1
|
Hu L, Qiu MJ, Fan WJ, Wang WE, Liu SH, Liu XQ, Liu SW, Shen ZJ, Zheng YF, Liu GC, Jia ZY, Wang XQ, Fang N. Characterization of GABAergic marker expression in prefrontal cortex in dexamethasone induced depression/anxiety model. Front Endocrinol (Lausanne) 2024; 15:1433026. [PMID: 39483976 PMCID: PMC11524930 DOI: 10.3389/fendo.2024.1433026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Background The pivotal responsibility of GABAergic interneurons is inhibitory neurotransmission; in this way, their significance lies in regulating the maintenance of excitation/inhibition (E/I) balance in cortical circuits. An abundance of glucocorticoids (GCs) exposure results in a disorder of GABAergic interneurons in the prefrontal cortex (PFC); the relationship between this status and an enhanced vulnerability to neuropsychiatric ailments, like depression and anxiety, has been identified, but this connection is still poorly understood because systematic and comprehensive research is lacking. Here, we aim to investigate the impact of dexamethasone (DEX, a GC receptor agonist) on GABAergic interneurons in the PFC of eight-week-old adult male mice. Methods A double-blind study was conducted where thirty-two mice were treated subcutaneously either saline or DEX (0.2 mg/10 ml per kg of body weight) dissolved in saline daily for 21 days. Weight measurements were taken at five-day intervals to assess the emotional changes in mice as well as the response to DEX treatment. Following the 21-day regimen of DEX injections, mice underwent examinations for depression/anxiety-like behaviours and GABAergic marker expression in PFC. Results In a depression/anxiety model generated by chronic DEX treatment, we found that our DEX procedure did trigger depression/anxiety-like behaviors in mice. Furthermore, DEX treatment reduced the expression levels of a GABA-synthesizing enzyme (GAD67), Reelin, calcium-binding proteins (parvalbumin and calretinin) and neuropeptides co-expressed in GABAergic neurons (somatostatin, neuropeptide Y and vasoactive intestinal peptide) in the PFC were reduced after 21 days of DEX treatment; these reductions were accompanied by decreases in brain size and cerebral cortex thickness. Conclusion Our results indicate that a reduction in the number of GABAergic interneurons may result in deficiencies in cortical inhibitory neurotransmission, potentially causing an E/I imbalance in the PFC; this insight suggests a potential breakthrough strategy for the treatment of depression and anxiety.
Collapse
Affiliation(s)
- Ling Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ming-Jing Qiu
- Neurological Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen-Juan Fan
- Luohe Medical College, Henan Province Engineering Research Center of Nutrition and Health, Luohe, China
| | - Wan-Er Wang
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Shao-Hao Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xiao-Qi Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Shi-Wei Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Ze-Jin Shen
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Ya-Fei Zheng
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Guang-Chao Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Zi-Yi Jia
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xiao-Qing Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Na Fang
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
2
|
Rappe A, Vihinen HA, Suomi F, Hassinen AJ, Ehsan H, Jokitalo ES, McWilliams TG. Longitudinal autophagy profiling of the mammalian brain reveals sustained mitophagy throughout healthy aging. EMBO J 2024:10.1038/s44318-024-00241-y. [PMID: 39367235 DOI: 10.1038/s44318-024-00241-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 10/06/2024] Open
Abstract
Mitophagy neutralizes mitochondrial damage, thereby preventing cellular dysfunction and apoptosis. Defects in mitophagy have been strongly implicated in age-related neurodegenerative disorders such as Parkinson's and Alzheimer's disease. While mitophagy decreases throughout the lifespan of short-lived model organisms, it remains unknown whether such a decline occurs in the aging mammalian brain-a question of fundamental importance for understanding cell type- and region-specific susceptibility to neurodegeneration. Here, we define the longitudinal dynamics of basal mitophagy and macroautophagy across neuronal and non-neuronal cell types within the intact aging mouse brain in vivo. Quantitative profiling of reporter mouse cohorts from young to geriatric ages reveals cell- and tissue-specific alterations in mitophagy and macroautophagy between distinct subregions and cell populations, including dopaminergic neurons, cerebellar Purkinje cells, astrocytes, microglia and interneurons. We also find that healthy aging is hallmarked by the dynamic accumulation of differentially acidified lysosomes in several neural cell subsets. Our findings argue against any widespread age-related decline in mitophagic activity, instead demonstrating dynamic fluctuations in mitophagy across the aging trajectory, with strong implications for ongoing theragnostic development.
Collapse
Affiliation(s)
- Anna Rappe
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Helena A Vihinen
- Electron Microscopy Unit (EMBI), Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Fumi Suomi
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Antti J Hassinen
- High Content Imaging and Analysis Unit (FIMM-HCA), Institute for Molecular Medicine, Helsinki Institute of Life Science, University of Helsinki, Tukholmankatu 8, Helsinki, 00290, Finland
| | - Homa Ehsan
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Eija S Jokitalo
- Electron Microscopy Unit (EMBI), Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Thomas G McWilliams
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
| |
Collapse
|
3
|
Asghar H, Siddiqui A, Batool L, Batool Z, Ahmed T. Post-exposure self-recovery reverses oxidative stress, ameliorates pathology and neurotransmitters imbalance and rescues spatial memory after time-dependent aluminum exposure in rat brain. Biometals 2024; 37:819-838. [PMID: 38233603 DOI: 10.1007/s10534-023-00570-1] [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: 08/31/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024]
Abstract
Aluminum is a potent neurotoxin, responsible for memory impairment and cognitive dysfunction. The neurotoxic effect of aluminum on cognitive impairment is well documented, however, exposure to aluminum in a time-dependent manner and post-exposure self-recovery still needs to be elaborated. This research aimed to (1) study the time-dependent effect of aluminum exposure by administering a total dose of 5850 mg/kg of Al over two different time periods: 30 and 45 days (130 and 195 mg/kg of AlCl3 respectively), and (2) study 20 days post-exposure self-recovery effect in both aluminum-exposed groups by giving distilled water. Cognitive abilities were investigated through Morris water maze test and hole board test and compared in both exposure and recovery groups. Oxidative stress markers and neurotransmitter levels were measured for both exposure and recovery groups. To understand the mechanism of aluminum exposure and recovery, immunohistochemical analysis of synaptophysin (Syp) and glial fibrillary acidic protein (GFAP) was performed. Results showed cognitive dysfunction, oxidative stress-induced damage, reduced neurotransmitter levels, decreased immunoreactivity of Syp, and increased GFAP. However, these parameters showed a larger improvement in the recovery group where rats were given aluminum for 30 days period in comparison to recovery group followed by 45 days of aluminum exposure. These results suggest that restoration of cognitive ability is affected by the duration of aluminum exposure. The study findings provide us with insight into the adverse effects of aluminum exposure and can be utilized to guide future preventive and therapeutic strategies against aluminum neurotoxicity.
Collapse
Affiliation(s)
- Humna Asghar
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Alveena Siddiqui
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Laraib Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan.
| |
Collapse
|
4
|
Sheng JA, Tobet SA. Maternal immune activation with toll-like receptor 7 agonist during mid-gestation alters juvenile and adult developmental milestones and behavior. J Neuroendocrinol 2024; 36:e13417. [PMID: 38822791 PMCID: PMC11296912 DOI: 10.1111/jne.13417] [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: 11/08/2023] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Infections during pregnancy are associated with increased risk for adult neuropsychiatric disease, such as major depressive disorder, schizophrenia, and autism spectrum disorder. In mouse models of maternal immune activation (MIA), different toll-like receptors (TLRs) are stimulated to initiate inflammatory responses in mother and fetus. The goal of this study was to determine sex-dependent aspects of MIA using a TLR7/8 agonist, Resiquimod (RQ), on neurodevelopment. RQ was administered to timed-pregnant mice on embryonic day (E) 12.5. At E15, maternal/fetal plasma cytokines were measured by enzyme-linked immunosorbent assay (ELISA). Maternal cytokines interleukin (IL)-6 and IL-10 were higher while tumor necrosis factor (TNF)-α and IL-17 were lower in pregnant dams exposed to RQ. Fetal cytokines (E15) were altered at the same timepoint with fetal plasma IL-6 and IL-17 greater after RQ compared to vehicle, while IL-10 and TNF-α were higher in male fetuses but not female. Other timed-pregnant dams were allowed to give birth. MIA with RQ did not alter the female to male ratio of offspring born per litter. Body weights were reduced significantly in both sexes at birth, and over the next 5 weeks. Offspring from RQ-injected mothers opened their eyes 5 days later than controls. Similarly, female offspring from RQ-injected mothers exhibited pubertal delay based on vaginal opening 2-3 days later than control females. On the behavioral side, juvenile and adult male and female MIA offspring exhibited less social-like behavior in a social interaction test. Anhedonia-like behavior was greater in MIA adult female mice. This study provides support for sex-dependent influences of fetal antecedents for altered brain development and behavioral outputs that could be indicative of increased susceptibility for adult disorders through immune mechanisms. Future studies are needed to determine neural cellular and molecular mechanisms for such programming effects.
Collapse
Affiliation(s)
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO
- Innovation Center on Sex Differences in Medicine, Mass General Hospital
| |
Collapse
|
5
|
Liao GY, Pettan-Brewer C, Ladiges W. Comparison of Age-Related Decline and Behavioral Validity in C57BL/6 and CB6F1 Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599036. [PMID: 38915625 PMCID: PMC11195229 DOI: 10.1101/2024.06.14.599036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Variability in physical resilience to aging prompts a comprehensive examination of underlying mechanisms across organs and individuals. We conducted a detailed exploration of behavioral and physiological differences between C57BL/6 and CB6F1 mice across various age groups. In behavioral assays, B6 mice displayed superior performance in rotarod tasks but higher anxiety while CB6F1 mice exhibited a decline in short-term memory with age. Grip strength, long-term memory, and voluntary wheel running declined similarly with age in both strains. Examining physiological phenotypes, B6 mice exhibited lower body fat percentages across ages compared to CB6F1 mice, though cataract severity worsened with age in both strains. Analysis of cardiac functions revealed differences between strains, with worsening left ventricular hypertrophy and structural heart abnormalities with age in CB6F1 mice along with higher blood pressure than B6. Lesion scores showed an age-related increase in heart, kidney, and liver lesions in both strains, while lung lesions worsened with age only in CB6F1 mice. This study underscores the validity of behavioral assays and geropathology assessment in reflecting age-related decline and emphasizes the importance of considering strain specificity when using mouse models to study human aging.
Collapse
Affiliation(s)
- Gerald Yu Liao
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Christina Pettan-Brewer
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Warren Ladiges
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Ünal D, Varol AB, Köse TB, Koçak EE. Morphological Correlates of Behavioral Variation in Autism Spectrum Disorder Groups in A Maternal Immune Activation Model. Noro Psikiyatr Ars 2024; 67:195-201. [PMID: 39258126 PMCID: PMC11382561 DOI: 10.29399/npa.28637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/16/2023] [Indexed: 09/12/2024] Open
Abstract
Introduction Clinical heterogeneity is one of the biggest challenges for researchers studying underlying neurobiological mechanisms in Autism Spectrum Disorder (ASD). We aimed to use polyinosinic-polycytidylic acid [Poly (I:C)] induced maternal immune activation mice model to investigate the behavioral variation and the role of brain circuits related to symptom clusters in ASD. For this purpose, behavioral tests were applied to offsprings and regional thickness was measured from histological brain sections in medial prefrontal cortex, hippocampus and striatum. Methods Pups of intraperitoneal Poly (I:C)-applied mothers (n: 14) and phosphate buffered saline-applied mothers (n: 6) were used for this study. We used three chamber socialization test and social memory test to evaluate social behavior deficit in mice. Marble burying test was used for assessing stereotypic behavior and new object recognition test for learning and cognitive flexibility. Three subgroups (n: 4 for each) were determined according to behavioral test parameters. Regional thickness was measured in medial prefrontal cortex, hippocampus and striatum and compared between subgroups. Results We detected that the behavioral differences were distributed in a spectrum as expected in the clinic and also detected increased hippocampus thickness in the stereotypic behavior dominant Poly (I:C) subgroup. Conclusion Poly (I:C) induced maternal immune activation model creates the behavioral variation and cortical development differences that are seen in relation with symptom groups in ASD.
