1
|
Hwang HG, Park JW, Lee HJ, Ko MY, Ka M, Lee YK, Choi J, In SA, Lee YE, Lee S, Kim MS, Kim JY. Akkermansia muciniphila reverses neuronal atrophy in Negr1 knockout mice with depression-like phenotypes. Gut Microbes 2025; 17:2508424. [PMID: 40388597 PMCID: PMC12091914 DOI: 10.1080/19490976.2025.2508424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/07/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025] Open
Abstract
Genetic predispositions can shape the gut microbiome, which in turn modulates host gene expression and impacts host physiology. The complex interplay between host genetics and the gut microbiome likely contributes to the development of neuropsychiatric disorders, yet the mechanisms behind these interactions remain largely unexplored. In this study, we investigated the gut microbiota in Negr1 knockout (KO) mice, which exhibit anxiety- and depression-like behaviors, as NEGR1 (neuronal growth regulator 1) is a cell adhesion molecule linked to neuronal development and neuropsychiatric disorders. Our findings show significant early-life alterations in the gut microbiota composition of Negr1 KO mice, most notably a marked reduction in Akkermansia spp. along with reduced dendritic arborization and spine density in the nucleus accumbens (NAc) and the dentate gyrus (DG) of the hippocampus. Remarkably, daily administration of an Akkermansia strain isolated from wild-type mice reversed the neuronal structural abnormalities and ameliorated anxiety- and depression-like behaviors in Negr1 KO mice. Transcriptomic profiling revealed upregulation of mitochondrial genome-encoded genes in the NAc and hippocampus of Negr1 KO mice, along with a predisposition toward a pro-inflammatory state in the colon of Negr1 KO mice. The Akkermansia supplementation downregulated these mitochondrial genes in the NAc and hippocampus and upregulated genes involved in T cell activation and immune homeostasis in the colon. These findings demonstrate a novel gene-microbiome interaction in the pathophysiology of Negr1 KO mice, positioning Akkermansia spp. as a key mediator that improves neuronal atrophy and modulates anxiety- and depression-like behaviors. Our study provides compelling evidence for bidirectional interactions between host genetics and the gut microbiome in modulating neuropsychiatric phenotypes, offering new insights for addressing genetically influenced mental disorders.
Collapse
Affiliation(s)
- Hee-Gon Hwang
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ji-Woo Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyo-Jin Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Moon Yi Ko
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Minhan Ka
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Jaeyoon Choi
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Su-A In
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ye-Eun Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Soojin Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Min-Soo Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Yoon Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
2
|
Wang P, Song X, Qiu J, Zhu X, Wu P, Liao Z, Xie J, Wang N, Zhao H. A comparative study on the acute-phase behavioral and pathological responses of closed head injury induced by cranial vertex and temporal lobe impacts in male rats. Exp Neurol 2025; 389:115259. [PMID: 40246007 DOI: 10.1016/j.expneurol.2025.115259] [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: 02/05/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
The outcomes of traumatic brain injury (TBI) are closely linked to the strength of mechanical loads applied to the head. However, the same mechanical load can lead to significant variations in injury outcomes depending on the location of impact. To compare the acute-phase behavioral and pathological effects of different impact locations on TBI outcomes, we conducted a closed head injury experimental study using male rats subjected to cranial vertex and temporal lobe impacts. The rats were injured by an impactor according to the experimental protocol established using the L4 (23) orthogonal table, and the behavioral and pathological outcomes were assessed. The contribution rates of impact location and strength to TBI were quantified using Analysis of Variance. The results indicated that impact strength played a dominant role in TBI and showed a positive correlation, while the role of impact location in TBI cannot be ignored. Behaviorally, cranial vertex impacts led to more severe coma, motor, memory, and anxiety deficits. Pathologically, cranial vertex impacts caused more severe diffuse axonal injury in the corpus callosum and brainstem. In the left hippocampus and amygdala, cell loss due to cranial vertex impacts was more pronounced than that caused by temporal lobe impacts, whereas the opposite was true on the right side. Notably, the pathological changes observed in the left (non-impact) hippocampus and amygdala due to temporal lobe impacts showed a stronger linear correlation with behavioral outcomes, suggesting that damage to the left side has greater predictive power for behavioral deficits. This suggests that the impact location is an important factor affecting TBI and should be considered in the study.
