by RSD. GlcN reduced neuroinflammation and attenuated Aβ accumulation induced by RSD. Repeated treatment of zebrafish with diazo-5-oxo-l-norleucine (DON), an inhibitor of HBP metabolism, resulted in cognitive dysfunction, neuroinflammation and Aβ accumulation, similar to the effects of RSD. The pathological changes induced by DON were restored to normal upon treatment with GlcN. Both the SD and DON-treated groups exhibited a common decrease in glutamate and γ-aminobutyric acid compared to the control group. Overexpression of OGT in zebrafish brain rescued RSD-induced neuronal dysfunction and neurodegeneration. RSD induced a decrease in O-GlcNAcylation of amyloid precursor protein and increase in β-secretase activity, which were reversed by GlcN treatment. Based on the collective findings, we propose that dysregulation of HBP and O-GlcNAc cycling in brain plays a crucial role in RSD-mediated progression of neurodegeneration and Alzheimer's disease pathogenesis. Targeting of this pathway may, therefore, offer an effective regulatory approach for treatment of sleep-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Jiwon Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea
| | - Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea.
- Department of Physiology and Biophysics, College of Medicine, Inha University, 100 Inha Ro, Nam-Gu, Incheon, 22212, Korea.
| |
Collapse
|
8
|
Sengupta A, Tudor JC, Cusmano D, Baur JA, Abel T, Weljie AM. Sleep deprivation and aging are metabolically linked across tissues. Sleep 2023; 46:zsad246. [PMID: 37738102 PMCID: PMC11502955 DOI: 10.1093/sleep/zsad246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/21/2023] [Indexed: 09/24/2023] Open
Abstract
STUDY OBJECTIVES Insufficient sleep is a concerning hallmark of modern society because sleep deprivation (SD) is a risk factor for neurodegenerative and cardiometabolic disorders. SD imparts an aging-like effect on learning and memory, although little is known about possible common molecular underpinnings of SD and aging. Here, we examine this question by profiling metabolic features across different tissues after acute SD in young adult and aged mice. METHODS Young adult and aged mice were subjected to acute SD for 5 hours. Blood plasma, hippocampus, and liver samples were subjected to UPLC-MS/MS-based metabolic profiling. RESULTS SD preferentially impacts peripheral plasma and liver profiles (e.g. ketone body metabolism) whereas the hippocampus is more impacted by aging. We further demonstrate that aged animals exhibit SD-like metabolic features at baseline. Hepatic alterations include parallel changes in nicotinamide metabolism between aging and SD in young animals. Overall, metabolism in young adult animals is more impacted by SD, which in turn induces aging-like features. A set of nine metabolites was classified (79% correct) based on age and sleep status across all four groups. CONCLUSIONS Our metabolic observations demonstrate striking parallels to previous observations in studies of learning and memory and define a molecular metabolic signature of sleep loss and aging.
Collapse
Affiliation(s)
- Arjun Sengupta
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer C Tudor
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current affiliation: Department of Biology, Saint Joseph’s University, Philadelphia, PA, USA
| | - Danielle Cusmano
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current Affiliation: Iowa Neuroscience Institute, Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 2312 PBDB, Iowa City, IA, USA
| | - Aalim M Weljie
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Koszałka A, Lustyk K, Pytka K. Sex-dependent differences in animal cognition. Neurosci Biobehav Rev 2023; 153:105374. [PMID: 37634555 DOI: 10.1016/j.neubiorev.2023.105374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
The differences in cognitive processes driven by biological sex are the issues that have gotten growing attention recently. Considering the increasing population suffering from various cognitive impairments and the development of therapeutic strategies, it is essential that we recognize the mechanisms responsible for discrepancies observed in male and female learning and memory functions. In this review, we discuss recent reports from preclinical studies on rodents regarding selected cognitive domains to explore the state of knowledge on sex-dependent differences and point to challenges encountered during such research. We focus on spatial, recognition, and emotional memory, as well as on executive functions, such as attention, cognitive flexibility, and working memory. This review will help to acknowledge sex-related differences in cognition and indicate some fields that lack sufficient data.
Collapse
Affiliation(s)
- Aleksandra Koszałka
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacodynamics, Medyczna 9, 30-688 Krakow, Poland; Jagiellonian University Medical College, Doctoral School of Medical and Health Sciences, Św. Łazarza 16, 31-530 Krakow, Poland
| | - Klaudia Lustyk
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacodynamics, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacodynamics, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
10
|
Wen J, Li Z, Zuo Z. Postoperative Learning and Memory Dysfunction Is More Severe in Males But Is Not Persistent and Transmittable to Next Generation in Young Adult Rats. J Neurosurg Anesthesiol 2023; 35:429-437. [PMID: 35605917 DOI: 10.1097/ana.0000000000000856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/18/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) affects numerous patients each year and is associated with poor outcomes. Currently, the duration of POCD is not known. This preclinical study determined whether POCD was persistent, different between sexes and transmittable to the next generation. METHODS Two-month-old Sprague-Dawley rats had left carotid artery exposure under isoflurane anesthesia and their learning and memory were assessed from 5 days, 2 months, and 4 months after surgery. Rats with or without surgery were mated when they were 4 or 6 months old, and the learning and memory of the offspring were tested at 2 months of age. RESULTS Males exposed to surgery took a longer time to identify the target box after training sessions in a Barnes maze and had less freezing behavior in context-related fear conditioning than control rats when the tests were started 5 days after surgery. Similarly, female rats had a poorer performance than control rats in the Barnes maze test from 5 days after surgery. However, these poorer performances were not observed when the tests were administered 2 or 4 months after surgery. The offspring of rats with surgery had a performance similar to that of the offspring of control rats. CONCLUSIONS Our results suggest that both male and female rats develop POCD but that the learning and memory dysfunction appears to be more severe in male rats. POCD may not be persistent and does not transmit to the next generation.