Collapse
Affiliation(s)
- Dilek Ünal
- Hacettepe University School of Medicine, Department of Child and Adolescent Psychiatry, Ankara, Turkey
| | - Aslıhan Bahadır Varol
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| | - Tansu Bilge Köse
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| | - Emine Eren Koçak
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| |
Collapse
|
7
|
Hudock J, Kenney JW. Aging in zebrafish is associated with reduced locomotor activity and strain dependent changes in bottom dwelling and thigmotaxis. PLoS One 2024; 19:e0300227. [PMID: 38696419 PMCID: PMC11065237 DOI: 10.1371/journal.pone.0300227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/25/2024] [Indexed: 05/04/2024] Open
Abstract
Aging is associated with a wide range of physiological and behavioral changes in many species. Zebrafish, like humans, rodents, and birds, exhibits gradual senescence, and thus may be a useful model organism for identifying evolutionarily conserved mechanisms related to aging. Here, we compared behavior in the novel tank test of young (6-month-old) and middle aged (12-month-old) zebrafish from two strains (TL and TU) and both sexes. We find that this modest age difference results in a reduction in locomotor activity in male fish. We also found that background strain modulated the effects of age on predator avoidance behaviors related to anxiety: older female TL fish increased bottom dwelling whereas older male TU fish decreased thigmotaxis. Although there were no consistent effects of age on either short-term (within session) or long-term (next day) habituation to the novel tank, strain affected the habituation response. TL fish tended to increase their distance from the bottom of the tank whereas TU fish had no changes in bottom distance but instead tended to increase thigmotaxis. Our findings support the use of zebrafish for the study of how age affects locomotion and how genetics interacts with age and sex to alter exploratory and emotional behaviors in response to novelty.
Collapse
Affiliation(s)
- Jacob Hudock
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States of America
| | - Justin W. Kenney
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States of America
| |
Collapse
|
8
|
Rodríguez-Ibarra C, Díaz-Urbina D, Zagal-Salinas AA, Medina-Reyes EI, Déciga-Alcaraz A, Hernández-Pando R, Chirino YI. Oral exposure to food grade titanium dioxide (E171) induces intestinal and behavioural alterations in adult mice but limited effects in young mice. J Trace Elem Med Biol 2024; 83:127409. [PMID: 38394968 DOI: 10.1016/j.jtemb.2024.127409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Food-grade titanium dioxide (E171), a white colourant widely used in ultra-processed food products, has been banned in the European Union. However, its usage is still permitted in medicines, and in several other countries. The estimated intake of E171 in children is higher than in adults, which led us to hypothesise that E171 induces differential effects depending on age, with adult mice being the most susceptible due to age, despite the lower dose. AIM To evaluate the effects of oral administration of E171 on intestinal permeability, ileum, and colon histology, and how these effects impact anxious and depressive behaviour in young and adult mice of both sexes. METHODS Young and adult mice of both sexes C57BL/6 mice received 10 mg/kgbw E171/3 times per week for 3 months. E171 was administered orally in water by pipetting, while control groups only received drinking water, then intestinal permeability, histology and animal behaviour were analysed. RESULTS E171 showed an amorphous shape, primary particles sized below 1 µm and anatase crystalline structure. Oral administration of E171 disrupted the intestinal permeability in adult male and female mice, but no effects were observed in young mice of both sexes. E171 promoted ileal adenoma formation in half of the adult female population, moreover hyperplastic crypts, and hyperplastic goblet cells at histological level in adult mice of both sexes. The colon presented hyperplastic goblet cells, hyperchromatic nuclei, increased proliferation and DNA damage in adult mice of both sexes. The anxiety and depressive behaviour were only altered in adult mice treated with E171, but no changes were detected in young animals of both sexes. CONCLUSIONS Adult mice displayed higher susceptibility in all parameters analysed in this study compared to young mice of both sexes.
Collapse
Affiliation(s)
- Carolina Rodríguez-Ibarra
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico
| | - Daniel Díaz-Urbina
- Laboratorio de Neurobiología de la Alimentación. Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico; Laboratory of Neurobiology on Compulsive Behaviors, The National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institute of Health, Bethesda, MD, USA
| | - Alejandro A Zagal-Salinas
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico
| | - Estefany I Medina-Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico
| | - Alejandro Déciga-Alcaraz
- Química de Aerosoles Orgánicos Atmosféricos, Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, CP 04510 Ciudad de México, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Belisario Domínguez Sección 16, Tlalpan, CP 14080 Ciudad de México, Mexico
| | - Yolanda I Chirino
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico.
| |
Collapse
|
9
|
Seo JY, Jo HR, Lee SH, Kim DG, Lee H, Kim YL, Choi YI, Jung SJ, Son H. TRPC4 deletion elicits behavioral defects in sociability by dysregulating expression of microRNA-138-2. iScience 2024; 27:108617. [PMID: 38188509 PMCID: PMC10770719 DOI: 10.1016/j.isci.2023.108617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/08/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
To investigate whether the defects in transient receptor potential canonical 4 (TRPC4), which is strongly expressed in the hippocampus, are implicated in ASD, we examined the social behaviors of mice in which Trpc4 was deleted (Trpc4-/-). Trpc4-/- mice displayed the core symptoms of ASD, namely, social disability and repetitive behaviors. In microarray analysis of the hippocampus, microRNA (miR)-138-2, the precursor of miR-138, was upregulated in Trpc4-/- mice. We also found that binding of Matrin3 (MATR3), a selective miR-138-2 binding nuclear protein, to miR-138-2 was prominently enhanced, resulting in the downregulation of miR-138 in Trpc4-/- mice. Some parameters of the social defects and repetitive behaviors in the Trpc4-/- mice were rescued by increased miR-138 levels following miR-138-2 infusion in the hippocampus. Together, these results suggest that Trpc4 regulates some signaling components that oppose the development of social behavioral deficits through miR-138 and provide a potential therapeutic strategy for ASD.
Collapse
Affiliation(s)
- Jee Young Seo
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Hye-Ryeong Jo
- Hanyang University Hospital for Rheumatic Diseases, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Seung Hoon Lee
- Hanyang University Hospital for Rheumatic Diseases, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Huiju Lee
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Ye Lim Kim
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Young In Choi
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Sung Jun Jung
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Hyeon Son
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| |
Collapse
|
10
|
Bannerman DM, Barkus C, Eltokhi A. Behavioral Analysis of NMDAR Function in Rodents: Tests of Long-Term Spatial Memory. Methods Mol Biol 2024; 2799:107-138. [PMID: 38727905 DOI: 10.1007/978-1-0716-3830-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
NMDAR-dependent forms of synaptic plasticity in brain regions like the hippocampus are widely believed to provide the neural substrate for long-term associative memory formation. However, the experimental data are equivocal at best and may suggest a more nuanced role for NMDARs and synaptic plasticity in memory. Much of the experimental data available comes from studies in genetically modified mice in which NMDAR subunits have been deleted or mutated in order to disrupt NMDAR function. Behavioral assessment of long-term memory in these mice has involved tests like the Morris watermaze and the radial arm maze. Here we describe these behavioral tests and some of the different testing protocols that can be used to assess memory performance. We discuss the importance of distinguishing selective effects on learning and memory processes from nonspecific effects on sensorimotor or motivational aspects of performance.
Collapse
Affiliation(s)
- David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK.
| | - Chris Barkus
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Ahmed Eltokhi
- Department of Biomedical Sciences, School of Medicine, Mercer University, Columbus, GA, USA
| |
Collapse
|
11
|
Seyed-Razavi Y, Kenyon BM, Qiu F, Harris DL, Hamrah P. A novel animal model of neuropathic corneal pain-the ciliary nerve constriction model. Front Neurosci 2023; 17:1265708. [PMID: 38144209 PMCID: PMC10749205 DOI: 10.3389/fnins.2023.1265708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Neuropathic pain arises as a result of peripheral nerve injury or altered pain processing within the central nervous system. When this phenomenon affects the cornea, it is referred to as neuropathic corneal pain (NCP), resulting in pain, hyperalgesia, burning, and photoallodynia, severely affecting patients' quality of life. To date there is no suitable animal model for the study of NCP. Herein, we developed an NCP model by constriction of the long ciliary nerves innervating the eye. Methods Mice underwent ciliary nerve constriction (CNC) or sham procedures. Safety was determined by corneal fluorescein staining to assess ocular surface damage, whereas Cochet-Bonnet esthesiometry and confocal microscopy assessed the function and structure of corneal nerves, respectively. Efficacy was assessed by paw wipe responses within 30 seconds of applying hyperosmolar (5M) saline at Days 3, 7, 10, and 14 post-constriction. Additionally, behavior was assessed in an open field test (OFT) at Days 7, 14, and 21. Results CNC resulted in significantly increased response to hyperosmolar saline between groups (p < 0.0001), demonstrating hyperalgesia and induction of neuropathic pain. Further, animals that underwent CNC had increased anxiety-like behavior in an open field test compared to controls at the 14- and 21-Day time-points (p < 0.05). In contrast, CNC did not result in increased corneal fluorescein staining or decreased sensation as compared to sham controls (p > 0.05). Additionally, confocal microscopy of corneal whole-mounts revealed that constriction resulted in only a slight reduction in corneal nerve density (p < 0.05), compared to naïve and sham groups. Discussion The CNC model induces a pure NCP phenotype and may be a useful model for the study of NCP, recapitulating features of NCP, including hyperalgesia in the absence of ocular surface damage, and anxiety-like behavior.
Collapse
Affiliation(s)
- Yashar Seyed-Razavi
- Center for Translational Ocular Immunology, Tufts Medical Center, Boston, MA, United States
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
| | - Brendan M. Kenyon
- Center for Translational Ocular Immunology, Tufts Medical Center, Boston, MA, United States
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, United States
| | - Fangfang Qiu
- Center for Translational Ocular Immunology, Tufts Medical Center, Boston, MA, United States
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
| | - Deshea L. Harris
- Center for Translational Ocular Immunology, Tufts Medical Center, Boston, MA, United States
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Boston, MA, United States
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, United States
- Departments of Neuroscience and Immunology, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
12
|
Sheng JA, Handa RJ, Tobet SA. Evaluating different models of maternal stress on stress-responsive systems in prepubertal mice. Front Neurosci 2023; 17:1292642. [PMID: 38130695 PMCID: PMC10733493 DOI: 10.3389/fnins.2023.1292642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Maternal adversity during pregnancy influences neurodevelopment in human and model animal offspring. Adversity can result from stressors coming from many different directions ranging from environmental to nutritional and physiological to immune (e.g., infection). Most stressors result in fetal overexposure to glucocorticoids that have been directly linked to long- and short-term negative impacts on neurological health of offspring. Neuropsychiatric diseases postulated to have fetal origins are diverse and include such things cardiovascular disease, obesity, affective disorders, and metabolic and immune disorders. Methods The experiments in the current study compare 3 stressors: prenatal exposure to dexamethasone (DEX), maternal high fat diet (HFD), and maternal caloric restriction (CR). Offspring of mothers with these treatments were examined prepubertally to evaluate stress responsiveness and stress-related behaviors in in male and female mice. Results Prenatal exposure to synthetic glucocorticoid, DEX, resulted in decreased neonatal body weights, reduced social interaction behavior, and hypoactive stress response offspring exposed to maternal DEX. Maternal CR resulted in decreased body weights and social interaction behavior in males and females and increased anxiety-like behavior and acute stress response only in males. HFD resulted in altered body weight gain in both sex offspring with decreased anxiety-like behavior in a female-biased manner. Discussion The idea that glucocorticoid responses to different stressors might serve as a common stimulus across stress paradigms is insufficient, given that different modes of prenatal stress produced differential effects. Opposite nutritional stressors produced similar outcomes for anxiety-like behavior in both sexes, social-like behavior in females, and a hyperactive adrenal stress response in males. One common theme among the three models of maternal stress (DEX, CR, and HFD) was consistent data showing their role in activating the maternal and fetal immune response. By tuning in on the more immediate immunological aspect on the developing fetus (e.g., hormones, cytokines), additional studies may tease out more direct outcomes of maternal stress in rodents and increase their translational value to human studies.