Collapse
Affiliation(s)
- Peng Wang
- State Key Laboratory of Automotive Simulation and Control, College of Automotive Engineering, Jilin University, Changchun, Jilin, China
| | - Xuewei Song
- State Key Laboratory of Automotive Simulation and Control, College of Automotive Engineering, Jilin University, Changchun, Jilin, China.
| | - Jinlong Qiu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiyan Zhu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Pengfei Wu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhikang Liao
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Jingru Xie
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Wang
- Department of Radiology, The First Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Hui Zhao
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
3
|
Alves RL, Gonçalves A, Voytyuk I, Harrison DC. Behaviour profile characterization of PS19 and rTg4510 tauopathy mouse models: A systematic review and a meta-analysis. Exp Neurol 2025; 389:115234. [PMID: 40185359 DOI: 10.1016/j.expneurol.2025.115234] [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: 01/17/2025] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The rTg4510 and PS19 mouse models are widely used in tauopathy research. Alzheimer's disease (AD) is the most prevalent among tauopathies. Behavioural tests are frequently used to assess emotional, cognitive, and motor behaviours in mouse models of AD. Cognitive deficits begin to manifest in rTg4510 mice around 3 months of age and in PS19 mice around 6 months. However, it's widely recognized that behavioural outcomes can vary due to environmental factors, health status, and husbandry practices, causing phenotypic differences between facilities. This study aims to consolidate current knowledge of the behavioural phenotypes of these two mouse models. We conducted a comprehensive literature review using keyword searches with Boolean operators across databases up to January 2024. Additional studies were included from manual searches. A total of 23 articles were reviewed for rTg4510 mice and 52 for PS19 mice. We extracted methodological details and key findings from each study. Results for rTg4510 mice show consistent findings regarding locomotion, memory and learning, and neurological dysfunction. However, the limited studies on motor and balance behaviour revealed no significant differences, while anxiety-like behaviour showed some inconsistencies. PS19 mice demonstrate more robust results for anxiety-like behaviour, memory and learning, and locomotion, while findings for balance and coordination are more inconsistent. Although there is overall coherence in certain aspects of the behavioural profiles of these tauopathy mouse models, it is crucial to recognize experimental heterogeneity and profile behavioural baselines to optimize the testing of both genetic and pharmacological interventions.
Collapse
Affiliation(s)
- Renata L Alves
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom.
| | | | - Iryna Voytyuk
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom
| | - David C Harrison
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom
| |
Collapse
|
4
|
Maejima I, Sato K. New aspects of a small GTPase RAB35 in brain development and function. Neural Regen Res 2025; 20:1971-1980. [PMID: 39254551 PMCID: PMC11691468 DOI: 10.4103/nrr.nrr-d-23-01543] [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: 09/12/2023] [Revised: 12/12/2023] [Accepted: 12/30/2023] [Indexed: 09/11/2024] Open
Abstract
In eukaryotic cells, organelles in the secretory, lysosomal, and endocytic pathways actively exchange biological materials with each other through intracellular membrane trafficking, which is the process of transporting the cargo of proteins, lipids, and other molecules to appropriate compartments via transport vesicles or intermediates. These processes are strictly regulated by various small GTPases such as the RAS-like in rat brain (RAB) protein family, which is the largest subfamily of the RAS superfamily. Dysfunction of membrane trafficking affects tissue homeostasis and leads to a wide range of diseases, including neurological disorders and neurodegenerative diseases. Therefore, it is important to understand the physiological and pathological roles of RAB proteins in brain function. RAB35, a member of the RAB family, is an evolutionarily conserved protein in metazoans. A wide range of studies using cultured mammalian cells and model organisms have revealed that RAB35 mediates various processes such as cytokinesis, endocytic recycling, actin bundling, and cell migration. RAB35 is also involved in neurite outgrowth and turnover of synaptic vesicles. We generated brain-specific Rab35 knockout mice to study the physiological roles of RAB35 in brain development and function. These mice exhibited defects in anxiety-related behaviors and spatial memory. Strikingly, RAB35 is required for the precise positioning of pyramidal neurons during hippocampal development, and thereby for normal hippocampal lamination. In contrast, layer formation in the cerebral cortex occurred superficially, even in the absence of RAB35, suggesting a predominant role for RAB35 in hippocampal development rather than in cerebral cortex development. Recent studies have suggested an association between RAB35 and neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. In this review, we provide an overview of the current understanding of subcellular functions of RAB35. We also provide insights into the physiological role of RAB35 in mammalian brain development and function, and discuss the involvement of RAB35 dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ikuko Maejima