Collapse
Affiliation(s)
- Jing Wen
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhisong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
| |
Collapse
|
11
|
Alzoubi KH, Halboup AM, Khabour OF, Alomari MA. The Protective Effects of the Combination of Vitamin E and Swimming Exercise on Memory Impairment Induced by Exposure to Waterpipe Smoke. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:304-312. [PMID: 35306997 DOI: 10.2174/1871527321666220318113635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/12/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Waterpipe smoking (WP) exposure involves a negative health impact, including memory deficit, which is attributed to the elevation of oxidative stress. Vitamin E (VitE) in combination with swimming exercise exerts protective effects that prevent memory impairment. In the current study, the modulation of WP-induced memory impairment by the combined effect of VitE and swimming exercise (SE) was investigated. METHODS Animals were exposed to WP one hour/day, five days per week for four weeks. Simultaneously, VitE (100 mg/kg, six days/week for four weeks) was administered via oral gavage, and the rats were made to swim one hour/day, five days/week for four weeks. Changes in memory were evaluated using radial arm water maze (RAWM), and oxidative stress biomarkers were examined in the hippocampus. RESULTS WP exposure induced short-term/long-term memory impairment (p<0.05). This impairment was prevented by a combination of VitE with SE (p<0.05). Additionally, this combination normalized the hippocampal catalase, GPx, and GSH/GSSG ratios that were modulated by WP (p<0.05). The combination further reduced TBARs levels below those of the control group (p<0.05). CONCLUSION WP-induced memory impairments were prevented by the combination of VitE with SE. This could be attributed to preserving the hippocampal oxidative mechanism by combining VitE and SE during WP exposure.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Abdulsalam M Halboup
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, University of Science and Technology, Sana\'a, Yemen
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Jordan
| | - Mahmoud A Alomari
- Department of Rehabilitation Sciences, Division of Physical Therapy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
12
|
Cloutier MÈ, Srivastava LK, Cermakian N. Exposure to Circadian Disruption During Adolescence Interacts With a Genetic Risk Factor to Modify Schizophrenia-relevant Behaviors in a Sex-dependent Manner. J Biol Rhythms 2022; 37:655-672. [PMID: 36168739 PMCID: PMC9749568 DOI: 10.1177/07487304221125363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
DTNBP1 is a gene associated with schizophrenia. Postmortem studies found a reduced expression of DTNBP1 in regions associated with schizophrenia in patients' brains. Sandy (Sdy) mice have a loss-of-function mutation in Dtnbp1 gene, resulting in behavioral deficits and brain changes similar to those seen in patients with schizophrenia. We previously showed that exposing adult Sdy mice to circadian disruption led to an exacerbation of schizophrenia-relevant behaviors. Here we asked whether the interaction between this genetic risk factor and circadian disruption occurs during adolescence, a period when environmental insults can promote schizophrenia symptoms, and whether sex affects this interaction. Starting at postnatal day 21, wild-type (WT) and Sdy males and females were housed for 4 weeks either in a 12 h light:12 h dark (LD 12:12) cycle or under chronic jetlag (CJL). Then, after 2 weeks in LD 12:12, behavioral assessments were conducted, including elevated plus maze (EPM), novel object recognition (NOR), social interaction, and prepulse inhibition (PPI) of acoustic startle. NOR and social novelty tests showed that, surprisingly, CJL during adolescence had opposite effects on WT and Sdy males, that is, behavioral deficits in WT males while rescuing preexisting deficits in Sdy mice. CJL led to decreased sociability in WT and Sdy mice while decreasing PPI only in females. Sdy mice showed decreased anxiety-like behavior compared with wild-type (WT), which was further accentuated by CJL in males. Thus, circadian disruption during adolescence, on its own or in association with Dtnbp1 mutation, can influence cognition, sociability, sensorimotor gating, and anxiety-like behaviors in a sex-dependent manner.
Collapse
Affiliation(s)
- Marie-Ève Cloutier
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada,Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Lalit K. Srivastava
- Douglas Mental Health University Institute, Montreal, QC, Canada,Department of Psychiatry, McGill University, Montreal, QC, Canada,Lalit K. Srivastava, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada; e-mail:
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, Montreal, QC, Canada,Department of Psychiatry, McGill University, Montreal, QC, Canada,Nicolas Cermakian, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada; e-mail:
| |
Collapse
|
13
|
Foakes C, Lawrence-Sidebottom D, Dralega AT, Harvey DO, Schmidt MA, Davis CJ. The rat Lux Actuating Search Task (LAST) and effects of sleep deprivation on task reversal performance. Neurobiol Sleep Circadian Rhythms 2022; 13:100081. [PMID: 35989719 PMCID: PMC9388875 DOI: 10.1016/j.nbscr.2022.100081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Sleep deprivation (SD) causes significant deficits in multiple aspects of cognition, including sustained attention and working memory. Investigating the neural processes underpinning these cognitive losses has proven challenging due to the confounds of current animal tasks; many employ appetitive or aversive stimuli to motivate behavior, while others lack task complexity that translates to human studies of executive function. We established the Lux Actuating Search Task (LAST) to circumvent these issues. The LAST is performed in a circular, open-field arena that requires rats to find an unmarked, quasi-randomly positioned target. Constant low-level floor vibrations motivate ambulation, while light intensity (determined by the rodent's proximity to the target destination) provides continuous visual feedback. The task has two paradigms that differ based on the relationship between the light intensity and target proximity: the Low Lux Target (LLT) paradigm and the High Lux Target paradigm (HLT). In this study, on days 1–6, the rats completed nine trials per day on one of the two paradigms. On day 7, the rats were either sleep deprived by gentle handling or were left undisturbed before undertaking the opposite (reversal) paradigm on days 7–9. Our results showed that SD significantly impeded the ability of Long Evans rats to learn the reversal paradigm, as indicated by increased times to target and increased failure percentages compared to rats whose sleep was undisturbed. Rats also showed reduced learning with the HLT paradigm, as the initial task or as the reversal task, likely due to the rodents' photophobia limiting their motivation to navigate toward a bright light, which is required to succeed. A continuous feedback paradigm examining the effects of sleep loss on cognitive flexibility in rats is introduced. Floor vibrations motivate and variable light intensity directs navigation to an unmarked location in an open field arena. The reversal of light intensity cues from light to dark and vice versa is disrupted by sleep deprivation.
Collapse
Affiliation(s)
- Callum Foakes
- Elson S. Floyd College of Medicine and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| | - Darian Lawrence-Sidebottom
- Elson S. Floyd College of Medicine and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| | - Aseru T Dralega
- Elson S. Floyd College of Medicine and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| | - Daniel O Harvey
- Elson S. Floyd College of Medicine and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| | - Michelle A Schmidt
- Elson S. Floyd College of Medicine and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| | - Christopher J Davis
- Elson S. Floyd College of Medicine and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| |
Collapse
|
14
|
Sur B, Lee B. Myricetin prevents sleep deprivation-induced cognitive impairment and neuroinflammation in rat brain via regulation of brain-derived neurotropic factor. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:415-425. [PMID: 36302617 PMCID: PMC9614391 DOI: 10.4196/kjpp.2022.26.6.415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 11/07/2022]
Abstract
Memory formation in the hippocampus is formed and maintained by circadian clock genes during sleep. Sleep deprivation (SD) can lead to memory impairment and neuroinflammation, and there remains no effective pharmacological treatment for these effects. Myricetin (MYR) is a common natural flavonoid that has various pharmacological activities. In this study, we investigated the effects of MYR on memory impairment, neuroinflammation, and neurotrophic factors in sleep-deprived rats. We analyzed SD-induced cognitive and spatial memory, as well as pro-inflammatory cytokine levels during SD. SD model rats were intraperitoneally injected with 10 and 20 mg/kg/day MYR for 14 days. MYR administration significantly ameliorated SD-induced cognitive and spatial memory deficits; it also attenuated the SD-induced inflammatory response associated with nuclear factor kappa B activation in the hippocampus. In addition, MYR enhanced the mRNA expression of brain-derived neurotropic factor (BDNF) in the hippocampus. Our results showed that MYR improved memory impairment by means of anti-inflammatory activity and appropriate regulation of BDNF expression. Our findings suggest that MYR is a potential functional ingredient that protects cognitive function from SD.