Collapse
Affiliation(s)
- Julietta A. Sheng
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Robert J. Handa
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Innovation Center on Sex Differences in Medicine, Mass General Hospital, Cambridge, MA, United States
| |
Collapse
|
13
|
Ryherd GL, Bunce AL, Edwards HA, Baumgartner NE, Lucas EK. Sex differences in avoidance behavior and cued threat memory dynamics in mice: Interactions between estrous cycle and genetic background. Horm Behav 2023; 156:105439. [PMID: 37813043 PMCID: PMC10810684 DOI: 10.1016/j.yhbeh.2023.105439] [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: 05/20/2023] [Revised: 09/11/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
Anxiety disorders are the most prevalent mental illnesses worldwide, exhibit high heritability, and affect twice as many women as men. To evaluate potential interactions between genetic background and cycling ovarian hormones on sex differences in susceptibility to negative valence behaviors relevant to anxiety disorders, we assayed avoidance behavior and cued threat memory dynamics in gonadally-intact adult male and female mice across four common inbred mouse strains: C57Bl/6J, 129S1/SVlmJ, DBA/2J, and BALB/cJ. Independent of sex, C57Bl/6J mice exhibited low avoidance but high threat memory, 129S1/SvlmJ mice high avoidance and high threat memory, DBA/2J mice low avoidance and low threat memory, and BALB/cJ mice high avoidance but low threat memory. Within-strain comparisons revealed reduced avoidance behavior in the high hormone phase of the estrous cycle (proestrus) compared to all other estrous phases in all strains except DBA/2J, which did not exhibit cycle-dependent behavioral fluctuations. Robust and opposing sex differences in threat conditioning and extinction training were found in the C57Bl/6J and 129S1/SvlmJ lines, whereas no sex differences were observed in the DBA/2J or BALB/cJ lines. C57Bl/6J males exhibited enhanced acute threat memory, whereas 129S1/SvlmJ females exhibited enhanced sustained threat memory, compared to their sex-matched littermates. These effects were not mediated by estrous cycle stage or sex differences in active versus passive defensive behavioral responses. Our data demonstrate that core features of behavioral endophenotypes relevant to anxiety disorders, such as avoidance and threat memory, are genetically driven yet dissociable and can be influenced further by cycling ovarian hormones.
Collapse
Affiliation(s)
- Garret L Ryherd
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Averie L Bunce
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Haley A Edwards
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Nina E Baumgartner
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA; Department of Psychiatry & Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth K Lucas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA; Department of Psychiatry & Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
14
|
Reiber M, von Schumann L, Buchecker V, Boldt L, Gass P, Bleich A, Talbot SR, Potschka H. Evidence-based comparative severity assessment in young and adult mice. PLoS One 2023; 18:e0285429. [PMID: 37862304 PMCID: PMC10588901 DOI: 10.1371/journal.pone.0285429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/21/2023] [Indexed: 10/22/2023] Open
Abstract
In animal-based research, welfare assessments are essential for ethical and legal reasons. However, accurate assessment of suffering in laboratory animals is often complicated by the multidimensional character of distress and pain and the associated affective states. The present study aimed to design and validate multidimensional composite measure schemes comprising behavioral and biochemical parameters based on a bioinformatics approach. Published data sets from induced and genetic mouse models of neurological and psychiatric disorders were subjected to a bioinformatics workflow for cross-model analyses. ROC analyses pointed to a model-specific discriminatory power of selected behavioral parameters. Principal component analyses confirmed that the composite measure schemes developed for adult or young mice provided relevant information with the level of group separation reflecting the expected severity levels. Finally, the validity of the composite measure schemes developed for adult and young mice was further confirmed by k-means-based clustering as a basis for severity classification. The classification systems allowed the allocation of individual animals to different severity levels and a direct comparison of animal groups and other models. In conclusion, the bioinformatics approach confirmed the suitability of the composite measure schemes for evidence-based comparative severity assessment in adult and young mice. In particular, we demonstrated that the composite measure schemes provide a basis for an individualized severity classification in control and experimental groups allowing direct comparison of severity levels across different induced or genetic models. An online tool (R package) is provided, allowing the application of the bioinformatics approach to severity assessment data sets regardless of the parameters or models used. This tool can also be used to validate refinement measures.
Collapse
Affiliation(s)
- Maria Reiber
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Lara von Schumann
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Verena Buchecker
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Lena Boldt
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Peter Gass
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Steven Roger Talbot
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| |
Collapse
|
15
|
Jiménez-Loygorri JI, Benítez-Fernández R, Viedma-Poyatos Á, Zapata-Muñoz J, Villarejo-Zori B, Gómez-Sintes R, Boya P. Mitophagy in the retina: Viewing mitochondrial homeostasis through a new lens. Prog Retin Eye Res 2023; 96:101205. [PMID: 37454969 DOI: 10.1016/j.preteyeres.2023.101205] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial function is key to support metabolism and homeostasis in the retina, an organ that has one of the highest metabolic rates body-wide and is constantly exposed to photooxidative damage and external stressors. Mitophagy is the selective autophagic degradation of mitochondria within lysosomes, and can be triggered by distinct stimuli such as mitochondrial damage or hypoxia. Here, we review the importance of mitophagy in retinal physiology and pathology. In the developing retina, mitophagy is essential for metabolic reprogramming and differentiation of retina ganglion cells (RGCs). In basal conditions, mitophagy acts as a quality control mechanism, maintaining a healthy mitochondrial pool to meet cellular demands. We summarize the different autophagy- and mitophagy-deficient mouse models described in the literature, and discuss the potential role of mitophagy dysregulation in retinal diseases such as glaucoma, diabetic retinopathy, retinitis pigmentosa, and age-related macular degeneration. Finally, we provide an overview of methods used to monitor mitophagy in vitro, ex vivo, and in vivo. This review highlights the important role of mitophagy in sustaining visual function, and its potential as a putative therapeutic target for retinal and other diseases.
Collapse
Affiliation(s)
- Juan Ignacio Jiménez-Loygorri
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Rocío Benítez-Fernández
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland
| | - Álvaro Viedma-Poyatos
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Juan Zapata-Muñoz
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Raquel Gómez-Sintes
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Patricia Boya
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Departament of Neuroscience and Movement Science, Faculty of Science and Medicine, University of Fribourg, 1700, Fribourg, Switzerland.
| |
Collapse
|
16
|
Gonzalez L, Sébrié C, Laroche S, Vaillend C, Poirier R. Delayed postnatal brain development and ontogenesis of behavior and cognition in a mouse model of intellectual disability. Neurobiol Dis 2023:106163. [PMID: 37270162 DOI: 10.1016/j.nbd.2023.106163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/05/2023] Open
Abstract
Intellectual disability (ID) is a neurodevelopmental disorder associated with impaired cognitive and adaptive behaviors and represents a major medical issue. Although ID-patients develop behavioral problems and are diagnosed during childhood, most behavioral studies in rodent models have been conducted in adulthood, missing precocious phenotypes expressed during this critical time-window characterized by intense brain plasticity. Here, we selectively assessed postnatal ontogenesis of behavioral and cognitive processes, as well as postnatal brain development in the male Rsk2-knockout mouse model of the Coffin-Lowry syndrome, an X-linked disorder characterized by ID and neurological abnormalities. While Rsk2-knockout mice were born healthy, a longitudinal MRI study revealed a transient secondary microcephaly and a persistent reduction of hippocampal and cerebellar volumes. Specific behavioral parameters from postnatal day 4 (P4) unveiled delayed acquisition of sensory-motor functions and alterations of spontaneous and cognitive behaviors during adolescence, which together, represent hallmarks of neurodevelopmental disorders. Together, our results suggest for the first time that RSK2, an effector of the MAPK signaling pathways, plays a crucial role in brain and cognitive postnatal development. This study also provides new relevant measures to characterize postnatal cognitive development of mouse models of ID and to design early therapeutic approaches.
Collapse
Affiliation(s)
- Laurine Gonzalez
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
| | - Catherine Sébrié
- Université Paris-Saclay CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 91401 Orsay, France
| | - Serge Laroche
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
| | - Roseline Poirier
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France.
| |
Collapse
|
17
|
Elzinga SE, Koubek EJ, Hayes JM, Carter A, Mendelson FE, Webber-Davis I, Lentz SI, Feldman EL. Modeling the innate inflammatory cGAS/STING pathway: sexually dimorphic effects on microglia and cognition in obesity and prediabetes. Front Cell Neurosci 2023; 17:1167688. [PMID: 37206668 PMCID: PMC10188944 DOI: 10.3389/fncel.2023.1167688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/06/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction The prevalence of obesity, prediabetes, and diabetes continues to grow worldwide. These metabolic dysfunctions predispose individuals to neurodegenerative diseases and cognitive impairment, including dementias such as Alzheimer's disease and Alzheimer's disease related dementias (AD/ADRD). The innate inflammatory cGAS/STING pathway plays a pivotal role in metabolic dysfunction and is an emerging target of interest in multiple neurodegenerative diseases, including AD/ADRD. Therefore, our goal was to establish a murine model to specifically target the cGAS/STING pathway to study obesity- and prediabetes-induced cognitive impairment. Methods We performed two pilot studies in cGAS knockout (cGAS-/-) male and female mice designed to characterize basic metabolic and inflammatory phenotypes and examine the impact of high-fat diet (HFD) on metabolic, inflammatory, and cognitive parameters. Results cGAS-/- mice displayed normal metabolic profiles and retained the ability to respond to inflammatory stimuli, as indicated by an increase in plasma inflammatory cytokine production in response to lipopolysaccharide injection. HFD feeding caused expected increases in body weight and decreases in glucose tolerance, although onset was accelerated in females versus males. While HFD did not increase plasma or hippocampal inflammatory cytokine production, it did alter microglial morphology to a state indicative of activation, particularly in female cGAS-/- mice. However, HFD negatively impacted cognitive outcomes in male, but not female animals. Discussion Collectively, these results suggest that cGAS-/- mice display sexually dimorphic responses to HFD, possibly based on differences in microglial morphology and cognition.
Collapse
Affiliation(s)
- Sarah E. Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Emily J. Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - A. Carter
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Faye E. Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Ian Webber-Davis
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Stephen I. Lentz
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
18
|
Ossorio-Salazar VA, D'Hooge R. Methodological shortcomings of preclinical research on chemotherapy-induced cognitive impairment. Neurosci Biobehav Rev 2023; 150:105198. [PMID: 37105339 DOI: 10.1016/j.neubiorev.2023.105198] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 04/29/2023]
Affiliation(s)
- Victoria A Ossorio-Salazar
- Laboratory of Biological Psychology & Leuven Brain Institute, Faculty of Psychology, KU Leuven, Tiensestraat 102, 3000 Leuven, Belgium.
| | - Rudi D'Hooge
- Laboratory of Biological Psychology & Leuven Brain Institute, Faculty of Psychology, KU Leuven, Tiensestraat 102, 3000 Leuven, Belgium.
| |
Collapse
|
19
|
Woodward E, Rangel-Barajas C, Ringland A, Logrip ML, Coutellier L. Sex-Specific Timelines for Adaptations of Prefrontal Parvalbumin Neurons in Response to Stress and Changes in Anxiety- and Depressive-Like Behaviors. eNeuro 2023; 10:ENEURO.0300-22.2023. [PMID: 36808099 PMCID: PMC9997696 DOI: 10.1523/eneuro.0300-22.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 02/22/2023] Open
Abstract
Women are twice as likely as men to experience emotional dysregulation after stress, resulting in substantially higher psychopathology for equivalent lifetime stress exposure, yet the mechanisms underlying this vulnerability remain unknown. Studies suggest changes in medial prefrontal cortex (mPFC) activity as a potential contributor. Whether maladaptive changes in inhibitory interneurons participate in this process, and whether adaptations in response to stress differ between men and women, producing sex-specific changes in emotional behaviors and mPFC activity, remained undetermined. This study examined whether unpredictable chronic mild stress (UCMS) in mice differentially alters behavior and mPFC parvalbumin (PV) interneuron activity by sex, and whether the activity of these neurons drives sex-specific behavioral changes. Four weeks of UCMS increased anxiety-like and depressive-like behaviors associated with FosB activation in mPFC PV neurons, particularly in females. After 8 weeks of UCMS, both sexes displayed these behavioral and neural changes. Chemogenetic activation of PV neurons in UCMS-exposed and nonstressed males induced significant changes in anxiety-like behaviors. Importantly, patch-clamp electrophysiology demonstrated altered excitability and basic neural properties on the same timeline as the emergence of behavioral effects: changes in females after 4 weeks and in males after 8 weeks of UCMS. These findings show, for the first time, that sex-specific changes in the excitability of prefrontal PV neurons parallel the emergence of anxiety-like behavior, revealing a potential novel mechanism underlying the enhanced vulnerability of females to stress-induced psychopathology and supporting further investigation of this neuronal population to identify new therapeutic targets for stress disorders.