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Ken Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|
5
|
Marathe CK, Thorat VG, Kokate IP, Pawar AT, Bhatt S. Antidepressant- and anxiolytic-like effects of linezolid in streptozotocin-induced diabetic mice via modulation of brain serotonin and plasma corticosterone activity. Biochem Biophys Res Commun 2025; 765:151843. [PMID: 40273625 DOI: 10.1016/j.bbrc.2025.151843] [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: 12/17/2024] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Depression and anxiety are comorbid with diabetes. Diabetes and depression have a bidirectional relationship. In this study, Linezolid was evaluated for the treatment of depression and anxiety comorbid with T2DM. Linezolid may exhibit antidepressant and anxiolytic-like effects via modulation of MAO enzyme activity. In the study, male Swiss Albino mice were divided into 9 groups (n = 70). Group 1 (normal control); Group 2 diabetes control (DC); Group 3, 4, and 5 (low, intermediate, high dose of Linezolid; LDL, IDL and HDL); Group 6 [DC + Paroxetine (PAR)], Group 7 [DC + Metformin (MET)]; Group 8 (DC + LDL + PAR); Group 9 (DC + LDL + PAR + MET). Except for the normal control group, all the other groups were induced with T2DM with Streptozotocin (50 mg/kg, i.p.) and Nicotinamide (120 mg/kg, i.p.). Mice showed glucose levels of more than 160 mg/dl were taken in the study. Following a 14-day treatment, behavioral and biochemical assessments were done using the FST, TST, EPM, and OFT, and blood glucose, plasma corticosterone, brain TNF-α and serotonin were measured. Group 3 to 9 showed their activity in behavioral models of depression and anxiety such as FST, TST, OFT, and EPM. The treatment groups (Groups 3 to 9) also have shown a significant (p < 0.05) decrease in blood glucose, plasma corticosterone, and brain TNF-α levels with respect to DC, while serotonin levels are increased in Groups 5, 8 & 9. The above findings suggest the beneficial effects of Linezolid in the treatment of depression and anxiety comorbid with T2DM.
Collapse
Affiliation(s)
- Chandan K Marathe
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, Maharashtra, India
| | - Vaishnavi G Thorat
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, Maharashtra, India
| | - Ishwar Purushottam Kokate
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, Maharashtra, India
| | - Anil T Pawar
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, Maharashtra, India
| | - Shvetank Bhatt
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, 411038, Maharashtra, India.
| |
Collapse
|
6
|
Rehn S, Raymond JS, Boakes RA, Kendig MD, Leenaars CHC. Behavioural and physiological effects of binge eating: A systematic review and meta-analysis of animal models. Neurosci Biobehav Rev 2025; 173:106135. [PMID: 40222574 DOI: 10.1016/j.neubiorev.2025.106135] [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: 08/27/2024] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Binge eating is defined as eating abnormally large amounts in a brief period of time. Many animal studies have examined the behavioural and physiological effects of binge eating of high-fat, high-sugar foods to model the consequences of human binge eating. The present systematic review of 199 rodent studies sought to identify the behavioural and physiological consequences of binge eating and determine whether changes were specific to binge eating or to general effects of exposure to a palatable diet. A meta-analysis of 18 rodent studies revealed that binge eating produces greater anxiety-like behaviour on the Elevated Plus-Maze with a small effect size and significant funnel plot asymmetry, suggesting that the true effect size is overestimated. A history of binge-like access generally increases progressive ratio breakpoint for the binged food, without altering 'liking' as measured by lick microstructure, suggesting that dissociable effects on 'wanting' but not 'liking' accompany binge eating behaviour and contribute to its persistence. Binge eating appears to enhance compulsive food-seeking behaviour and prevent stress-induced reductions in intake but does not appear to alter depression-like behaviour or locomotor activity. Notably, binge eating may produce comparable metabolic impairments to those observed after extended continuous exposure to a palatable diet despite no overall effects on body weight outcomes in most studies.