Collapse
Affiliation(s)
- Bongjun Sur
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Korea,Center for Converging Humanities, Kyung Hee University, Seoul 02447, Korea,Correspondence Bombi Lee, E-mail:
| |
Collapse
|
15
|
Dong TN, Kramár EA, Beardwood JH, Al-Shammari A, Wood MA, Keiser AA. Temporal endurance of exercise-induced benefits on hippocampus-dependent memory and synaptic plasticity in female mice. Neurobiol Learn Mem 2022; 194:107658. [PMID: 35811066 PMCID: PMC9901197 DOI: 10.1016/j.nlm.2022.107658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/20/2022] [Accepted: 07/04/2022] [Indexed: 02/08/2023]
Abstract
Exercise facilitates hippocampal neurogenesis and neuroplasticity that in turn, promotes cognitive function. Our previous studies have demonstrated that in male mice, voluntary exercise enables hippocampus-dependent learning in conditions that are normally subthreshold for long-term memory formation in sedentary animals. Such cognitive enhancement can be maintained long after exercise has ceased and can be re-engaged by a subsequent subthreshold exercise session, suggesting exercise-induced benefits are temporally dynamic. In females, the extent to which the benefits of exercise can be maintained and the mechanisms underlying this maintenance have yet to be defined. Here, we examined the exercise parameters required to initiate and maintain the benefits of exercise in female C57BL/6J mice. Using a subthreshold version of the hippocampus-dependent task called object-location memory (OLM) task, we show that 14d of voluntary exercise enables learning under subthreshold acquisition conditions in female mice. Following the initial exercise, a 7d sedentary delay results in diminished performance, which can be re-facilitated when animals receive 2d of reactivating exercise following the sedentary delay. Assessment of estrous cycle reveals enhanced wheel running activity during the estrus phase relative to the diestrus phase, whereas estrous phase on training or test had no effect on OLM performance. Utilizing the same exercise parameters, we demonstrate that 14d of exercise enhances long-term potentiation (LTP) in the CA1 region of the hippocampus, an effect that persists throughout the sedentary delay and following the reactivating exercise session. Previous studies have proposed exercise-induced BDNF upregulation as the mechanism underlying exercise-mediated benefits on synaptic plasticity and cognition. However, our assessment of hippocampal Bdnf mRNA expression following memory retrieval reveals no difference between exercise conditions and control, suggesting that persistent Bdnf upregulation may not be required for maintenance of exercise-induced benefits. Together, our data indicate that 14d of voluntary exercise can initiate long-lasting benefits on neuroplasticity and cognitive function in female mice, establishing the first evidence on the temporal endurance of exercise-induced benefits in females.
Collapse
Affiliation(s)
- T N Dong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - E A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - J H Beardwood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A Al-Shammari
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - M A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A A Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States.
| |
Collapse
|
16
|
Boi L, Petralla S, Monti B, Talani G, Sanna E, Pisu MG, Calderisi G, Maciocco E, Serra M, Concas A, Porcu P. Chronic treatment with hormonal contraceptives alters hippocampal BDNF and histone H3 post-translational modifications but not learning and memory in female rats. Horm Behav 2022; 144:105218. [PMID: 35785712 DOI: 10.1016/j.yhbeh.2022.105218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/11/2022] [Accepted: 06/20/2022] [Indexed: 12/11/2022]
Abstract
Hormonal contraceptives prevent ovulation with subsequent reduction in endogenous levels of estradiol, progesterone and its neuroactive metabolite allopregnanolone. These neurosteroids modulate several brain functions, including neuronal plasticity, cognition and memory. We hypothesized that hormonal contraceptives might affect synaptic plasticity, learning and memory, as a consequence of suppressed endogenous hormones levels. Female rats were orally treated with a combination of ethinyl estradiol (EE, 0.020 mg) and levonorgestrel (LNG, 0.060 mg) once daily for four weeks. Decreased hippocampal brain-derived neurotrophic factor (BDNF) levels and altered histone H3 post-translational modifications (PTMs) were observed 14 days after discontinuation from chronic EE-LNG treatment. These effects were not accompanied by alterations in long-term plasticity at glutamatergic synapses, recognition memory in the novel object and novel place location tests, or spatial learning, memory, and behavioral flexibility in the Morris water maze test. Thus, decreased BDNF content does not affect synaptic plasticity and cognitive performance; rather it might be relevant for the occurrence of certain psychiatric symptoms, reported by some women using hormonal contraceptives. These results provide the first evidence of hippocampal epigenetic changes induced by hormonal contraceptives and complement previous studies on the neurobiological actions of hormonal contraceptives; the finding that effects of chronic EE-LNG treatment on BDNF content and histone PTMs are observed 14 days after drug discontinuation warrants further investigation to better understand the implications of such long-term consequences for women's health.
Collapse
Affiliation(s)
- Laura Boi
- Department of Life and Environment Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Cagliari, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giuseppe Talani
- Neuroscience Institute, CNR - National Research Council of Italy, Cagliari, Italy
| | - Enrico Sanna
- Department of Life and Environment Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Cagliari, Italy; Neuroscience Institute, CNR - National Research Council of Italy, Cagliari, Italy
| | | | - Giulia Calderisi
- Department of Life and Environment Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Cagliari, Italy
| | - Elisabetta Maciocco
- Neuroscience Institute, CNR - National Research Council of Italy, Cagliari, Italy
| | - Mariangela Serra
- Department of Life and Environment Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Cagliari, Italy
| | - Alessandra Concas
- Department of Life and Environment Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Cagliari, Italy
| | - Patrizia Porcu
- Neuroscience Institute, CNR - National Research Council of Italy, Cagliari, Italy.
| |
Collapse
|
17
|
Rubio Arzola P, Shansky RM. Considering Organismal Physiology in Laboratory Studies of Rodent Behavior. Annu Rev Neurosci 2022; 45:387-402. [PMID: 35395164 DOI: 10.1146/annurev-neuro-111020-085500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Any experiment conducted in a rodent laboratory is done so against the backdrop of each animal's physiological state at the time of the experiment. This physiological state can be the product of multiple factors, both internal (e.g., animal sex, strain, hormone cycles, or circadian rhythms) and external (e.g., housing conditions, social status, and light/dark phases). Each of these factors has the potential to influence experimental outcomes, either independently or via interactions with others, and yet there is little consistency across laboratories in terms of the weight with which they are considered in experimental design. Such discrepancies-both in practice and in reporting-likely contribute to the perception of a reproducibility crisis in the field of behavioral neuroscience. In this review, we discuss how several of these sources of variability can impact outcomes within the realm of common learning and memory paradigms. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
| | - Rebecca M Shansky
- Department of Psychology, Northeastern University, Boston, Massachusetts, USA;
| |
Collapse
|
18
|
Lu C, Wei Z, Jiang N, Chen Y, Wang Y, Li S, Wang Q, Fan B, Liu X, Wang F. Soy isoflavones protects against cognitive deficits induced by chronic sleep deprivation via alleviating oxidative stress and suppressing neuroinflammation. Phytother Res 2022; 36:2072-2080. [PMID: 35373399 DOI: 10.1002/ptr.7354] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/21/2021] [Accepted: 11/27/2021] [Indexed: 12/30/2022]
Abstract
Mounting evidence suggests that there is a close association between chronic sleep deprivation (CSD) and cognitive deficits. The animal model of CSD-induced cognitive deficits is commonly used to seek potential treatments. Soy isoflavones (SI) have been reported to possess antioxidant, anti-inflammation, and neuroprotective effects. In the present study, the effects of SI on CSD-induced memory impairment were investigated. The mice were subjected to the sleep interruption apparatus and continuously sleep deprived for 2 weeks, while orally administrated with SI (10, 20, and 40 mg/kg) or Modafinil (MOD,100 mg/kg) during the CSD process. Immediately after the SD protocol, cognitive performance of mice was evaluated by the object location recognition (OLR) test, the novel object recognition (NOR) test, and the Morris water maze (MWM) task, as well as the hippocampus, was extracted for evaluation of oxidative stress parameters and inflammation levels through biochemical parameter assay and western blotting analysis. The results showed that SI administration remarkably improved the cognitive performance of CSD-treated mice in OLR, NOR, and MWM tests. In addition, SI significantly elevated total antioxidant capacity and superoxide dismutase enzyme activities, decreased malondialdehyde level, promoting antioxidant element nuclear erythroid-2-related factor 2, and its downstream targets, including heme oxygenase 1, and quinone oxidoreductase 1 protein expressions. Moreover, SI treatment significantly suppressed nuclear factor kappa B p65, nitric oxide synthase, and cyclooxygenase 2 activation, as well as the pro-inflammatory cytokines (Tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]) release in the hippocampus of CSD-treated mice. In summary, the current study provides an insight into the potential of SI in treatment of cognitive deficits by CSD.