Collapse
Affiliation(s)
- Emma Woodward
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| | - Claudia Rangel-Barajas
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Amanda Ringland
- Department of Psychology, The Ohio State University, Columbus, Ohio 43210
| | - Marian L Logrip
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Laurence Coutellier
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Department of Psychology, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
20
|
Liu S, Crawford J, Tao F. Assessing Orofacial Pain Behaviors in Animal Models: A Review. Brain Sci 2023; 13:390. [PMID: 36979200 PMCID: PMC10046781 DOI: 10.3390/brainsci13030390] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/25/2022] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Orofacial pain refers to pain occurring in the head and face, which is highly prevalent and represents a challenge to clinicians, but its underlying mechanisms are not fully understood, and more studies using animal models are urgently needed. Currently, there are different assessment methods for analyzing orofacial pain behaviors in animal models. In order to minimize the number of animals used and maximize animal welfare, selecting appropriate assessment methods can avoid repeated testing and improve the reliability and accuracy of research data. Here, we summarize different methods for assessing spontaneous pain, evoked pain, and relevant accompanying dysfunction, and discuss their advantages and disadvantages. While the behaviors of orofacial pain in rodents are not exactly equivalent to the symptoms displayed in patients with orofacial pain, animal models and pain behavioral assessments have advanced our understanding of the pathogenesis of such pain.
Collapse
Affiliation(s)
| | | | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| |
Collapse
|
21
|
Collier JJ, Oláhová M, McWilliams TG, Taylor RW. Mitochondrial signalling and homeostasis: from cell biology to neurological disease. Trends Neurosci 2023; 46:137-152. [PMID: 36635110 DOI: 10.1016/j.tins.2022.12.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/18/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023]
Abstract
Efforts to understand how mitochondrial dysfunction contributes to neurodegeneration have primarily focussed on the role of mitochondria in neuronal energy metabolism. However, progress in understanding the etiological nature of emerging mitochondrial functions has yielded new ideas about the mitochondrial basis of neurological disease. Studies aimed at deciphering how mitochondria signal through interorganellar contacts, vesicular trafficking, and metabolic transmission have revealed that mitochondrial regulation of immunometabolism, cell death, organelle dynamics, and neuroimmune interplay are critical determinants of neural health. Moreover, the homeostatic mechanisms that exist to protect mitochondrial health through turnover via nanoscale proteostasis and lysosomal degradation have become integrated within mitochondrial signalling pathways to support metabolic plasticity and stress responses in the nervous system. This review highlights how these distinct mitochondrial pathways converge to influence neurological health and contribute to disease pathology.
Collapse
Affiliation(s)
- Jack J Collier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | - Monika Oláhová
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Thomas G McWilliams
- Translational Stem Cell Biology & Metabolism Program, Research Programs Unit, University of Helsinki, Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and Children, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
22
|
Ding X, Wen Y, Ma X, Zhang Y, Cheng Y, Liu Z, Hu W, Xia Y. Pyridoxal 5'-phosphate alleviates prenatal pyridaben exposure-induced anxiety-like behaviors in offspring. ENVIRONMENTAL SCIENCE AND ECOTECHNOLOGY 2023; 13:100224. [PMID: 36437888 PMCID: PMC9691908 DOI: 10.1016/j.ese.2022.100224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 06/16/2023]
Abstract
Pyridaben (PY) is a widely used organochlorine acaricide, which can be detected in the peripheral blood of pregnant women. Available evidence suggests that PY has reproductive toxicity. However, it remains uncertain whether prenatal PY exposure impacts neurobehavioral development in offspring. Here, we administered PY to pregnant mice at a dose of 0.5 and 5 mg kg-1 day-1 via gavage and observed anxiety-like behaviors in PY offspring aged five weeks. We then integrated the metabolome and transcriptome of the offspring's brain to explore the underlying mechanism. Metabolome data indicated that the vitamin B6 metabolism pathway was significantly affected, and the pyridoxal 5'-phosphate (PLP) concentration and the active form of vitamin B6 was significantly reduced. Moreover, the transcriptome data showed that both PLP generation-related Pdxk and anxiety-related Gad1 were significantly down-regulated. Meanwhile, there was a decreasing trend in the concentration of GABA in the hippocampal DG region. Next, we supplemented PLP at a dose of 20 mg kg-1 day-1 to the PY offspring via intraperitoneal injection at three weeks. We found up-regulated expression of Pdxk and Gad1 and restored anxiety-like behaviors. This study suggests that prenatal exposure to PY can disrupt vitamin B6 metabolism, reduce the concentration of PLP, down-regulate the expression levels of Pdxk and Gad1, inhibit the production of GABA, and ultimately lead to anxiety-like behaviors in offspring.
Collapse
Affiliation(s)
- Xingwang Ding
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Ya Wen
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Department of Chronic Non-Communicable Disease Control, Wuxi Liangxi District Center for Disease Control and Prevention, Wuxi, 214011, China
| | - Xuan Ma
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Yuepei Zhang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Yuting Cheng
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Zhaofeng Liu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Weiyue Hu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| |
Collapse
|
23
|
Clapcote SJ. How can we obtain truly translational mouse models to improve clinical outcomes in schizophrenia? Dis Model Mech 2022; 15:dmm049970. [PMID: 36441105 PMCID: PMC10655820 DOI: 10.1242/dmm.049970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Schizophrenia is a serious mental illness affecting 0.7% of the world's population. Despite over 50 years of schizophrenia drug identification and development, there have been no fundamental advances in the treatment of schizophrenia since the 1980s. Complex genetic aetiology and elusive pathomechanisms have made it difficult for researchers to develop models that sufficiently reflect pathophysiology to support effective drug discovery. However, recent large-scale, well-powered genomic studies have identified risk genes that represent tractable entry points to decipher disease mechanisms in heterogeneous patient populations and develop targeted treatments. Replicating schizophrenia-associated gene variants in mouse models is an important strategy to start understanding their pathogenicity and role in disease biology. Furthermore, longitudinal studies in a wide range of genetic mouse models from early postnatal life are required to assess the progression of this disease through developmental stages to improve early diagnostic strategies and enable preventative measures. By expanding and refining our approach to schizophrenia research, we can improve prevention strategies and treatment of this debilitating disease.
Collapse
|
24
|
Reiber M, Miljanovic N, Schönhoff K, Palme R, Potschka H. Behavioral phenotyping of young Scn1a haploinsufficient mice. Epilepsy Behav 2022; 136:108903. [PMID: 36240579 DOI: 10.1016/j.yebeh.2022.108903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
Dravet syndrome is a rare, severe, infancy-onset epileptic encephalopathy associated with a high premature mortality. In most patients, Dravet syndrome is caused by a heterozygous loss-of-function mutation in the SCN1A gene encoding the alpha 1 subunit of the sodium channel. Of the variety of SCN1A variants identified in patients with Dravet syndrome, SCN1A missense mutations occur in one-third of cases. The novel Scn1a-A1783V mouse model of Dravet syndrome carries the human Ala1783Val missense variant. Recently, the behavioral phenotype of Scn1a-A1783V haploinsufficient adult mice has been characterized, which may provide a valuable basis for assessment of novel therapeutic approaches. However, there is still limited information on the developmental course of behavioral alterations in the Scn1a-A1783V mouse model, which is of particular relevance for conclusions about face validity and severity classification of the model. Based on reference data from young wildtype mice, we analyzed selected behavioral parameters and fecal corticosterone metabolites in the Scn1a-A1783V mouse model during post-weaning development. Differences in the preference for a sweet saccharin solution between Dravet mice and wildtype mice were observed once mice reached sexual maturity. Nest building behavior was already influenced by the Scn1a genotype during prepubescence. Sexually mature Dravet mice showed a significantly reduced burrowing performance as compared to their wildtype littermates. In the open-field test, pronounced hyperactivity and increased thigmotactic behavior were evident in prepubescent and sexually mature Dravet mice. Analysis of Irwin scores revealed several genotype-dependent changes in handling-associated parameters during the course of adolescence. The information obtained provides insight into the age-dependence of behavioral patterns in the novel Scn1a-A1783V mouse model of Dravet syndrome. In addition, the dataset confirms the suitability of the applied behavioral composite measure scheme for evidence-based assessment of cumulative severity in genetic mouse lines.
Collapse
Affiliation(s)
- Maria Reiber
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Nina Miljanovic
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Katharina Schönhoff
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Rupert Palme
- Department of Biomedical Sciences, Unit of Physiology, Pathophysiology and Experimental Endocrinology, University of Veterinary Medicine, Vienna, Austria
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany.
| |
Collapse
|
25
|
Herrera-Rivero M, Bohn L, Witten A, Jüngling K, Kaiser S, Richter SH, Stoll M, Sachser N. Transcriptional profiles in the mouse amygdala after a cognitive judgment bias test largely depend on the genotype. Front Mol Neurosci 2022; 15:1025389. [DOI: 10.3389/fnmol.2022.1025389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
Background: The amygdala is crucial for emotional cognitive processing. Affective or emotional states can bias cognitive processes, including attention, memory, and decision-making. This can result in optimistic or pessimistic behaviors that are partially driven by the activation of the amygdala. The resulting emotional cognitive bias is a common feature of anxiety and mood disorders, both of which are interactively influenced by genetic and environmental factors. It is also known that emotional cognitive biases can be influenced by environmental factors. However, little is known about the effects of genetics and/or gene-environment interactions on emotional cognitive biases. We investigated the effects of the genetic background and environmental enrichment on the transcriptional profiles of the mouse amygdala following a well-established cognitive bias test.Methods: Twenty-four female C57BL/6J and B6D2F1N mice were housed either in standard (control) conditions or in an enriched environment. After appropriate training, the cognitive bias test was performed on 19 mice that satisfactorily completed the training scheme to assess their responses to ambiguous cues. This allowed us to calculate an “optimism score” for each mouse. Subsequently, we dissected the anterior and posterior portions of the amygdala to perform RNA-sequencing for differential expression and other statistical analyses.Results: In general, we found only minor changes in the amygdala’s transcriptome associated with the levels of optimism in our mice. In contrast, we observed wide molecular effects of the genetic background in both housing environments. The C57BL/6J animals showed more transcriptional changes in response to enriched environments than the B6D2F1N mice. We also generally found more dysregulated genes in the posterior than in the anterior portion of the amygdala. Gene set overrepresentation analyses consistently implicated cellular metabolic responses and immune processes in the differences observed between mouse strains, while processes favoring neurogenesis and neurotransmission were implicated in the responses to environmental enrichment. In a correlation analysis, lipid metabolism in the anterior amygdala was suggested to influence the levels of optimism.Conclusions: Our observations underscore the importance of selecting appropriate animal models when performing molecular studies of affective conditions or emotional states, and suggest an important role of immune and stress responses in the genetic component of emotion regulation.
Collapse
|
26
|
Michetti C, Falace A, Benfenati F, Fassio A. Synaptic genes and neurodevelopmental disorders: From molecular mechanisms to developmental strategies of behavioral testing. Neurobiol Dis 2022; 173:105856. [PMID: 36070836 DOI: 10.1016/j.nbd.2022.105856] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022] Open
Abstract
Synaptopathies are a class of neurodevelopmental disorders caused by modification in genes coding for synaptic proteins. These proteins oversee the process of neurotransmission, mainly controlling the fusion and recycling of synaptic vesicles at the presynaptic terminal, the expression and localization of receptors at the postsynapse and the coupling between the pre- and the postsynaptic compartments. Murine models, with homozygous or heterozygous deletion for several synaptic genes or knock-in for specific pathogenic mutations, have been developed. They have proved to be extremely informative for understanding synaptic physiology, as well as for clarifying the patho-mechanisms leading to developmental delay, epilepsy and motor, cognitive and social impairments that are the most common clinical manifestations of neurodevelopmental disorders. However, the onset of these disorders emerges during infancy and adolescence while the behavioral phenotyping is often conducted in adult mice, missing important information about the impact of synaptic development and maturation on the manifestation of the behavioral phenotype. Here, we review the main achievements obtained by behavioral testing in murine models of synaptopathies and propose a battery of behavioral tests to improve classification, diagnosis and efficacy of potential therapeutic treatments. Our aim is to underlie the importance of studying behavioral development and better focusing on disease onset and phenotypes.