Collapse
Affiliation(s)
- Simone Rehn
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia; School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Joel S Raymond
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia; Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, Sydney, NSW 2050, Australia; Department of Psychiatry and Brain Health Institute, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Robert A Boakes
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael D Kendig
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| | - Cathalijn H C Leenaars
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
7
|
Mirehei M, Motamedi F, Maghsoudi N, Mansouri Z, Naderi S, Khodagholi F, Abbaszadeh F. Effects of Bufexamac, a class IIb HDAC inhibitor, on behavior and neuropathological features in an Aβ-induced rat model of Alzheimer's disease. Exp Gerontol 2025; 204:112746. [PMID: 40185252 DOI: 10.1016/j.exger.2025.112746] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
It has been suggested that Alzheimer's disease (AD), a progressive neurological condition, can potentially be treated through epigenetic means by targeting histone deacetylases (HDACs), enzymes that regulate gene expression. In this study, we investigated the molecular mechanisms of Bufexamac, in an animal model of AD. Bufexamac specifically targets Class IIb HDACs, which are particularly relevant in the context of neuroinflammation and neurodegeneration. This selectivity may reduce off-target effects commonly associated with broader-spectrum HDAC inhibitors, such as pan-HDAC inhibitors, which can affect multiple HDAC classes and potentially lead to undesirable side effects. Male rats injected with Aβ25-35 for AD-like symptoms were treated with 20 μg/rat Bufexamac for 8 days. Cognitive function, depression, and anxiety were assessed through behavioral tests, while Western blotting, H&E staining, and ELISA were used to detect protein expression, morphological changes, and enzyme activity. Bufexamac treatment markedly improved cognitive and behavioral impairments in Aβ-injected rats and regulated the key proteins related to neuroinflammation (GFAP, Iba1), histone, and α-tubulin acetylation. Simultaneously, it decreased the expression of proteins in the stromal interaction molecule (STIM) pathway. Furthermore, Bufexamac lowered the activity of monoamine oxidase enzymes, elevated the count of healthy neurons, and ameliorated neuronal structure in the hippocampus. Overall, these findings suggest that Bufexamac could be a more targeted therapy for AD than other non-selective HDAC inhibitors, which often have diverse functions and potential side effects. Bufexamac enhances cognitive function and alleviates depression and anxiety by regulating proteins related to neuroinflammation, histone, and α-tubulin acetylation, as well as modulating STIM levels and MAO activity.
Collapse
Affiliation(s)
- Monireh Mirehei
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Maghsoudi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Mansouri
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudabeh Naderi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Abbaszadeh
- Neurobiology Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Pathak D, Singh KP. Neurobehavioral manifestations in female rats after intermittent exposure to an anticancer agent, paclitaxel. Behav Pharmacol 2025:00008877-990000000-00135. [PMID: 40397101 DOI: 10.1097/fbp.0000000000000833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Paclitaxel (PTX), a widely used chemotherapeutic agent, causes both peripheral and central neurotoxicity, leading to significant behavioral impairments. However, inadequate literature is available on PTX-induced neurobehavioral sequelae associated with anxiety, depression, and cognition in adults during and after chemotherapy. Therefore, the present study aimed to investigate neurobehavioral impairments in adult female rats following PTX exposure, with a specific focus on anxiety-like behaviors and cognitive functions such as learning and memory. In this study, we used adult female Wistar rats aged 10-12 weeks (average weight: 180 ± 5 g) and administered clinically relevant therapeutic doses of PTX (1.6 and 3.2 mg/kg body weight) intravenously once weekly for 6 weeks, simulating the clinical chemotherapy regimen. Neurobehavioral assessments were conducted after the first and sixth doses of PTX using validated mazes, including the photoactometer, open-field maze, elevated plus-maze (EPM; for anxiety-like behaviors), and the step-down latency test (SDL; for cognitive performance). Neurobehavioral patterns were recorded using autotracking software (ANY-maze, Stoelting Co., Wood Dale, Illinois, USA). Our findings revealed substantially reduced locomotor activity in the photoactometer, increased anxiety-like behaviors with amplified fear emotionality in the open-field and EPM tests, and memory impairment in the SDL test. These results suggest that the manifestation of anxiogenic and cognitive behavioral changes is associated with the administration of a higher dose (3.2 mg/kg) of PTX. In conclusion, our study indicates that PTX causes significant neurobehavioral impairments in rats after exposure to equivalent therapeutic doses of PTX.
Collapse
Affiliation(s)
- Deepika Pathak
- Neurobiology Laboratory, Department of Zoology, University of Allahabad, Prayagraj, India
| | | |
Collapse
|
9
|
Lord MN, Spaulding MO, Hoffman JR, Basma RK, Noble EE. Edible cannabinoids impact meal structure and food impulsivity in female rats. iScience 2025; 28:112415. [PMID: 40330886 PMCID: PMC12051634 DOI: 10.1016/j.isci.2025.112415] [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: 12/02/2024] [Revised: 02/04/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Cannabinoid receptor agonists increase eating in a dose-dependent manner. However, the behavioral mechanisms by which cannabinoids modulate food intake control aren't clear, particularly in females. We utilized a rodent model of cannabinoid administration modeling a common route of cannabinoid consumption in humans: edibles. Herein, we administered the dual cannabinoid receptor agonist CP55940 in edible form to female rats and observed acute increases in standard chow intake due to an increase in meal size with no change in meal number. We further observed that the hyperphagic dose of edible CP55940 increases impulsive responding for sucrose, but this did not coincide with changes in motivation for sucrose. Finally, cannabinoids can affect anxiety-like behavior, but the acutely hyperphagic dose used in our studies had no effect on anxiety-like behavior. We conclude that edible cannabinoid administration delays satiation and increases impulsive eating behavior without impacting food motivation, potentially by reducing inhibitory control.