Collapse
Affiliation(s)
- Cong Lu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Zhen Wei
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China.,College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ning Jiang
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongquan Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Shuying Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Qiong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Bei Fan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Xinmin Liu
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China.,College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
19
|
Massadeh AM, Alzoubi KH, Milhem AM, Rababa'h AM, Khabour OF. Evaluating the effect of selenium on spatial memory impairment induced by sleep deprivation. Physiol Behav 2022; 244:113669. [PMID: 34871651 DOI: 10.1016/j.physbeh.2021.113669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/19/2021] [Accepted: 12/01/2021] [Indexed: 01/09/2023]
Abstract
Sleep deprivation (SD) impairs memory due to disturbing oxidative stress parameters. Selenium is a main component of several antioxidant enzymes and provides a neuroprotective effect. The present study aimed to investigate the potential neuroprotective effect of chronic selenium administration on cognitive impairments induced by chronic SD. Adult male Wister rats were randomly assigned into five groups (n = 12/group). The SD was induced in rats using modified multiple platform model. Selenium (6 µg/kg of animal's body weight) was administered to rats via oral gavage for 6 weeks. The spatial learning and memory were assessed using the radial arm water maze (RAWM). Moreover, we measured the levels of reduced glutathione (GSH), oxidized glutathione (GSSG) and GSH/GSSG, catalase, glutathione peroxidase (GPx), superoxide dismutase (SOD), thiobarbituric acid reactive substances (TBARS) and brain derived neurotrophic factor (BDNF) in the hippocampus. The results indicate that short- and long-term memory were impaired by chronic sleep deprivation (P < 0.05), while selenium administration prevented this effect. Moreover, selenium normalized antioxidants activities which were reduced by SD such as: catalase (P < 0.05), and SOD (P < 0.05), and significantly enhanced the ratio of GSH/GSSG in sleep-deprived rats (P < 0.05), without significant alteration of BDNF (P > 0.05), GSH (P > 0.05), or TBARS levels (P > 0.05). In conclusion, chronic SD induced memory impairment, and chronic treatment with selenium prevented this impairment by normalizing antioxidant enzymes activities in the hippocampus.
Collapse
Affiliation(s)
- Adnan M Massadeh
- Department of Medicinal Chemistry and Pharmacognosy , Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110 Jordan.
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE; Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110 Jordan
| | - Amal M Milhem
- Department of Medicinal Chemistry and Pharmacognosy , Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110 Jordan
| | - Abeer M Rababa'h
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110 Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110 Jordan
| |
Collapse
|
20
|
Mehrbeheshti N, Esmaili Z, Ahmadi M, Moosavi M. A dose response effect of oral aluminum nanoparticle on novel object recognition memory, hippocampal caspase-3 and MAPKs signaling in mice. Behav Brain Res 2022; 417:113615. [PMID: 34606775 DOI: 10.1016/j.bbr.2021.113615] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/30/2022]
Abstract
The increasing use of aluminum nanoparticles (nano-Al) leads to increased human exposure and might affect human health. Considering the suggested connection between aluminum exposure and Alzheimer's disease (AD) pathogenesis, there is a concern about the effect of nano-Al on cognitive function and brain health. This study was aimed to assess the effect of a 5-day oral gavage of aluminum oxide nanoparticle (nano-Al) on memory and the phosphorylation levels of hippocampal p38, JNK (c-Jun N-terminal kinase), ERK (extracellular signal-regulated kinase) as well as cleaved caspase-3 in mice. Adult male NMRI mice were treated with nano-Al in doses 5 and 10 mg/kg/oral gavage for 5 days. The test session of novel object recognition (NOR) task was performed on day 5. Following the NOR test, the hippocampi were isolated for western blot analysis to determine the total and phosphorylated levels of p38, JNK, ERK as well as cleaved caspase-3 proteins. The results showed that nano-Al oral gavage in doses of 5 and 10 mg/kg impairs NOR memory in mice. Moreover, the memory impairing effect of nano-Al coincided with a dose dependent increase in phosphorylated p38 and cleaved caspase-3 in the hippocampus. It also increased the ratio of phosphorylated to total content of ERK in the hippocampus while JNK signaling was not affected by nano-Al. This study showed that nano-Al in doses as low as 5 and 10 mg/ kg ingested for 5 days impairs NOR memory and activates p38, ERK and cleaved caspase-3 in the hippocampus.
Collapse
Affiliation(s)
- Nahid Mehrbeheshti
- Shiraz Neuroscience Research Centre, Shiraz University of Medical sciences, Shiraz, Iran
| | - Zahra Esmaili
- Nanobiology and Nanomedicine Research Centre, Shiraz University of Medical sciences, Shiraz, Iran
| | - Mojdeh Ahmadi
- Nanobiology and Nanomedicine Research Centre, Shiraz University of Medical sciences, Shiraz, Iran
| | - Maryam Moosavi
- Nanobiology and Nanomedicine Research Centre, Shiraz University of Medical sciences, Shiraz, Iran.
| |
Collapse
|
21
|
Frye HE, Izumi Y, Harris AN, Williams SB, Trousdale CR, Sun MY, Sauerbeck AD, Kummer TT, Mennerick S, Zorumski CF, Nelson EC, Dougherty JD, Morón JA. Sex Differences in the Role of CNIH3 on Spatial Memory and Synaptic Plasticity. Biol Psychiatry 2021; 90:766-780. [PMID: 34548146 PMCID: PMC8571071 DOI: 10.1016/j.biopsych.2021.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND CNIH3 is an AMPA receptor (AMPAR) auxiliary protein prominently expressed in the dorsal hippocampus (dHPC), a region that plays a critical role in spatial memory and synaptic plasticity. However, the effects of CNIH3 on AMPAR-dependent synaptic function and behavior have not been investigated. METHODS We assessed a gain-of-function model of Cnih3 overexpression in the dHPC and generated and characterized a line of Cnih3-/- C57BL/6 mice. We assessed spatial memory through behavioral assays, protein levels of AMPAR subunits and synaptic proteins by immunoblotting, and long-term potentiation in electrophysiological recordings. We also utilized a super-resolution imaging workflow, SEQUIN (Synaptic Evaluation and Quantification by Imaging of Nanostructure), for analysis of nanoscale synaptic connectivity in the dHPC. RESULTS Overexpression of Cnih3 in the dHPC improved short-term spatial memory in female mice but not in male mice. Cnih3-/- female mice exhibited weakened short-term spatial memory, reduced dHPC synapse density, enhanced expression of calcium-impermeable AMPAR (GluA2-containing) subunits in synaptosomes, and attenuated long-term potentiation maintenance compared with Cnih3+/+ control mice; Cnih3-/- males were unaffected. Further investigation revealed that deficiencies in spatial memory and changes in AMPAR composition and synaptic plasticity were most pronounced during the metestrus phase of the estrous cycle in female Cnih3-/- mice. CONCLUSIONS This study identified a novel effect of sex and estrous on CNIH3's role in spatial memory and synaptic plasticity. Manipulation of CNIH3 unmasked sexually dimorphic effects on spatial memory, synaptic function, AMPAR composition, and hippocampal plasticity. These findings reinforce the importance of considering sex as a biological variable in studies of memory and hippocampal synaptic function.