Collapse
Affiliation(s)
- Caterina Michetti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy.
| | - Antonio Falace
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Fassio
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
27
|
Rappe A, McWilliams TG. Mitophagy in the aging nervous system. Front Cell Dev Biol 2022; 10:978142. [PMID: 36303604 PMCID: PMC9593040 DOI: 10.3389/fcell.2022.978142] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/07/2022] [Indexed: 02/01/2024] Open
Abstract
Aging is characterised by the progressive accumulation of cellular dysfunction, stress, and inflammation. A large body of evidence implicates mitochondrial dysfunction as a cause or consequence of age-related diseases including metabolic disorders, neuropathies, various forms of cancer and neurodegenerative diseases. Because neurons have high metabolic demands and cannot divide, they are especially vulnerable to mitochondrial dysfunction which promotes cell dysfunction and cytotoxicity. Mitophagy neutralises mitochondrial dysfunction, providing an adaptive quality control strategy that sustains metabolic homeostasis. Mitophagy has been extensively studied as an inducible stress response in cultured cells and short-lived model organisms. In contrast, our understanding of physiological mitophagy in mammalian aging remains extremely limited, particularly in the nervous system. The recent profiling of mitophagy reporter mice has revealed variegated vistas of steady-state mitochondrial destruction across different tissues. The discovery of patients with congenital autophagy deficiency provokes further intrigue into the mechanisms that underpin neural integrity. These dimensions have considerable implications for targeting mitophagy and other degradative pathways in age-related neurological disease.
Collapse
Affiliation(s)
- Anna Rappe
- Translational Stem Cell Biology and Metabolism Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Thomas G. McWilliams
- Translational Stem Cell Biology and Metabolism Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
28
|
Elzinga SE, Henn R, Murdock BJ, Kim B, Hayes JM, Mendelson F, Webber-Davis I, Teener S, Pacut C, Lentz SI, Feldman EL. cGAS/STING and innate brain inflammation following acute high-fat feeding. Front Immunol 2022; 13:1012594. [PMID: 36248795 PMCID: PMC9556783 DOI: 10.3389/fimmu.2022.1012594] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/16/2022] [Indexed: 02/05/2023] Open
Abstract
Obesity, prediabetes, and diabetes are growing in prevalence worldwide. These metabolic disorders are associated with neurodegenerative diseases, particularly Alzheimer's disease and Alzheimer's disease related dementias. Innate inflammatory signaling plays a critical role in this association, potentially via the early activation of the cGAS/STING pathway. To determine acute systemic metabolic and inflammatory responses and corresponding changes in the brain, we used a high fat diet fed obese mouse model of prediabetes and cognitive impairment. We observed acute systemic changes in metabolic and inflammatory responses, with impaired glucose tolerance, insulin resistance, and alterations in peripheral immune cell populations. Central inflammatory changes included microglial activation in a pro-inflammatory environment with cGAS/STING activation. Blocking gap junctions in neuron-microglial co-cultures significantly decreased cGAS/STING activation. Collectively these studies suggest a role for early activation of the innate immune system both peripherally and centrally with potential inflammatory crosstalk between neurons and glia.
Collapse
Affiliation(s)
- Sarah E. Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Rosemary Henn
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Benjamin J. Murdock
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Faye Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Ian Webber-Davis
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Sam Teener
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Stephen I. Lentz
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, United States
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
29
|
Liu M, Yu C, Zhang Z, Song M, Sun X, Piálek J, Jacob J, Lu J, Cong L, Zhang H, Wang Y, Li G, Feng Z, Du Z, Wang M, Wan X, Wang D, Wang YL, Li H, Wang Z, Zhang B, Zhang Z. Whole-genome sequencing reveals the genetic mechanisms of domestication in classical inbred mice. Genome Biol 2022; 23:203. [PMID: 36163035 PMCID: PMC9511766 DOI: 10.1186/s13059-022-02772-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background The laboratory mouse was domesticated from the wild house mouse. Understanding the genetics underlying domestication in laboratory mice, especially in the widely used classical inbred mice, is vital for studies using mouse models. However, the genetic mechanism of laboratory mouse domestication remains unknown due to lack of adequate genomic sequences of wild mice. Results We analyze the genetic relationships by whole-genome resequencing of 36 wild mice and 36 inbred strains. All classical inbred mice cluster together distinctly from wild and wild-derived inbred mice. Using nucleotide diversity analysis, Fst, and XP-CLR, we identify 339 positively selected genes that are closely associated with nervous system function. Approximately one third of these positively selected genes are highly expressed in brain tissues, and genetic mouse models of 125 genes in the positively selected genes exhibit abnormal behavioral or nervous system phenotypes. These positively selected genes show a higher ratio of differential expression between wild and classical inbred mice compared with all genes, especially in the hippocampus and frontal lobe. Using a mutant mouse model, we find that the SNP rs27900929 (T>C) in gene Astn2 significantly reduces the tameness of mice and modifies the ratio of the two Astn2 (a/b) isoforms. Conclusion Our study indicates that classical inbred mice experienced high selection pressure during domestication under laboratory conditions. The analysis shows the positively selected genes are closely associated with behavior and the nervous system in mice. Tameness may be related to the Astn2 mutation and regulated by the ratio of the two Astn2 (a/b) isoforms. Supplementary Information The online version contains supplementary material available at 10.1186/s13059-022-02772-1.
Collapse
Affiliation(s)
- Ming Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,International Society of Zoological Sciences, Beijing, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Caixia Yu
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.,National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Zhichao Zhang
- Novogene Bioinformatics Institute, Beijing, China.,Glbizzia Biosciences, Beijing, China
| | - Mingjing Song
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiuping Sun
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing, China
| | - Jaroslav Piálek
- House Mouse Group, Research Facility Studenec, Institute of Vertebrate Biology of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jens Jacob
- Julius Kühn-Institute, Federal Research Centre for Cultivated Plants, Institute for Plant Protection in Horticulture and Forests / Institute for Epidemiology and Pathogen Diagnostics, Münster, Germany
| | - Jiqi Lu
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Cong
- Institute of Plant Protection, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Hongmao Zhang
- School of Life Sciences, Central China Normal University, Wuhan, Hubei, China
| | - Yong Wang
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyong Feng
- Plant Protection Research Institute Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Zhenglin Du
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.,National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Meng Wang
- Novogene Bioinformatics Institute, Beijing, China
| | - Xinru Wan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Dawei Wang
- Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hongjun Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306, USA
| | - Bing Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China. .,International Society of Zoological Sciences, Beijing, China. .,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
30
|
Xu M, Qi S, Calhoun V, Dai J, Yu B, Zhang K, Pei M, Li C, Wei Y, Jiang R, Zhi D, Huang Z, Qiu Z, Liang Z, Sui J. Aberrant brain functional and structural developments in MECP2 duplication rats. Neurobiol Dis 2022; 173:105838. [PMID: 35985556 PMCID: PMC9631682 DOI: 10.1016/j.nbd.2022.105838] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Transgenic animal models with homologous etiology provide a promising way to pursue the neurobiological substrates of the behavioral deficits in autism spectrum disorder (ASD). Gain-of-function mutations of MECP2 cause MECP2 duplication syndrome, a severe neurological disorder with core symptoms of ASD. However, abnormal brain developments underlying the autistic-like behavioral deficits of MECP2 duplication syndrome are rarely investigated. To this end, a human MECP2 duplication (MECP2-DP) rat model was created by the bacterial artificial chromosome transgenic method. Functional and structural magnetic resonance imaging (MRI) with high-field were performed on 16 male MECP2-DP rats and 15 male wildtype rats at postnatal 28 days, 42 days, and 56 days old. Multimodal fusion analyses guided by locomotor-relevant metrics and social novelty time separately were applied to identify abnormal brain networks associated with diverse behavioral deficits induced by MECP2 duplication. Aberrant functional developments of a core network primarily composed of the dorsal medial prefrontal cortex (dmPFC) and retrosplenial cortex (RSP) were detected to associate with diverse behavioral phenotypes in MECP2-DP rats. Altered developments of gray matter volume were detected in the hippocampus and thalamus. We conclude that gain-of-function mutations of MECP2 induce aberrant functional activities in the default-mode-like network and aberrant volumetric changes in the brain, resulting in autistic-like behavioral deficits. Our results gain critical insights into the biomarker of MECP2 duplication syndrome and the neurobiological underpinnings of the behavioral deficits in ASD.
Collapse
Affiliation(s)
- Ming Xu
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Shile Qi
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Vince Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia Institute of Technology, Georgia State University, Emory University, Atlanta, GA 30303, USA
| | - Jiankun Dai
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Yu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kaiwei Zhang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengchao Pei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenjian Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Yusheng Wei
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Rongtao Jiang
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Dongmei Zhi
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhimin Huang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Zilong Qiu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhifeng Liang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Jing Sui
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
31
|
Seemiller LR, Logue SF, Gould TJ. Inbred mouse strain differences in alcohol and nicotine addiction-related phenotypes from adolescence to adulthood. Pharmacol Biochem Behav 2022; 218:173429. [PMID: 35820468 PMCID: PMC11524176 DOI: 10.1016/j.pbb.2022.173429] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/18/2022] [Accepted: 07/06/2022] [Indexed: 11/20/2022]
Abstract
Understanding the genetic basis of a predisposition for nicotine and alcohol use across the lifespan is important for public health efforts because genetic contributions may change with age. However, parsing apart subtle genetic contributions to complex human behaviors is a challenge. Animal models provide the opportunity to study the effects of genetic background and age on drug-related phenotypes, while controlling important experimental variables such as amount and timing of drug exposure. Addiction research in inbred, or isogenic, mouse lines has demonstrated genetic contributions to nicotine and alcohol abuse- and addiction-related behaviors. This review summarizes inbred mouse strain differences in alcohol and nicotine addiction-related phenotypes including voluntary consumption/self-administration, initial sensitivity to the drug as measured by sedative, hypothermic, and ataxic effects, locomotor effects, conditioned place preference or place aversion, drug metabolism, and severity of withdrawal symptoms. This review also discusses how these alcohol and nicotine addiction-related phenotypes change from adolescence to adulthood.
Collapse
Affiliation(s)
- Laurel R Seemiller
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Sheree F Logue
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA.
| |
Collapse
|
32
|
Lalonde R, Strazielle C. The Hole-Board Test in Mutant Mice. Behav Genet 2022; 52:158-169. [PMID: 35482162 DOI: 10.1007/s10519-022-10102-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/01/2022] [Indexed: 11/02/2022]
Abstract
First described by Boissier and Simon in (Ther Recreat J 17:1225-1232, 1962), the hole-board has become a recognized test of anxiety and spatial memory. Benzodiazepines acting at the GABAA-BZD site increase hole-pokes in rats and mice, indicating a loss in behavioral inhibition concordant with the behavior of mutant mice deficient in the GABA transporter. Hole-poking also depends on arousal mechanisms dependent on dopaminergic transmission, as indicated by drug and null mutant studies. In addition, the behavior is modified in natural and null mutants affecting the cerebellum as well as null mutants affecting neuropeptides, growth factors, cell adhesion, and inflammation. Further research is required to determine convergences between genetic and pharmacological effects.
Collapse
Affiliation(s)
- Robert Lalonde
- Laboratory of Stress, Immunity, Pathogens (EA7300), Medical School, University of Lorraine, 54500, Vandœuvre-les-Nancy, France
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, Pathogens (EA7300), Medical School, University of Lorraine, 54500, Vandœuvre-les-Nancy, France. .,CHRU Nancy, Vandœuvre-les-Nancy, France.
| |
Collapse
|
33
|
Mickael ME, Bhaumik S, Chakraborti A, Umfress AA, van Groen T, Macaluso M, Totenhagen J, Sorace AG, Bibb JA, Standaert DG, Basu R. RORγt-Expressing Pathogenic CD4 + T Cells Cause Brain Inflammation during Chronic Colitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2054-2066. [PMID: 35379749 PMCID: PMC10103644 DOI: 10.4049/jimmunol.2100869] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/11/2022] [Indexed: 01/09/2023]
Abstract
Neurobehavioral disorders and brain abnormalities have been extensively reported in both Crohn's disease and ulcerative colitis patients. However, the mechanism causing neuropathological disorders in inflammatory bowel disease patients remains unknown. Studies have linked the Th17 subset of CD4+ T cells to brain diseases associated with neuroinflammation and cognitive impairment, including multiple sclerosis, ischemic brain injury, and Alzheimer's disease. To better understand how CD4+ T lymphocytes contribute to brain pathology in chronic intestinal inflammation, we investigated the development of brain inflammation in the T cell transfer model of chronic colitis. Our findings demonstrate that CD4+ T cells infiltrate the brain of colitic Rag1 -/- mice in proportional levels to colitis severity. Colitic mice developed hypothalamic astrogliosis that correlated with neurobehavioral disorders. Moreover, the brain-infiltrating CD4+ T cells expressed Th17 cell transcription factor retinoic acid-related orphan receptor γt (RORγt) and displayed a pathogenic Th17 cellular phenotype similar to colonic Th17 cells. Adoptive transfer of RORγt-deficient naive CD4+ T cells failed to cause brain inflammation and neurobehavioral disorders in Rag1 -/- recipients, with significantly less brain infiltration of CD4+ T cells. The finding is mirrored in chronic dextran sulfate sodium-induced colitis in Rorcfl/fl Cd4-Cre mice that showed lower frequency of brain-infiltrating CD4+ T cells and astrogliosis despite onset of significantly more severe colitis compared with wild-type mice. These findings suggest that pathogenic RORγt+CD4+ T cells that aggravate colitis migrate preferentially into the brain, contributing to brain inflammation and neurobehavioral disorders, thereby linking colitis severity to neuroinflammation.