Collapse
Affiliation(s)
- Magen N. Lord
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30606, USA
| | - Mai O. Spaulding
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30606, USA
| | - Jessica R. Hoffman
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30606, USA
| | - Rawad K. Basma
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30606, USA
| | - Emily E. Noble
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30606, USA
| |
Collapse
|
10
|
Harris SA, Sgro M, Salberg S, Li C, Vlassopoulos E, Smith M, Semple BD, Chinnery HR, Mychasiuk R. Shaking into deficits: investigating behavioural and neuropathological outcomes associated with a novel preclinical model of infant abusive head trauma. Acta Neuropathol Commun 2025; 13:100. [PMID: 40375338 DOI: 10.1186/s40478-025-02029-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
Abusive head trauma (AHT) resulting from violent shaking and whiplash-induced brain injury by a caregiver, is the leading cause of abusive mortality and morbidity in children. Cerebral oedema is common in survivors of AHT. While many children may initially appear behaviourally asymptomatic or present with non-specific symptoms following the AHT, deficits often emerge later in childhood. Additionally, AHTs are frequently repetitive, with a single child likely to experience multiple AHTs. Despite the prevalence of AHT, the mechanisms that lead to brain pathology and the latent emergence of behavioural deficits are poorly understood, and there is a paucity of preclinical, small animal models to investigate the biology and cumulative effects of repetitive injuries. This study aimed to develop a preclinical model of repetitive AHT and subsequently examine alterations in gene expression, cell types, and early adolescent behaviour. Mice were placed on a 400 rpm shaking device for 60s. This was repeated one, three, or five times throughout the neonatal development period (postnatal days (P)8-12). Injured mice initially displayed no overt behavioural changes compared to uninjured controls; however, in adolescence (P40-45) they later developed deficits in socialisation and thermal nociception. Further, alterations in the expression of genes involved in growth, cell damage, and development were observed in the brains of injured mice, along with an increase in white matter cells and evidence of blood-brain barrier leakage. This novel preclinical model of AHT provides a valuable platform for exploring diagnostic biomarkers and potential therapeutic interventions for children with an AHT.
Collapse
Affiliation(s)
- Sydney A Harris
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Marissa Sgro
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Sabrina Salberg
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Crystal Li
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Elaina Vlassopoulos
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Madeleine Smith
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Bridgette D Semple
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Science, University of Western Australia, Lions Eye Institute, Perth, Western Australia, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia.
- Department of Neuroscience, School of Translational Medicine, Monash University, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
11
|
Ni WH, Wang K, Wang Y, Lu J, Lu CT, Rong W, Gu YF, Qian WJ, Zhang HL. Long-term folic acid treatment relieves chronic inflammatory pain and pain-induced anxiety by reducing MMP2 expression in rats. Neuropharmacology 2025; 269:110352. [PMID: 39938859 DOI: 10.1016/j.neuropharm.2025.110352] [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: 12/09/2024] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
Chronic inflammatory pain is a top priority for arthritis patients seeking medical care. Despite the availability of NSAIDs and glucocorticoids, pain management becomes increasingly challenging due to central and peripheral sensitization. Previous studies have shown that Matrix metalloproteinase 2 (MMP2) promotes neuroinflammation by cleaving extracellular matrix proteins and activating pro-inflammatory cytokines. Folic acid acts as a promising candidate for the treatment of neuroinflammatory diseases due to its neuroprotective effects. However, the role of folic acid in inflammatory pain remains unclear. This study investigated the analgesic mechanisms of folic acid in inflammatory pain. Adult rats underwent inflammatory pain by injecting complete freund's adjuvant (CFA) into the right hindpaw. Behavioral tests were used to assess the paw withdrawal threshold (PWT) and paw withdrawal latency (PWL). The results demonstrated that CFA injection induced abnormal mechanical and thermal pain and increased MMP2 expression in L3-L5 DRG and SDH of CFA rats. MMP2 was mainly expressed in neurons rather than glial cells in L3-L5 DRG of CFA rats. We further discovered that MMP2 inhibitor auraptene or knockdown alleviated inflammatory pain in CFA rats. Interestingly, we observed that long-term folic acid treatment reversed MMP2 overexpression, resulting in sustained relief of chronic inflammatory pain. Consistently, long-term folic acid treatment also relieved pain-induced anxiety. These results indicated that folic acid had a protective role in chronic inflammatory pain and pain-induced anxiety by repressing MMP2 expression. Folic acid or auraptene might be promising therapeutic options for the treatment of chronic inflammatory pain.