Collapse
Affiliation(s)
- Hannah E Frye
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri; Program in Neuroscience, Washington University in St. Louis, St. Louis, Missouri
| | - Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Alexis N Harris
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Sidney B Williams
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher R Trousdale
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri
| | - Min-Yu Sun
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew D Sauerbeck
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Terrance T Kummer
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Steven Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Elliot C Nelson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Joseph D Dougherty
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Jose A Morón
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri; Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
22
|
Smiley CE, McGonigal JT, Nimchuk KE, Gass JT. Optogenetic manipulation of the prelimbic cortex during fear memory reconsolidation alters fear extinction in a preclinical model of comorbid PTSD/AUD. Psychopharmacology (Berl) 2021; 238:3193-3206. [PMID: 34347171 DOI: 10.1007/s00213-021-05935-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/12/2021] [Indexed: 12/27/2022]
Abstract
RATIONALE AND OBJECTIVE Post-traumatic stress disorder (PTSD) and alcohol use disorder (AUD) are disorders of learning and memory that often occur comorbidly. Exposure to trauma-related cues can increase alcohol intake in PTSD patients that are using alcohol to self-medicate. The recurrence of anxiety symptoms with subsequent alcohol use may initiate a destructive cycle where stress and alcohol exposure impair the function of the prefrontal cortex (PFC). While the incidence of these disorders has steadily increased, current therapies and treatments often lack efficacy. Thus, investigation into the underlying neurocircuitry responsible for the establishment and maintenance of these disorders is necessary to develop novel treatment targets. METHODS The present study examined the effects of ethanol exposure on the ability to create new learned associations around previously conditioned fear cues in a rat model. Animals were exposed to fear conditioning followed by chronic intermittent ethanol to translationally model trauma exposure followed by alcohol abuse. Optogenetics was used to inhibit the prelimbic (PrL) or infralimbic (IfL) cortex during fear memory reconsolidation, and fear behaviors were measured during subsequent extinction and spontaneous recovery tests. Results and conclusion Chronic ethanol exposure led to deficits in fear extinction learning and increased freezing during spontaneous recovery, both of which were prevented following inhibition of the PrL, but not the IfL, during memory reconsolidation. These results support the involvement of the PrL in fear learning and memory, and strongly suggest that the PrL could serve as a potential target for the treatment of the learning and memory deficits that occur following exposure to stress and alcohol.
Collapse
Affiliation(s)
- C E Smiley
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA.
| | - J T McGonigal
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA
| | - K E Nimchuk
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA
| | - J T Gass
- Department of Neuroscience, Basic Science Building, Medical University of South Carolina, 173 Ashley Avenue, Room 403, Charleston, SC, 29425, USA
- Department of Biomedical Sciences, James H. Quillen College of Medicine & VA Medical Center, PO Box 70582, Johnson City, TN, 37614, USA
| |
Collapse
|
23
|
Arora S, Dharavath RN, Bansal Y, Bishnoi M, Kondepudi KK, Chopra K. Neurobehavioral alterations in a mouse model of chronic partial sleep deprivation. Metab Brain Dis 2021; 36:1315-1330. [PMID: 33740181 DOI: 10.1007/s11011-021-00693-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
The night shift paradigm induces a state of chronic partial sleep deprivation (CPSD) and enhances the vulnerability to neuronal dysfunction. However, the specific neuronal impact of CPSD has not been thoroughly explored to date. In the current study, the night shift condition was mimicked in female Swiss albino mice. The classical sleep deprivation model, i.e., Modified Multiple Platform (MMP) method, was used for 8 h/day from Monday to Friday with Saturday and Sunday as a weekend off for nine weeks. Following nine weeks of night shift schedule, their neurobehavioral profile and physiological parameters were assessed along with the activity of the mitochondrial complexes, oxidative stress, serotonin levels, and inflammatory markers in the brain. Mice showed an overall hyperactive behavioral profile including hyperlocomotion, aggression, and stereotyped behavior accompanied by decreased activity of mitochondrial enzymes and serotonin levels, increased oxidative stress and inflammatory markers in whole brain homogenates. Collectively, the study points towards the occurrence of a hyperactive behavioral profile akin to mania and psychosis as a potential consequence of CPSD.
Collapse
Affiliation(s)
- Shiyana Arora
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Yashika Bansal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Mahendra Bishnoi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India.
| |
Collapse
|
24
|
Aleshin VA, Graf AV, Artiukhov AV, Boyko AI, Ksenofontov AL, Maslova MV, Nogués I, di Salvo ML, Bunik VI. Physiological and Biochemical Markers of the Sex-Specific Sensitivity to Epileptogenic Factors, Delayed Consequences of Seizures and Their Response to Vitamins B1 and B6 in a Rat Model. Pharmaceuticals (Basel) 2021; 14:ph14080737. [PMID: 34451834 PMCID: PMC8400147 DOI: 10.3390/ph14080737] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 01/28/2023] Open
Abstract
The disturbed metabolism of vitamins B1 or B6, which are essential for neurotransmitters homeostasis, may cause seizures. Our study aims at revealing therapeutic potential of vitamins B1 and B6 by estimating the short- and long-term effects of their combined administration with the seizure inductor pentylenetetrazole (PTZ). The PTZ dose dependence of a seizure and its parameters according to modified Racine’s scale, along with delayed physiological and biochemical consequences the next day after the seizure are assessed regarding sexual dimorphism in epilepsy. PTZ sensitivity is stronger in the female than the male rats. The next day after a seizure, sex differences in behavior and brain biochemistry arise. The induced sex differences in anxiety and locomotor activity correspond to the disappearance of sex differences in the brain aspartate and alanine, with appearance of those in glutamate and glutamine. PTZ decreases the brain malate dehydrogenase activity and urea in the males and the phenylalanine in the females. The administration of vitamins B1 and B6 24 h before PTZ delays a seizure in female rats only. This desensitization is not observed at short intervals (0.5–2 h) between the administration of the vitamins and PTZ. With the increasing interval, the pyridoxal kinase (PLK) activity in the female brain decreases, suggesting that the PLK downregulation by vitamins contributes to the desensitization. The delayed effects of vitamins and/or PTZ are mostly sex-specific and interacting. Our findings on the sex differences in sensitivity to epileptogenic factors, action of vitamins B1/B6 and associated biochemical events have medical implications.
Collapse
Affiliation(s)
- Vasily A. Aleshin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.A.A.); (A.V.A.); (A.I.B.)
- A.N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.V.G.); (A.L.K.)
| | - Anastasia V. Graf
- A.N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.V.G.); (A.L.K.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
- Faculty of Nano-, Bio-, Informational, Cognitive and Socio-Humanistic Sciences and Technologies at Moscow Institute of Physics and Technology, 123098 Moscow, Russia
| | - Artem V. Artiukhov
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.A.A.); (A.V.A.); (A.I.B.)