Collapse
Affiliation(s)
| | - Suniti Bhaumik
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Ayanabha Chakraborti
- Department of Surgery, Neuroscience, and Neurology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Alan A Umfress
- Department of Surgery, Neuroscience, and Neurology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Thomas van Groen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Matthew Macaluso
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL
| | - John Totenhagen
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL.,Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL; and
| | - James A Bibb
- Department of Surgery, Neuroscience, and Neurology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL.,Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Rajatava Basu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL;
| |
Collapse
|
34
|
Cabrera-Muñoz EA, Olvera-Hernández S, Vega-Rivera NM, Meneses-San Juan D, Reyes-Haro D, Ortiz-López L, Ramírez Rodríguez GB. Environmental Enrichment Differentially Activates Neural Circuits in FVB/N Mice, Inducing Social Interaction in Females but Agonistic Behavior in Males. Neurochem Res 2022; 47:781-794. [PMID: 34978003 DOI: 10.1007/s11064-021-03487-8] [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: 08/12/2021] [Revised: 10/14/2021] [Accepted: 11/09/2021] [Indexed: 01/17/2023]
Abstract
Environmental enrichment induces behavioral and structural modifications in rodents and influences the capability of mice to cope with stress. However, little is understood about hippocampal neurogenesis and the appearance of social/agonistic (aggressive) behavior upon activation of different neuronal circuits in FVB/N mice. Thus, in this study we hypothesized that environmental enrichment differentially regulates neurogenesis, neural circuit activation and social/agonistic behavior in male and female FVB/N mice. We explored the (1) neurogenic process as an indicative of neuroplasticity, (2) neuronal activation in the limbic system, and (3) social behavior using the resident-intruder test. On postnatal day 23 (PD23), mice were assigned to one of two groups: Standard Housing or Environmental Enrichment. At PD53, rodents underwent the resident-intruder test to evaluate social behaviors. Results revealed that environmental enrichment increased neurogenesis and social interaction in females. In males, environmental enrichment increased neurogenesis and agonistic behavior. Enriched male mice expressed higher levels of agonistic-related behavior than female mice housed under the same conditions. Neural circuit analysis showed lower activation in the amygdala of enriched males and higher activation in enriched females than their respective controls. Enriched females also showed higher activation in the frontal cortex without differences in male groups. Moreover, the insular cortex was less activated in females than in males. Thus, our results indicate that environmental enrichment has different effects on neuroplasticity and social/agonistic behavior in FVB/N mice, suggesting the relevance of sexual dimorphism in response to environmental stimuli.
Collapse
Affiliation(s)
- Edith Araceli Cabrera-Muñoz
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Sandra Olvera-Hernández
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Nelly Maritza Vega-Rivera
- Laboratorio of Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco 101, C.P. 14370, México City, México
| | - David Meneses-San Juan
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Daniel Reyes-Haro
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología. Universidad Nacional Autónoma de México, Campus Juriquilla. Boulevard Juriquilla 3001, C.P. 76230, Juriquilla, Querétaro, México
| | - Leonardo Ortiz-López
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Gerardo Bernabé Ramírez Rodríguez
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México.
| |
Collapse
|
35
|
Development of behavioral patterns in young C57BL/6J mice: a home cage-based study. Sci Rep 2022; 12:2550. [PMID: 35169182 PMCID: PMC8847349 DOI: 10.1038/s41598-022-06395-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 11/23/2022] Open
Abstract
Evidence exists that behavioral patterns only stabilize once mice reach adulthood. Detailed information about the course of behavioral patterns is of particular relevance for neuroscientific research and for the assessment of cumulative severity in genetically modified mice. The analysis considered five age groups focusing on behavioral assessments in the animals’ familiar home cage environment during the adolescence phase. We confirmed age- and sex-specific differences for several of the behavioral parameters and fecal corticosterone metabolites. Interestingly, an age-dependent decline in saccharin preference was detected in female mice. Regardless of sex, relevant levels of burrowing activity were only observed during later developmental phases. The development of nest complexity following the offer of new material was affected by age in female mice. In female and male mice, an age-dependency was evident for wheel running reaching a peak at P 50. A progressive increase with age was also observed for Open field activity. The data sets provide guidance for behavioral studies and for development of composite measure schemes for evidence-based severity assessment in young mice. Except for the burrowing test, the different behavioral tests can be applied in different age groups during post-weaning development. However, age- and sex-specific characteristics need to be considered.
Collapse
|
36
|
Tran T, Mach J, Gemikonakli G, Wu H, Allore H, Howlett SE, Little CB, Hilmer SN. Diurnal effects of polypharmacy with high drug burden index on physical activities over 23 h differ with age and sex. Sci Rep 2022; 12:2168. [PMID: 35140291 PMCID: PMC8828819 DOI: 10.1038/s41598-022-06039-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 01/18/2023] Open
Abstract
Aging, polypharmacy (concurrent use of ≥ 5 medications), and functional impairment are global healthcare challenges. However, knowledge of the age/sex-specific effects of polypharmacy is limited, particularly on daily physical activities. Using continuous monitoring, we demonstrated how polypharmacy with high Drug Burden Index (DBI-cumulative anticholinergic/sedative exposure) affected behaviors over 23 h in male/female, young/old mice. For comparison, we also evaluated how different drug regimens (polypharmacy/monotherapy) influenced activities in young mice. We found that after 4 weeks of treatment, high DBI (HDBI) polypharmacy decreased exploration (reduced mean gait speed and climbing) during the habituation period, but increased it during other periods, particularly in old mice during the transition to inactivity. After HDBI polypharmacy, mean gait speed consistently decreased throughout the experiment. Some behavioral declines after HDBI were more marked in females than males, indicating treatment × sex interactions. Metoprolol and simvastatin monotherapies increased activities in young mice, compared to control/polypharmacy. These findings highlight that in mice, some polypharmacy-associated behavioral changes are greater in old age and females. The observed diurnal behavioral changes are analogous to drug-induced delirium and sundowning seen in older adults. Future mechanistic investigations are needed to further inform considerations of age, sex, and polypharmacy to optimize quality use of medicines.
Collapse
Affiliation(s)
- Trang Tran
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW, 2065, Australia.
- Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia.
| | - John Mach
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW, 2065, Australia
- Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
| | - Gizem Gemikonakli
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW, 2065, Australia
- Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
| | - Harry Wu
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW, 2065, Australia
- Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
| | - Heather Allore
- Department of Internal Medicine, Yale University, New Haven, CT, 06510, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06510, USA
| | - Susan E Howlett
- Department of Pharmacology and Medicine (Geriatric Medicine), Dalhousie University, Halifax, NS, B3H 2E1, Canada
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW, 2065, Australia
| | - Sarah N Hilmer
- Laboratory of Ageing and Pharmacology, Kolling Institute, Faculty of Medicine and Health, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW, 2065, Australia
- Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
| |
Collapse
|
37
|
Wilcox JM, Consoli DC, Paffenroth KC, Spitznagel BD, Calipari ES, Bowman AB, Harrison FE. Manganese-induced hyperactivity and dopaminergic dysfunction depend on age, sex and YAC128 genotype. Pharmacol Biochem Behav 2022; 213:173337. [PMID: 35063467 PMCID: PMC8833139 DOI: 10.1016/j.pbb.2022.173337] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/17/2021] [Accepted: 01/10/2022] [Indexed: 02/03/2023]
Abstract
Manganese (Mn) is an essential micronutrient but is neurotoxic in excess. Environmental and genetic factors influence vulnerability to Mn toxicity, including sex, age, and the autosomal dominant mutation that causes Huntington disease (HD). To better understand the differential effects of Mn in wild-type (WT) versus YAC128 mice, we examined impacts of Mn exposure across different ages and sexes on disease-relevant behavioral tasks and dopamine dynamics. Young (3-week) and aged (12-month) WT and YAC128 mice received control (70 ppm) or high (2400 ppm) Mn diet for 8 weeks followed by a battery of behavioral tasks. In young female WT mice, high Mn diet induced hyperactivity across two independent behavioral tasks. Changes in the expression of tyrosine hydroxylase (TH) were consistent with the behavioral data in young females such that elevated TH in YAC128 on control diet was decreased by high Mn diet. Aged YAC128 mice showed the expected disease-relevant behavioral impairments in females and decreased TH expression, but we observed no significant effects of Mn diet in either genotype of the aged group. Fast-scan cyclic voltammetry recorded dopamine release and clearance in the nucleus accumbens of eight-month-old WT and YAC128 mice following acute Mn exposure (3×/1 week subcutaneous injections of 50 mg/kg MnCl[2]-tetrahydrate or saline). In WT mice, Mn exposure led to faster dopamine clearance that resembled saline treated YAC128 mice. Mn treatment increased dopamine release only in YAC128 mice, possibly indirectly correcting the faster dopamine clearance observed in saline treated YAC128 mice. The same exposure paradigm led to decreased dopamine and serotonin and metabolites (3-MT, HVA and 5-HIAA) in striatum and increased glutamate in YAC128 mice but not WT mice. These studies confirm an adverse effect of Mn in young, female WT animals and support a role for Mn exposure in stabilizing dopaminergic dysfunction and motivated behavior in early HD.
Collapse
Affiliation(s)
- Jordyn M. Wilcox
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN,corresponding author: Jordyn M. Wilcox, PhD, , 2215 Garland Ave, Medical Research Building IV, 7445, Nashville, TN 37232
| | - David C. Consoli
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | | | - Brittany D. Spitznagel
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | - Erin S. Calipari
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN,Departments of Pharmacology, Molecular Physiology and Biophysics, Psychiatry and Behavioral Sciences; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN
| | - Fiona E. Harrison
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| |
Collapse
|
38
|
Bozhko DV, Myrov VO, Kolchanova SM, Polovian AI, Galumov GK, Demin KA, Zabegalov KN, Strekalova T, de Abreu MS, Petersen EV, Kalueff AV. Artificial intelligence-driven phenotyping of zebrafish psychoactive drug responses. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110405. [PMID: 34320403 DOI: 10.1016/j.pnpbp.2021.110405] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/26/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023]
Abstract
Zebrafish (Danio rerio) are rapidly emerging in biomedicine as promising tools for disease modelling and drug discovery. The use of zebrafish for neuroscience research is also growing rapidly, necessitating novel reliable and unbiased methods of neurophenotypic data collection and analyses. Here, we applied the artificial intelligence (AI) neural network-based algorithms to a large dataset of adult zebrafish locomotor tracks collected previously in a series of in vivo experiments with multiple established psychotropic drugs. We first trained AI to recognize various drugs from a wide range of psychotropic agents tested, and then confirmed prediction accuracy of trained AI by comparing several agents with known similar behavioral and pharmacological profiles. Presenting a framework for innovative neurophenotyping, this proof-of-concept study aims to improve AI-driven movement pattern classification in zebrafish, thereby fostering drug discovery and development utilizing this key model organism.