Collapse
Affiliation(s)
- Wen-Hui Ni
- Center for Translational Medicine, Department of Renal Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, 215600, China
| | - Ke Wang
- Department of Pain, Suzhou Wuzhong People's Hospital, Suzhou, 215128, China
| | - Yun Wang
- Center for Translational Medicine, Department of Renal Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, 215600, China
| | - Jia Lu
- Center for Translational Medicine, Department of Renal Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, 215600, China
| | - Chun-Ting Lu
- Department of Pain, Suzhou Wuzhong People's Hospital, Suzhou, 215128, China
| | - Wen Rong
- Department of Pain, Suzhou Wuzhong People's Hospital, Suzhou, 215128, China
| | - Yi-Feng Gu
- Center for Translational Medicine, Department of Renal Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, 215600, China
| | - Wen-Juan Qian
- Center for Translational Medicine, Department of Renal Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, 215600, China.
| | - Hai-Long Zhang
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
12
|
Lysaker CR, Kugler BA, Csikos V, Birky CJ, Gilmore CA, Wenger M, Franczak E, Davis X, Hauger BM, Allen JA, Troutwine BR, Gonalez-Duran L, McCoin CS, Chauhan M, Harris JL, Frazier AL, Winter MK, Koch LG, Britton SL, Thyfault JP, Wilkins HM. A polygenetic rat model of divergent aerobic capacity reveals a liver-brain interaction impacting Alzheimer's disease-like phenotypes. J Physiol 2025. [PMID: 40349321 DOI: 10.1113/jp286750] [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: 11/09/2024] [Accepted: 04/16/2025] [Indexed: 05/14/2025] Open
Abstract
The interaction between liver and brain health is an emerging complex relationship implicated in Alzheimer's disease (AD). Divergence in aerobic capacity influences liver and brain health independently; however, whether these factors converge to influence AD risk is mechanistically unknown. Bile acid metabolism has been implicated as a link between liver and brain health and is modulated by aerobic capacity. Here, we examined rats selectively bred for high vs. low intrinsic aerobic capacity [high and low-capacity runner (HCR or LCR)] on indices of hepatic metabolism and brain health following a chronic low-fat, high-fat, or high-fat diet with bile acid sequestrant from 6 to 12 months of age. Pre- and post-diet measures included learning, memory, and brain volume metabolite levels. We additionally quantified brain and liver Aβ and proteins associated with Aβ production and clearance, as well as liver and brain mitochondrial energetics and liver bile acid species. We found that not only did aerobic capacity and diet influence mitochondrial function, but also it modified Aβ levels across the liver and brain. Additionally, aerobic capacity and diet altered bile acid profiles and brain hippocampal metabolite levels. The addition of bile acid sequestrant lowered brain Aβ levels in a sexually dimorphic manner. Aerobic capacity but not diet altered cognitive outcomes. Our results indicate that aerobic capacity and diet-induced liver health alterations modulate brain health with respect to metabolism and AD-like pathologies, whereas a stimulation of faecal bile acid loss could have positive effects on lowering brain Aβ. KEY POINTS: Aerobic capacity and diet-induced alterations to liver function alter liver bile acid species and faecal energy loss. Aerobic capacity and diet alter both brain and liver Aβ homeostasis. Aerobic capacity modulates brain and hippocampal volume in addition to brain metabolism. Aerobic capacity influences learning in middle-aged rats.