- A.N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.V.G.); (A.L.K.)
| | - Alexandra I. Boyko
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.A.A.); (A.V.A.); (A.I.B.)
| | - Alexander L. Ksenofontov
- A.N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.V.G.); (A.L.K.)
| | - Maria V. Maslova
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Isabel Nogués
- Research Institute of Terrestrial Ecosystems, National Research Council, Via Salaria km 29.300, Monterotondo, 00015 Rome, Italy;
| | - Martino L. di Salvo
- Department of Biological Sciences A. Rossi Fanelli, Sapienza University, 00185 Rome, Italy;
| | - Victoria I. Bunik
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.A.A.); (A.V.A.); (A.I.B.)
- A.N. Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (A.V.G.); (A.L.K.)
- Department of Biochemistry, Sechenov University, Trubetskaya, 8, bld. 2, 119991 Moscow, Russia
- Correspondence:
| |
Collapse
|
25
|
Alonso A, Genzel L, Gomez A. Sex and Menstrual Phase Influences on Sleep and Memory. CURRENT SLEEP MEDICINE REPORTS 2021. [DOI: 10.1007/s40675-020-00201-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Abstract
Purposes of Review
This review highlights the effect of sex differences in sleep mediated memory consolidation and cognitive performance. In addition, the role of menstrual cycle and the fluctuating level of sexual hormones (mainly oestrogen and progesterone) are stressed.
Recent Findings
The literature indicates that sex hormones mediate and orchestrate the differences observed in performance of females in comparison with males in a variety of tasks and can also be related to how sleep benefits cognition. Although the exact mechanism of such influence is not clear, it most likely involves differential activation of brain areas, sensitivity to neuromodulators (mainly oestrogen), circadian regulation of sleep and temperature, as well as modification of strategies to solve tasks across the menstrual cycle.
Summary
With the evidence presented here, we hope to encourage researchers to develop appropriate paradigms to study the complex relationship between menstrual cycle, sleep (its regulation, architecture and electrophysiological hallmarks) and performance in memory and other cognitive tasks.
Collapse
|
26
|
Jackson-Cowan L, Eldahshan W, Dumanli S, Dong G, Jamil S, Abdul Y, Althomali W, Baban B, Fagan SC, Ergul A. Delayed Administration of Angiotensin Receptor (AT2R) Agonist C21 Improves Survival and Preserves Sensorimotor Outcomes in Female Diabetic Rats Post-Stroke through Modulation of Microglial Activation. Int J Mol Sci 2021; 22:ijms22031356. [PMID: 33572986 PMCID: PMC7866408 DOI: 10.3390/ijms22031356] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
About 70% of stroke victims present with comorbid diseases such as diabetes and hypertension. The integration of comorbidities in pre-clinical experimental design is important in understanding the mechanisms involved in the development of stroke injury and recovery. We recently showed that administration of compound C21, an angiotensin II type 2 receptor agonist, at day 3 post-stroke improved sensorimotor outcomes by lowering neuroinflammation in diabetic male animals. In the current study, we hypothesized that a delayed administration of C21 would also lower chronic inflammation post-stroke in diabetic female animals. Young female diabetic rats were subjected to 1 h of middle cerebral artery occlusion (MCAO). Three days post-stroke, rats were administered C21 or vehicle in drinking water at a dose of 0.12 mg/kg/day for 4 weeks. The impact of C21 on microglial polarization was analyzed by flow cytometry in vivo and in vitro. Compound 21 treatment improved fine motor skills after MCAO through modulation of the microglia/macrophage inflammatory properties. In addition, C21 increased M2 polarization and reduced the M1:M2 ratio in vitro. In conclusion, delayed administration of C21 downregulates post-stroke inflammation in female diabetic animals. C21 may be a useful therapeutic option to lower neuro-inflammation and improve the post-stroke recovery in diabetes.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Cell Line
- Cognition/drug effects
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/physiopathology
- Female
- Infarction, Middle Cerebral Artery/complications
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/physiopathology
- Mice
- Microglia/drug effects
- Microglia/pathology
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Rats
- Rats, Wistar
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/metabolism
- Stroke/complications
- Stroke/drug therapy
- Stroke/physiopathology
Collapse
Affiliation(s)
- LaDonya Jackson-Cowan
- Department of Medicine, Augusta University/University of Georgia Medical Partnership, Athens, GA 30602, USA
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Wael Eldahshan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Selin Dumanli
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Guangkuo Dong
- Department Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Sarah Jamil
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Yasir Abdul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Waleed Althomali
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Babak Baban
- Department of Oral Biology, Dental College of Georgia, Augusta, GA 30912, USA
| | - Susan C Fagan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Adviye Ergul
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
27
|
Luo M, Song B, Zhu J. Electroacupuncture: A New Approach for Improved Postoperative Sleep Quality After General Anesthesia. Nat Sci Sleep 2020; 12:583-592. [PMID: 32922103 PMCID: PMC7457783 DOI: 10.2147/nss.s261043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 08/04/2020] [Indexed: 11/23/2022] Open
Abstract
General anesthesia produces a state of drug-induced unconsciousness that is controlled by the extent and duration of administered agents. Whether inhalation or intravenous in formulation, such agents may interfere with normal sleep-wake cycles, impairing postoperative sleep quality and creating complications. Electroacupuncture is a new approach widely applied in clinical practice during recent years. This particular technology helps regulate neurotransmitter concentrations in the brain, lowering norepinephrine and dopamine levels to improve sleep quality. It also alleviates surgical pain that degrades postoperative sleep quality after general anesthesia by downregulating immune activity (SP, NK-1, and COX-1) and upregulating serotonin receptor (5-HT1AR, 5-HT2AR) and endocannabinoid expression levels. However, large-scale, multicenter studies are still needed to determine the optimal duration, frequency, and timing of electroacupuncture for such use.