Collapse
Affiliation(s)
| | | | | | | | | | - Konstantin A Demin
- Institite of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Almazov National Medical Research Center, St. Petersburg, Russia; Neurobiology Program, Sirius University, Sochi, Russia
| | - Konstantin N Zabegalov
- Institite of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Ural Federal University, Ekaterinburg, Russia; Neurobiology Program, Sirius University, Sochi, Russia; Group of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Tatiana Strekalova
- Maastricht University, Maastricht, Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov Moscow State Medical University, Moscow, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil; Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | | | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia; ZENEREI, LLC, Slidell, LA, USA; Group of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia.
| |
Collapse
|
39
|
Pitzer C, Kurpiers B, Eltokhi A. Gait performance of adolescent mice assessed by the CatWalk XT depends on age, strain and sex and correlates with speed and body weight. Sci Rep 2021; 11:21372. [PMID: 34725364 PMCID: PMC8560926 DOI: 10.1038/s41598-021-00625-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022] Open
Abstract
The automatization of behavioral tests assessing motor activity in rodent models is important for providing robust and reproducible results and evaluating new therapeutics. The CatWalk system is an observer-independent, automated and computerized technique for the assessment of gait performance in rodents. This method has previously been used in adult rodent models of CNS-based movement disorders such as Parkinson's and Huntington's diseases. As motor and gait abnormalities in neuropsychiatric disorders are observed during infancy and adolescence, it became important to validate the CatWalk XT in the gait analysis of adolescent mice and unravel factors that may cause variations in gait performance. Three adolescent wild-type inbred mouse strains, C57BL/6N, DBA/2 and FVB/N, were tested using the CatWalk XT (Version 10.6) for suitable detection settings to characterize several gait parameters at P32 and P42. The same detection settings being suitable for C57BL/6N and DBA/2 mice allowed a direct comparison between the two strains. On the other hand, due to their increased body weight and size, FVB/N mice required different detection settings. The CatWalk XT reliably measured the temporal, spatial, and interlimb coordination parameters in the investigated strains during adolescence. Additionally, significant effects of sex, development, speed and body weight within each strain confirmed the sensitivity of motor and gait functions to these factors. The CatWalk gait analysis of rodents during adolescence, taking the effect of age, strain, sex, speed and body weight into consideration, will decrease intra-laboratory discrepancies and increase the face validity of rodent models of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany.
| | - Barbara Kurpiers
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, USA.
| |
Collapse
|
40
|
Eltokhi A, Kurpiers B, Pitzer C. Baseline Depression-Like Behaviors in Wild-Type Adolescent Mice Are Strain and Age but Not Sex Dependent. Front Behav Neurosci 2021; 15:759574. [PMID: 34690714 PMCID: PMC8529326 DOI: 10.3389/fnbeh.2021.759574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Depression is a major neuropsychiatric disorder, decreasing the ability of hundreds of millions of individuals worldwide to function in social, academic, and employment settings. Beyond the alarming public health problem, depression leads to morbidity across the entire age including adolescence and adulthood. Modeling depression in rodents has been used to understand the pathophysiological mechanisms behind this disorder and create new therapeutics. Although women are two times more likely to be diagnosed with depression compared to men, behavioral experiments on rodent models of depression are mainly performed in males based on the assumption that the estrous cycles in females may affect the behavioral outcome and cause an increase in the intrinsic variability compared to males. Still, the inclusion of female rodents in the behavioral analysis is mandatory to establish the origin of sex bias in depression. Here, we investigated the baseline depression-like behaviors in male and female mice of three adolescent wild-type inbred strains, C57BL/6N, DBA/2, and FVB/N, that are typically used as background strains for mouse models of neuropsychiatric disorders. Our experiments, performed at two different developmental stages during adolescence (P22-P26 and P32-P36), revealed strain but no sex differences in a set of depression-related tests, including tail suspension, sucrose preference and forced swim tests. Additionally, the 10-day interval during this sensitive period uncovered a strong impact on the behavioral outcome of C57BL/6N and FVB/N mice, highlighting a significant effect of maturation on behavioral patterns. Since anxiety-related behavioral tests are often performed together with depression tests in mouse models of neuropsychiatric disorders, we extended our study and included hyponeophagia as an anxiety test. Consistent with a previous study revealing sex differences in other anxiety tests in adolescent mice, male and females mice behaved differently in the hyponeophagia test at P27. Our study gives insight into the behavioral experiments assessing depression and stresses the importance of considering strain, age and sex when evaluating neuropsychiatric-like traits in rodent models.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Barbara Kurpiers
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
41
|
Torres ERS, Stanojlovic M, Zelikowsky M, Bonsberger J, Hean S, Mulligan C, Baldauf L, Fleming S, Masliah E, Chesselet MF, Fanselow MS, Richter F. Alpha-synuclein pathology, microgliosis, and parvalbumin neuron loss in the amygdala associated with enhanced fear in the Thy1-aSyn model of Parkinson's disease. Neurobiol Dis 2021; 158:105478. [PMID: 34390837 PMCID: PMC8447919 DOI: 10.1016/j.nbd.2021.105478] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
In Parkinson's disease (PD), the second most common neurodegenerative disorder, non-motor symptoms often precede the development of debilitating motor symptoms and present a severe impact on the quality of life. Lewy bodies containing misfolded α-synuclein progressively develop in neurons throughout the peripheral and central nervous system, which may be correlated with the early development of non-motor symptoms. Among those, increased fear and anxiety is frequent in PD and thought to result from pathology outside the dopaminergic system, which has been the focus of symptomatic treatment to alleviate motor symptoms. Alpha-synuclein accumulation has been reported in the amygdala of PD patients, a brain region critically involved in fear and anxiety. Here we asked whether α-synuclein overexpression alone is sufficient to induce an enhanced fear phenotype in vivo and which pathological mechanisms are involved. Transgenic mice expressing human wild-type α-synuclein (Thy1-aSyn), a well-established model of PD, were subjected to fear conditioning followed by extinction and then tested for extinction memory retention followed by histopathological analysis. Thy1-aSyn mice showed enhanced tone fear across acquisition and extinction compared to wild-type littermates, as well as a trend to less retention of fear extinction. Immunohistochemical analysis of the basolateral nucleus of the amygdala, a nucleus critically involved in tone fear learning, revealed extensive α-synuclein pathology, with accumulation, phosphorylation, and aggregation of α-synuclein in transgenic mice. This pathology was accompanied by microgliosis and parvalbumin neuron loss in this nucleus, which could explain the enhanced fear phenotype. Importantly, this non-motor phenotype was detected in the pre-clinical phase, prior to dopamine loss in Thy1-aSyn mice, thus replicating observations in patients. Results obtained in this study suggest a possible mechanism by which increased anxiety and maladaptive fear processing may occur in PD, opening a door for therapeutic options and further early biomarker research.
Collapse
Affiliation(s)
- Eileen Ruth S Torres
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Milos Stanojlovic
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Moriel Zelikowsky
- Department of Psychology, Staglin Center for Brain and Behavioral Health, UCLA, Los Angeles, CA 90095, USA; Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Jana Bonsberger
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sindalana Hean
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Caitlin Mulligan
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Leonie Baldauf
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sheila Fleming
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Eliezer Masliah
- Department of Neurosciences, UCSD School of Medicine, La Jolla, CA 92093, USA
| | | | - Michael S Fanselow
- Department of Psychology, Staglin Center for Brain and Behavioral Health, UCLA, Los Angeles, CA 90095, USA
| | - Franziska Richter
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, Hanover, Germany.
| |
Collapse
|
42
|
Grieco F, Bernstein BJ, Biemans B, Bikovski L, Burnett CJ, Cushman JD, van Dam EA, Fry SA, Richmond-Hacham B, Homberg JR, Kas MJH, Kessels HW, Koopmans B, Krashes MJ, Krishnan V, Logan S, Loos M, McCann KE, Parduzi Q, Pick CG, Prevot TD, Riedel G, Robinson L, Sadighi M, Smit AB, Sonntag W, Roelofs RF, Tegelenbosch RAJ, Noldus LPJJ. Measuring Behavior in the Home Cage: Study Design, Applications, Challenges, and Perspectives. Front Behav Neurosci 2021; 15:735387. [PMID: 34630052 PMCID: PMC8498589 DOI: 10.3389/fnbeh.2021.735387] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
The reproducibility crisis (or replication crisis) in biomedical research is a particularly existential and under-addressed issue in the field of behavioral neuroscience, where, in spite of efforts to standardize testing and assay protocols, several known and unknown sources of confounding environmental factors add to variance. Human interference is a major contributor to variability both within and across laboratories, as well as novelty-induced anxiety. Attempts to reduce human interference and to measure more "natural" behaviors in subjects has led to the development of automated home-cage monitoring systems. These systems enable prolonged and longitudinal recordings, and provide large continuous measures of spontaneous behavior that can be analyzed across multiple time scales. In this review, a diverse team of neuroscientists and product developers share their experiences using such an automated monitoring system that combines Noldus PhenoTyper® home-cages and the video-based tracking software, EthoVision® XT, to extract digital biomarkers of motor, emotional, social and cognitive behavior. After presenting our working definition of a "home-cage", we compare home-cage testing with more conventional out-of-cage tests (e.g., the open field) and outline the various advantages of the former, including opportunities for within-subject analyses and assessments of circadian and ultradian activity. Next, we address technical issues pertaining to the acquisition of behavioral data, such as the fine-tuning of the tracking software and the potential for integration with biotelemetry and optogenetics. Finally, we provide guidance on which behavioral measures to emphasize, how to filter, segment, and analyze behavior, and how to use analysis scripts. We summarize how the PhenoTyper has applications to study neuropharmacology as well as animal models of neurodegenerative and neuropsychiatric illness. Looking forward, we examine current challenges and the impact of new developments. Examples include the automated recognition of specific behaviors, unambiguous tracking of individuals in a social context, the development of more animal-centered measures of behavior and ways of dealing with large datasets. Together, we advocate that by embracing standardized home-cage monitoring platforms like the PhenoTyper, we are poised to directly assess issues pertaining to reproducibility, and more importantly, measure features of rodent behavior under more ethologically relevant scenarios.
Collapse
Affiliation(s)
| | - Briana J Bernstein
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | | | - Lior Bikovski
- Myers Neuro-Behavioral Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| | - C Joseph Burnett
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jesse D Cushman
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | | | - Sydney A Fry
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Bar Richmond-Hacham
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | | | - Michael J Krashes
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Vaishnav Krishnan
- Laboratory of Epilepsy and Emotional Behavior, Baylor Comprehensive Epilepsy Center, Departments of Neurology, Neuroscience, and Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States
| | - Sreemathi Logan
- Department of Rehabilitation Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Maarten Loos
- Sylics (Synaptologics BV), Amsterdam, Netherlands
| | - Katharine E McCann
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | | | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Thomas D Prevot
- Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Gernot Riedel
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Lianne Robinson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Mina Sadighi
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - William Sonntag
- Department of Biochemistry & Molecular Biology, Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | | | | | - Lucas P J J Noldus
- Noldus Information Technology BV, Wageningen, Netherlands
- Department of Biophysics, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
43
|
Varga TG, de Toledo Simões JG, Siena A, Henrique E, da Silva RCB, Dos Santos Bioni V, Ramos AC, Rosenstock TR. Haloperidol rescues the schizophrenia-like phenotype in adulthood after rotenone administration in neonatal rats. Psychopharmacology (Berl) 2021; 238:2569-2585. [PMID: 34089344 DOI: 10.1007/s00213-021-05880-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
Neuropsychiatric disorders are multifactorial disturbances that encompass several hypotheses, including changes in neurodevelopment. It is known that brain development disturbances during early life can predict psychosis in adulthood. As we have previously demonstrated, rotenone, a mitochondrial complex I inhibitor, could induce psychiatric-like behavior in 60-day-old rats after intraperitoneal injections from the 5th to the 11th postnatal day. Because mitochondrial deregulation is related to psychiatric disorders and the establishment of animal models is a high-value preclinical tool, we investigated the responsiveness of the rotenone (Rot)-treated newborn rats to pharmacological agents used in clinical practice, haloperidol (Hal), and methylphenidate (MPD). Taken together, our data show that Rot-treated animals exhibit hyperlocomotion, decreased social interaction, and diminished contextual fear conditioning response at P60, consistent with positive, negative, and cognitive deficits of schizophrenia (SZ), respectively, that were reverted by Hal, but not MPD. Rot-treated rodents also display a prodromal-related phenotype at P35. Overall, our results seem to present a new SZ animal model as a consequence of mitochondrial inhibition during a critical neurodevelopmental period. Therefore, our study is crucial not only to elucidate the relevance of mitochondrial function in the etiology of SZ but also to fulfill the need for new and trustworthy experimentation models and, likewise, provide possibilities to new therapeutic avenues for this burdensome disorder.