Collapse
Affiliation(s)
- Colton R Lysaker
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Benjamin A Kugler
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Vivien Csikos
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Cole J Birky
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Caleb A Gilmore
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Madi Wenger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas Center for Metabolism and Obesity Research, University of Kansas Medical Center, Kansas City, KS, USA
| | - Edziu Franczak
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas Center for Metabolism and Obesity Research, University of Kansas Medical Center, Kansas City, KS, USA
- Diabetes Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Xin Davis
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas Center for Metabolism and Obesity Research, University of Kansas Medical Center, Kansas City, KS, USA
- Diabetes Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brittany M Hauger
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Julie A Allen
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Benjamin R Troutwine
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | | | - Colin S McCoin
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas Center for Metabolism and Obesity Research, University of Kansas Medical Center, Kansas City, KS, USA
| | - Munish Chauhan
- Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Janna L Harris
- Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alexandria L Frazier
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michelle K Winter
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, USA
| | - Steven L Britton
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - John P Thyfault
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas Center for Metabolism and Obesity Research, University of Kansas Medical Center, Kansas City, KS, USA
- Diabetes Institute, University of Kansas Medical Center, Kansas City, KS, USA
- Research Service, Kansas City VA Medical Center Department of Veterans Affairs, Kansas City, MO, USA
| | - Heather M Wilkins
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
13
|
Syed OA, Petranker R, Tsang B. The effect of psychedelic microdosing on animal behavior: A review with recommendations for the field. Neurosci Biobehav Rev 2025; 174:106204. [PMID: 40348309 DOI: 10.1016/j.neubiorev.2025.106204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Microdosing, the repeated use of psychedelic substances at low doses, is growing in popularity among recreational consumers. While this practice is associated with many benefits to mood, well-being and health, research in this area is in its early stages and predominantly centered on human applications. In this narrative review, we synthesize the findings from studies investigating the effects of microdosing on the behaviors of three animal species: rats, mice, and zebrafish. A total of 12 studies were identified that implemented a microdosing regimen of LSD, psilocybin, or DMT in these animal models. Overall, microdosing caused little changes in behaviors associated with anxiety- and depressive-like states. Moreover, while microdosing was well-tolerated across species, further research is needed to capture specific safety concerns. Finally, we critically appraise the studies included in this review based on their methodologies and discuss further avenues of research to advance the preclinical literature on psychedelic microdosing. Specifically, we recommend that future research prioritize the replication of existing findings to inform the development of robust study designs and dosing protocols, as well as establish standardized methodologies to enable effective comparisons across different animal models. Furthermore, future investigations should explore the therapeutic potential of mescaline microdosing, examine sex-dependent effects, and extend research to additional models of psychiatric conditions, including those related to obsessive-compulsive disorder and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Omer A Syed
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Rotem Petranker
- Department of Psychology, Neuroscience, and Behaviour, McMaster University, Hamilton, ON, Canada
| | - Benjamin Tsang
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Chen CH, Yu KC, Hsu LJ, Chiu WT, Hsu KS. Pro-inflammatory macrophages contribute to developing comorbid anxiety-like behaviors through gastrointestinal vagal afferent signaling in experimental colitis mice. Brain Behav Immun 2025; 128:620-633. [PMID: 40348137 DOI: 10.1016/j.bbi.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 04/09/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Anxiety symptoms are commonly observed in individuals with inflammatory bowel disease (IBD), but the mechanistic link between IBD and comorbid anxiety remains incompletely understood. Our previous study revealed that vagal gut-brain signaling contributes to driving comorbid anxiety-like behaviors in dextran sulfate sodium (DSS)-induced colitis mice, but how vagus nerve senses and transmits information to the brain in response to changes in the colonic microenvironment following DSS treatment remain elusive. Here, we identify a critical contribution of pro-inflammatory CD86+ macrophages to activate gut-innervating vagal afferents and ultimately drive anxiety-like behaviors in DSS-treated mice. An increased number of F4/80+ macrophages accumulated closely with gut-innervating vagal afferent fibers following DSS treatment. Depletion of macrophages alleviated DSS-induced anxiety-like behaviors, whereas peripheral delivery of lipopolysaccharide-activated M1 macrophages promoted anxiety-like behaviors, which were prevented by bilateral vagal afferent ablation. Moreover, differential expression levels of anxiety-like behaviors were positively correlated with neuronal activity changes in the nucleus tractus solitarius, locus coeruleus, and basolateral amygdala. Finally, treatment with either anti-α4β7 integrin antagonist vedolizumab or neutralizing anti-interleukin-1β monoclonal antibody effectively alleviated DSS-induced anxiety-like behaviors. Collectively, these findings unravel a mechanism of macrophage-to-vagus nerve communication via cytokine signaling responsible for comorbid anxiety associated with experimental colitis and suggest that pro-inflammatory CD86+ macrophages may represent a potential therapeutic target for psychological comorbidities in patients with IBD.