Collapse
Affiliation(s)
- Man Luo
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Bijia Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
- Department of Anesthesiology, Friendship Hospital of Capital Medical University, Beijing, People’s Republic of China
| | - Junchao Zhu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| |
Collapse
|
28
|
Zhang W, Huo T, Li A, Wu X, Feng C, Liu J, Jiang H. Identification of neurotoxicity markers induced by realgar exposure in the mouse cerebral cortex using lipidomics. JOURNAL OF HAZARDOUS MATERIALS 2020; 389:121567. [PMID: 32061421 DOI: 10.1016/j.jhazmat.2019.121567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/12/2019] [Accepted: 10/29/2019] [Indexed: 06/10/2023]
Abstract
Realgar is a traditional Chinese medicine containing arsenic and has neurotoxicity. This study used realgar exposure mice model, neurobehavioral tests, analytical chemistry, molecular biology and nontargeted lipidomics to explore the mechanism of realgar damages the nervous system. The arsenic contained in realgar passed through the BBB and accumulated in the brain. Neurons, synapses and myelin showed abnormal changes in the cerebral cortex. The number of autophagosomes were incresed as well as levels of MDA, Lp-PLA2, and cPLA2 but the CAT level was significant reduced. Finally, the cognition and memory of mice were decreased. Nontargeted lipidomics detected 34 lipid subclasses including 1603 lipid molecules. The levels of the LPC and LPE were significantly increased. Under the condition of variable importance for the projection (VIP)>1 and P < 0.05, only 28 lipid molecules satisfied the criteria. The lipid molecular markers SM (d36:2), PE (18:2/22:6) and PE (36:3) which were filtered by receiver operating characteristic (ROC) curve (AUC>0.8 or AUC<0.2) were used to identify the neurotoxicity induced by realgar. Therefore, realgar induces neurotoxicity through exacerbating oxidative damage and lipid dysfunction. Providing research basis for the clinical diagnosis and treatment of realgar-induced neurotoxicity.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Taoguang Huo
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Aihong Li
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Xinyu Wu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Cong Feng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jieyu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Hong Jiang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| |
Collapse
|
29
|
Protective effects of Genistein on the cognitive deficits induced by chronic sleep deprivation. Phytother Res 2020; 34:846-858. [DOI: 10.1002/ptr.6567] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/03/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
|
30
|
Sharma R, Sahota P, Thakkar MM. Sleep Loss Immediately After Fear Memory Reactivation Attenuates Fear Memory Reconsolidation. Neuroscience 2020; 428:70-75. [PMID: 31917354 DOI: 10.1016/j.neuroscience.2019.12.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/12/2019] [Accepted: 12/15/2019] [Indexed: 10/25/2022]
Abstract
Permanently stored memories become labile through a process called reactivation. Once reactivated, these memories need reconsolidation to become permanent. Sleep is critical for memory consolidation. Is sleep necessary for memory reconsolidation? We hypothesized that sleep loss immediately after fear reactivation (FR) will prevent memory reconsolidation. To test our hypothesis, two experiments were performed in adult male C57BL/6J mice exposed to contextual fear conditioning paradigm with inescapable foot shock as unconditional stimulus (US) and contextual cage as conditional stimulus (CS). Sleep loss was achieved either by 5 h of sleep deprivation (SD; Experiment 1) or by systemic infusion of modafinil (200 mg/Kg, ip), an FDA approved wake-promoting agent (Experiment 2). One hour after light-onset, fear memory acquisition (FMA) was performed on Day 1. Mice were allowed to explore CS for 5 min followed by presentation of US (7 foot-shocks; 0.5 mA, 2.0 s duration) at pseudorandom intervals. Controls were exposed to similar CS but no shocks were delivered. On Day 2, mice were exposed to CS for 2 min (without US; for FR) followed by either sleep loss or no sleep loss. On Day 3, fear memory recall (FMR) was performed by exposing mice to CS (without US) for 12 min. Percent time spent in freezing was monitored during FC, FR and FMR. Our results suggested that as compared to sleeping controls, mice with sleep loss immediately after FR displayed a significant reduction in percent time freezing during FMR. These results suggest that sleep loss may prevent memory reconsolidation.
Collapse
Affiliation(s)
- Rishi Sharma
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri-School of Medicine, Columbia, MO 65201, United States
| | - Pradeep Sahota
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri-School of Medicine, Columbia, MO 65201, United States
| | - Mahesh M Thakkar
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri-School of Medicine, Columbia, MO 65201, United States.
| |
Collapse
|
31
|
Wang H, Zhang L, Zhang Z, Li Y, Luo Q, Yuan S, Yan F. Perioperative Sleep Disturbances and Postoperative Delirium in Adult Patients: A Systematic Review and Meta-Analysis of Clinical Trials. Front Psychiatry 2020; 11:570362. [PMID: 33173517 PMCID: PMC7591683 DOI: 10.3389/fpsyt.2020.570362] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/14/2020] [Indexed: 01/11/2023] Open
Abstract
Background: The aim of this systematic review and meta-analysis of clinical trials was to investigate the effects of perioperative sleep disturbances on postoperative delirium (POD). Methods: Authors searched for studies (until May 12, 2020) reporting POD in patients with sleep disturbances following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Results: We identified 29 relevant trials including 55,907 patients. We divided these trials into three groups according to study design: Seven retrospective observational trials, 12 prospective observational trials, and 10 randomized controlled trials. The results demonstrated that perioperative sleep disturbances were significantly associated with POD occurrence in observational groups [retrospective: OR = 0.56, 95% CI: [0.33, 0.93], I 2 = 91%, p for effect = 0.03; prospective: OR = 0.27, 95% CI: [0.20, 0.36], I 2 = 25%, p for effect < 0.001], but not in the randomized controlled trial group [OR = 0.58, 95% CI: [0.34, 1.01], I 2 = 68%, p for effect = 0.05]. Publication bias was assessed using Egger's test. We used a one-by-one literature exclusion method to address high heterogeneity. Conclusions: Perioperative sleep disturbances were potential risk factors for POD in observational trials, but not in randomized controlled trials.
Collapse
Affiliation(s)
- Hongbai Wang
- Department of Anesthesiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Liang Zhang
- Department of Anesthesiology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Zhe Zhang
- Department of Anesthesiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Yinan Li
- Department of Anesthesiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Qipeng Luo
- Department of Anesthesiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Su Yuan
- Department of Anesthesiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| | - Fuxia Yan
- Department of Anesthesiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Fuwai Hospital, Beijing, China
| |
Collapse
|
32
|
APOE genetic background and sex confer different vulnerabilities to postnatal chlorpyrifos exposure and modulate the response to cholinergic drugs. Behav Brain Res 2019; 376:112195. [DOI: 10.1016/j.bbr.2019.112195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 08/20/2019] [Accepted: 08/28/2019] [Indexed: 01/30/2023]
|
33
|
Fidalgo S, Patel M, Quadir A, Sadiq W, Gard PR. Decreased behavioural and neurochemical effects of angiotensin IV following prenatal alcohol exposure in the mouse. Neuropeptides 2019; 77:101931. [PMID: 31079845 DOI: 10.1016/j.npep.2019.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/26/2019] [Accepted: 05/01/2019] [Indexed: 11/30/2022]
Abstract
Angiotensin IV (ang IV) is known to improve learning and memory in animal models but the mechanism is unclear. We have previously demonstrated sex differences in the pro-cognitive effects of ang IV, and that prenatal alcohol exposure (PAE) abolishes these effects. This study aimed to explore a possible mechanism underlying the sex differences and the effects of PAE in male mice. Mouse breeding harems received 5% ethanol in drinking water throughout pregnancy and lactation in a two-bottle schedule. The effects of ang IV were assessed in offspring at 4 months of age using the open field test, novel object recognition test and elevated plus maze. Aminopeptidase activity of brain insulin-regulated aminopeptidase (IRAP), a putative target of ang IV, was determined. As seen in a previous similar study, ang IV administered immediately after the second training trial significantly improved novel object recognition 24 h later in male mice but not female. PAE abolished this pro-cognitive effect in males. PAE also increased anxiety-like behaviour in male but not female offspring. Ang IV decreased the aminopeptidase activity of brain IRAP in control male, but not female, mice; PAE abolished this inhibitory effect. Ang IV improved memory consolidation in male but not female mice and PAE abolished this effect in the males. While the effects of PAE may be related to increased anxiety; ang IV decreased the aminopeptidase activity in male but not female mice and PAE abolished this inhibitory effect. The results therefore suggest that improvements in learning and memory induced by peripheral administration of ang IV correlate with a reduction of the enzyme activity of IRAP. This is the first demonstration that ang IV administered peripherally can induce long-term (24 h) changes in IRAP function which are probably not simple competitive inhibition and the first demonstration that PAE alters IRAP activity.