Collapse
Affiliation(s)
- Thiago Garcia Varga
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | | | - Amanda Siena
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, Av. Prof. Lineu Prestes, 1524 - Ed. Biomédicas I, 2º andar, São Paulo, SP, 05508-900, Brazil
| | - Elisandra Henrique
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | | | | | - Aline Camargo Ramos
- Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Tatiana Rosado Rosenstock
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, Av. Prof. Lineu Prestes, 1524 - Ed. Biomédicas I, 2º andar, São Paulo, SP, 05508-900, Brazil. .,Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
44
|
Percelay S, Freret T, Turnbull N, Bouet V, Boulouard M. Combination of MAP6 deficit, maternal separation and MK801 in female mice: A 3-hit animal model of neurodevelopmental disorder with cognitive deficits. Behav Brain Res 2021; 413:113473. [PMID: 34280461 DOI: 10.1016/j.bbr.2021.113473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 11/26/2022]
Abstract
Schizophrenia is a major psychiatric disease still lacking efficient treatment, particularly for cognitive deficits. To go further in research of new treatments that would encompass all the symptoms associated with this pathology, preclinical animal models need to be improved. To date, the aetiology of schizophrenia is unknown, but there is increasing evidence to highlight its multifactorial nature. We built a new neurodevelopmental mouse model gathering a triple factor combination (3-M): a genetic factor (partial deletion of MAP6 gene), an early stress (maternal separation) and a late pharmacological factor (MK801 administration, 0.05 mg/kg, i.p., daily for 5 days). The effects of each factor and of their combination were investigated on several behaviours including cognitive functions. While each individual factor induced slight deficits in one or another behavioural test, 3-M conditioning induces a wider phenotype with hyperlocomotion and cognitive deficits (working memory and social recognition). This study confirms the hypothesis that genetic, environmental and pharmacological factors, even if not deleterious by themselves, could act synergistically to induce a deleterious behavioural phenotype. It moreover encourages the use of such combined models to improve translational research on neurodevelopmental disorders.
Collapse
Affiliation(s)
- Solenn Percelay
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France.
| | - Thomas Freret
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| | - Nicole Turnbull
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| | - Valentine Bouet
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| | - Michel Boulouard
- Normandie Université, UNICAEN, INSERM, COMETE, CYCERON, CHU Caen, 14000, Caen, France
| |
Collapse
|
45
|
Ganley IG, Whitworth AJ, McWilliams TG. Comment on "mt-Keima detects PINK1-PRKN mitophagy in vivo with greater sensitivity than mito-QC". Autophagy 2021; 17:4477-4479. [PMID: 33818280 PMCID: PMC8726702 DOI: 10.1080/15548627.2021.1907269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Ian G Ganley
- MRC Protein Phosphorylation & Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | | | - Thomas G McWilliams
- Translational Stem Cell Biology and Metabolism, Research Programs Unit, Faculty of Medicine,University of Helsinki, Helsinki, Finland.,Department of Anatomy, Faculty of Medicine,University of Helsinki, Helsinki, Finland
| |
Collapse
|
46
|
Comprehensive characterization of motor and coordination functions in three adolescent wild-type mouse strains. Sci Rep 2021; 11:6497. [PMID: 33753800 PMCID: PMC7985312 DOI: 10.1038/s41598-021-85858-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
Neuropsychiatric disorders are often associated with motor and coordination abnormalities that have important implications on the etiology, pathophysiology, and management of these disorders. Although the onset of many neuropsychiatric disorders including autism spectrum disorder, schizophrenia, and attention-deficit hyperactivity disorder emerges mainly during infancy and adolescence, most of the behavioral studies in mice modeling neuropsychiatric phenotypes are performed in adult animals, possibly missing valuable phenotypic information related to the effect of synaptic maturation during development. Here, we examined which behavioral tests assessing both motor and coordination functions can be performed in mice at two different adolescent stages. As strain and sex affect mouse behavior, our experiments covered both male and female mice of three inbred wild-type strains, C57BL/6N, DBA/2, and FVB/N. Adolescent mice of both postnatal days (P)22-30 and P32-40 developmental stages were capable of mastering common motor and coordination tests. However, results differed significantly between strains and sexes. Moreover, the 10-day interval between the two tested cohorts uncovered a strong difference in the behavioral results, confirming the significant impact of maturation on behavioral patterns. Interestingly, the results of distinct behavioral experiments were directly correlated with the weight of mice, which may explain the lack of reproducibility of some behavioral results in genetically-modified mice. Our study paves the way for better reproducibility of behavioral tests by addressing the effect of the developmental stage, strain, sex, and weight of mice on achieving the face validity of neuropsychiatric disorder-associated motor dysfunctions.
Collapse
|
47
|
Sarver DC, Lei X, Wong GW. FAM19A (TAFA): An Emerging Family of Neurokines with Diverse Functions in the Central and Peripheral Nervous System. ACS Chem Neurosci 2021; 12:945-958. [PMID: 33621067 DOI: 10.1021/acschemneuro.0c00757] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cytokines and chemokines have diverse and pleiotropic functions in peripheral tissues and in the brain. Recent studies uncovered a novel family of neuron-derived secretory proteins, or neurokines, distantly related to chemokines. The FAM19A family comprises five ∼12-15 kDa secretory proteins (FAM19A1-5), also known as TAFA1-5, that are predominantly detected in the central and peripheral nervous system. FAM19A expression in the central nervous system is dynamically regulated during development and in the postnatal brain. As secreted ligands, FAM19A proteins appear to bind to different classes of cell surface receptors (e.g., GPCRs and neurexins). Functional studies using gain- and loss-of-function mouse models established nonredundant roles for each FAM19A family member in regulating diverse physiological processes ranging from locomotor activity and food intake to learning and memory, anxiety- and depressive-like behaviors, social communication, repetitive behaviors, and somatosensory functions. This review summarizes major advances as well as the limitations and knowledge gaps in understanding the regulation and diverse biological functions of this conserved family of neurokines.
Collapse
Affiliation(s)
- Dylan C. Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Xia Lei
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - G. William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
48
|
Xu K, Wang M, Zhou W, Pu J, Wang H, Xie P. Chronic D-ribose and D-mannose overload induce depressive/anxiety-like behavior and spatial memory impairment in mice. Transl Psychiatry 2021; 11:90. [PMID: 33531473 PMCID: PMC7854712 DOI: 10.1038/s41398-020-01126-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
The effects of different forms of monosaccharides on the brain remain unclear, though neuropsychiatric disorders undergo changes in glucose metabolism. This study assessed cell viability responses to five commonly consumed monosaccharides-D-ribose (RIB), D-glucose, D-mannose (MAN), D-xylose and L-arabinose-in cultured neuro-2a cells. Markedly decreased cell viability was observed in cells treated with RIB and MAN. We then showed that high-dose administration of RIB induced depressive- and anxiety-like behavior as well as spatial memory impairment in mice, while high-dose administration of MAN induced anxiety-like behavior and spatial memory impairment only. Moreover, significant pathological changes were observed in the hippocampus of high-dose RIB-treated mice by hematoxylin-eosin staining. Association analysis of the metabolome and transcriptome suggested that the anxiety-like behavior and spatial memory impairment induced by RIB and MAN may be attributed to the changes in four metabolites and 81 genes in the hippocampus, which is involved in amino acid metabolism and serotonin transport. In addition, combined with previous genome-wide association studies on depression, a correlation was found between the levels of Tnni3k and Tbx1 in the hippocampus and RIB induced depressive-like behavior. Finally, metabolite-gene network, qRT-PCR and western blot analysis showed that the insulin-POMC-MEK-TCF7L2 and MAPK-CREB-GRIN2A-CaMKII signaling pathways were respectively associated with RIB and MAN induced depressive/anxiety-like behavior and spatial memory impairment. Our findings clarified our understanding of the biological mechanisms underlying RIB and MAN induced depressive/anxiety-like behavior and spatial memory impairment in mice and highlighted the deleterious effects of high-dose RIB and MAN as long-term energy sources.
Collapse
Affiliation(s)
- Ke Xu
- grid.203458.80000 0000 8653 0555Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China ,grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Mingyang Wang
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Wei Zhou
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Juncai Pu
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Haiyang Wang
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China. .,NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China. .,Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China. .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
49
|
MIYAZAKI T, ABE H, UCHIDA H, TAKAHASHI T. Translational medicine of the glutamate AMPA receptor. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:1-21. [PMID: 33431723 PMCID: PMC7859086 DOI: 10.2183/pjab.97.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/30/2020] [Indexed: 05/05/2023]
Abstract
Psychiatric and neurological disorders severely hamper patient's quality of life. Despite their high unmet needs, the development of diagnostics and therapeutics has only made slow progress. This is due to limited evidence on the biological basis of these disorders in humans. Synapses are essential structural units of neurotransmission, and neuropsychiatric disorders are considered as "synapse diseases". Thus, a translational approach with synaptic physiology is crucial to tackle these disorders. Among a variety of synapses, excitatory glutamatergic synapses play central roles in neuronal functions. The glutamate α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) is a principal component of glutamatergic neurotransmission; therefore, it is considered to be a promising translational target. Here, we review the limitations of current diagnostics and therapeutics of neuropsychiatric disorders and advocate the urgent need for the promotion of translational medicine based on the synaptic physiology of AMPAR. Furthermore, we introduce our recent translational approach to these disorders by targeting at AMPARs.
Collapse
Affiliation(s)
- Tomoyuki MIYAZAKI
- Yokohama City University Graduate School of Medicine, Department of Physiology, Yokohama, Kanagawa, Japan
| | - Hiroki ABE
- Yokohama City University Graduate School of Medicine, Department of Physiology, Yokohama, Kanagawa, Japan
| | - Hiroyuki UCHIDA
- Keio University School of Medicine, Department of Neuropsychiatry, Tokyo, Japan
| | - Takuya TAKAHASHI
- Yokohama City University Graduate School of Medicine, Department of Physiology, Yokohama, Kanagawa, Japan
| |
Collapse
|
50
|
Kang KK, Kim YI, Seo MS, Sung SE, Choi JH, Lee S, Jung YS, Cho JY, Hwang DY, Park SJ, Kim KS. A comparative study of the phenotype with kainic acid-induced seizure in DBA/2 mice from three different sources. Lab Anim Res 2020; 36:39. [PMID: 33134158 PMCID: PMC7594308 DOI: 10.1186/s42826-020-00072-y] [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: 08/03/2020] [Accepted: 10/13/2020] [Indexed: 11/10/2022] Open
Abstract
The kainic acid-induced seizure mouse model is widely used in epilepsy research. In this study, we applied kainic acid to the subcutaneous injections of three different sources of DBA/2 mice to compare and evaluate the seizure response. The three mouse sources consisted of DBA/2Kor1 (Korea FDA source), DBA/2A (USA source), and DBA/2 (Japan source), and were purchased from different vendors. To compare the responses of DBA/2 mice to kainic acid injections, we examined the survival rate, seizure phenotype scoring, and behavioral changes. We also evaluated brain lesions using histopathological analysis. Following the administration of kainic acid, almost half of the cohort survived, and the seizure phenotype displayed a moderate level of sensitivity (2 ~ 4 out of 6). In the histopathologic analysis, there was no change in morphological features, and levels of glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba-1) increased in the kainic acid-treated groups. However, there was no difference in the neuronal nuclei (NeuN) expression level. All the data showed that the responses in the kainic acid-treated group were similar across the three strains. In conclusion, our results suggest that the three sources of DBA/2 mice (DBA/2Kor1, DBA/2A, and DBA/2B) have similar pathological responses to kainic acid-induced seizures.
Collapse
Affiliation(s)
- Kyung-Ku Kang
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | | | - Min-Soo Seo
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Soo-Eun Sung
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Joo-Hee Choi
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Sijoon Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan, 46241 Korea
| | - Joon Young Cho
- Exercise Biochemistry Laboratory, Korea National Sport University, 88-15 Oryung-dong, Songpa-gu, Seoul, 138-763 Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, 50463 Korea
| | - Sang-Joon Park
- Department of Histology, College of Veterinary Medicine, Kyungpook National University, Daegu, 702-701 Korea
| | - Kil Soo Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea.,College of Veterinary Medicine, Kyungpook National University, 80 Daehakro, Buk-gu, Daegu, 41566 Korea
| |
Collapse
|