Collapse
Affiliation(s)
- Sara Fidalgo
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton BN2 4GJ, UK
| | - Mira Patel
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton BN2 4GJ, UK
| | - Angela Quadir
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton BN2 4GJ, UK.
| | - Wafia Sadiq
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton BN2 4GJ, UK
| | - Paul R Gard
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Moulsecoomb, Brighton BN2 4GJ, UK.
| |
Collapse
|
34
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
35
|
Stefanello FV, Fontana BD, Ziani PR, Müller TE, Mezzomo NJ, Rosemberg DB. Exploring Object Discrimination in Zebrafish: Behavioral Performance and Scopolamine-Induced Cognitive Deficits at Different Retention Intervals. Zebrafish 2019; 16:370-378. [DOI: 10.1089/zeb.2018.1703] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Flavia V. Stefanello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Barbara D. Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Paola R. Ziani
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Talise E. Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Nathana J. Mezzomo
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Denis B. Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, Louisiana
| |
Collapse
|
36
|
Preoperative Sleep Disturbance Exaggerates Surgery-Induced Neuroinflammation and Neuronal Damage in Aged Mice. Mediators Inflamm 2019; 2019:8301725. [PMID: 31011286 PMCID: PMC6442479 DOI: 10.1155/2019/8301725] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/23/2018] [Indexed: 12/20/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is defined as new cognitive impairment (memory impairment and impaired performance) after surgery, especially in aged patients. Sleep disturbance is a common phenomenon before surgery that has been increasingly thought to affect patient recovery. However, little is known about the functional impact of preoperative sleep disturbance on POCD. Here, we showed that tibial fracture surgery induced cognitive deficit and production of proinflammatory cytokines interleukin-6 (IL-6) and IL-1β, along with microglia and astrocyte activation, neuronal damage, and blood-brain barrier (BBB) disruption. Preoperative sleep disturbance enhanced the surgery-induced neuroinflammation, neuronal damage, BBB disruption, and memory impairment 24 h after surgery. Taken together, these results demonstrated that preoperative sleep disturbance aggravated postoperative cognitive function in aged mice and the mechanism may be related to central nervous system (CNS) inflammation and neuronal damage.
Collapse
|
37
|
Hou J, Shen Q, Wan X, Zhao B, Wu Y, Xia Z. REM sleep deprivation-induced circadian clock gene abnormalities participate in hippocampal-dependent memory impairment by enhancing inflammation in rats undergoing sevoflurane inhalation. Behav Brain Res 2019; 364:167-176. [PMID: 30779975 DOI: 10.1016/j.bbr.2019.01.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/18/2018] [Accepted: 01/18/2019] [Indexed: 12/31/2022]
Abstract
Sleep disturbance can result in memory impairment, and both sleep and hippocampal memory formation are maintained by circadian clock genes. Although preoperative sleep deprivation is known to be an independent risk factor for postoperative cognitive dysfunction (POCD) after inhalation anesthesia, the circadian mechanisms involved are currently unclear. To examine this issue, we constructed models of rapid eye movement sleep deprivation (RSD) and POCD after sevoflurane inhalation, to evaluate the circadian mechanisms underlying preoperative sleep deprivation-induced POCD after sevoflurane inhalation. Morris water maze probe test performance revealed that RSD aggravated the hippocampal-dependent memory impairment induced by sevoflurane anesthesia, and the recovery period of memory impairment was prolonged for more than a week by sleep deprivation. Western blot analysis revealed that sleep deprivation inhibited hippocampal Bmal1 and Egr1 expression for more than 7 days after sevoflurane inhalation. Importantly, hippocampal Per2 expression levels were first decreased by sevoflurane inhalation then increased from the third day by sleep deprivation. Sleep deprivation enhanced the expression of hippocampal inflammatory factors IL-1β and IL-6 after sevoflurane inhalation. In addition, sevoflurane inhalation activated the plasma expression of S100β and IL-6, particularly after sleep deprivation. Sleep deprivation aggravated pathogenic impairment of pyramidal neurons and activated astrocytes in CA1 after sevoflurane inhalation. These results suggest that preoperative RSD aggravates hippocampal memory impairment by enhancing neuroinflammatory injuries after sevoflurane inhalation, which is related to hippocampal clock gene abnormalities.
Collapse
Affiliation(s)
- Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Qianni Shen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Xing Wan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
38
|
Pantier LK, Li J, Christian CA. Estrous Cycle Monitoring in Mice with Rapid Data Visualization and Analysis. Bio Protoc 2019; 9:e3354. [PMID: 32695847 DOI: 10.21769/bioprotoc.3354] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The estrous cycle provides a readout of reproductive health in female laboratory rodents, and estrous cycle stage can be an important physiological variable. Accurate assessment of estrous cycle stage is also important in producing timed pregnancies for developmental studies. Here, we provide a protocol for evaluation of estrous cycle stage through a minimally invasive procedure of acquiring cells lining the vaginal cavity and immediate microscopic visual assessment of these cells without drying or staining. When performed over several consecutive days, the pattern of progression through the four main stages of the estrous cycle, and disruptions to this pattern, can be determined. We also present software that enables more efficient cycle stage data analysis and pattern visualization. These protocols and tools will thus facilitate the incorporation of female animals in laboratory experiments and enhance the assessment of relationships between the reproductive cycle and overall physiology and behavior.
Collapse
Affiliation(s)
- Leanna K Pantier
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jiang Li
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Catherine A Christian
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
39
|
Noise-induced sleep disruption increases weight gain and decreases energy metabolism in female rats. Int J Obes (Lond) 2018; 43:1759-1768. [PMID: 30568267 PMCID: PMC6584067 DOI: 10.1038/s41366-018-0293-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 01/28/2023]
Abstract
Background/objectives: Inadequate sleep increases obesity and environmental noise contributes to poor sleep. However, women may be more vulnerable to noise and hence more susceptible to sleep disruption-induced weight gain than men. In male rats, exposure to environmental (i.e. ambient) noise disrupts sleep and increases feeding and weight gain. However, the effects of environmental noise on sleep and weight gain in female rats are unknown. Thus, this study was designed to determine whether noise exposure would disturb sleep, increase feeding and weight gain and alter the length of the estrous cycle in female rats. Subjects/methods: Female rats (12-weeks old) were exposed to noise for 17d (8h/d during the light period) to determine the effects of noise on weight gain and food intake. In a separate set of females, estrous cycle phase and length, EEG, EMG, spontaneous physical activity and energy expenditure were recorded continuously for 27d during baseline (control, 9d), noise exposure (8h/d, 9d) and recovery (9d) from sleep disruption. Results: Noise exposure significantly increased weight gain and food intake compared to females that slept undisturbed. Noise also significantly increased wakefulness, reduced sleep and resulted in rebound sleep during the recovery period. Total energy expenditure was significantly lower during both noise exposure and recovery due to lower energy expenditure during spontaneous physical activity and sleep. Notably, noise did not alter the estrous cycle length. Conclusions: As previously observed in male rats, noise exposure disrupted sleep and increased weight gain in females but did not alter the length of the estrous cycle. This is the first demonstration of weight gain in female rats during sleep disruption. We conclude that the sleep disruption caused by exposure to environmental noise is a significant tool for determining how sleep loss contributes to obesity in females.
Collapse
|