Collapse
Affiliation(s)
- Chin-Hao Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kuan-Chieh Yu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Li-Jin Hsu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
15
|
da Silva Joaquim L, da Rosa LR, Strickert Y, Machado RS, Lanzzarin E, Bernardes G, de Souza Ramos S, de Novais LR, Steiner B, Farias B, Mathias K, Martins HM, Lins EMF, Chaves JS, Camilo D, da Silva LE, de Oliveira MP, da Silva MR, Barcelos PMP, Santos FP, Bobinski F, Rezin GT, Yonamine M, Inserra A, Petronilho F, de Bitencourt RM. Ayahuasca reverses ischemic stroke-induced neuroinflammation and oxidative stress. Behav Brain Res 2025; 485:115521. [PMID: 40043852 DOI: 10.1016/j.bbr.2025.115521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/21/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Ischemic stroke is a leading cause of death and disability worldwide. Survivors face disability and psychiatric sequelae resulting from ischemia-induced cell death and associated neuroinflammation, and oxidative stress. Herbal medicines have been shown to elicit neuroprotective effects following stroke due to their anti-inflammatory and antioxidant effects. Preliminary evidence suggests that Ayahuasca (AYA), a decoction made from the vine Banisteriopsis caapi containing β-carbolines and the shrub Psychotria viridis containing N, N-Dimethyltryptamine, might attenuate ischemia-induced neuroinflammation and oxidative stress. Therefore, in this study we investigated the putative protective effects of AYA in the middle cerebral artery occlusion (MCAO) model of ischemic stroke. METHODS Wistar rats were subjected to the MCAO stroke model or sham surgery on day 0. After 24-h, rats were treated for three days with AYA (2 and 4 mL/kg, gavage) or saline. Neurological score was assessed for 72-h post-stroke. Rats were tested in the elevated plus maze, open field, and novel object recognition tests to assess locomotion, anxiety-like behavior, and recognition memory. Interleukin (IL)-6, IL-10 myeloperoxidase (MPO) activity, and the nitrite/nitrate (N/N) concentrations were determined in the prefrontal cortex (PFC), hippocampus (HPC), hypothalamus (HYP) and cortex. as markers of inflammation. Oxidative stress was quantified in the same brain areas as measured by the levels of thiobarbituric acid reactive species (TBARS), protein carbonylation, and superoxide dismutase (SOD), and catalase (CAT) activity. Mitochondrial metabolism was assessed quantifying the activity of complex 1(CI), CII, citrate synthase (CS), succinate dehydrogenase (SDH), and creatine kinase (CK). RESULTS No differences were observed regarding neurological deficits, locomotion, anxiety-like behavior, and recognition memory. However, AYA reversed the stroke-induced increase in IL-6 levels in the PFC and the HPC, IL-10 in the PFC, HPC, and HYP, MPO activity in the PFC, and N/N concentration and CAT activity in the HYP. Moreover, AYA decreased TBARS levels in the PFC and HPC and brain-derived neurotrophic factor (BDNF) in the PFC, and increased SOD activity in the cortex. Lastly, AYA increased CI activity in the HPC and cortex and decreased SDH and CK activity in the HPC. CONCLUSION AYA administration following ischemic stroke modulates oxidative stress and neuroinflammation in the PFC, HPC, and HYP. Despite no significant improvements in neurological or behavioral scores, these molecular changes suggest a neuroprotective role of AYA. Future studies should explore the timing of AYA treatment and putative long-term effects on functional recovery, as well as its potential in other brain regions critical for cognitive and motor functions.
Collapse
Affiliation(s)
- Larissa da Silva Joaquim
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Lara Rodrigues da Rosa
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Yasmin Strickert
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Richard Simon Machado
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil; Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Everton Lanzzarin
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Gabriela Bernardes
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Suelen de Souza Ramos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Linério Ribeiro de Novais
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Beatriz Steiner
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Brenno Farias
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Helena Mafra Martins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Elisa Mitkus Flores Lins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Jéssica Schaefer Chaves
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Douglas Camilo
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Pablo Michel Pereira Barcelos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabiana Pereira Santos
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Maurício Yonamine
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio Inserra
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Rafael Mariano de Bitencourt
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil.
| |
Collapse
|
16
|
Poliński P, Miret Cuesta M, Zamora-Moratalla A, Mantica F, Cantero-Recasens G, Viana C, Sabariego-Navarro M, Normanno D, Iñiguez LP, Morenilla-Palao C, Ordoño P, Bonnal S, Ellis JD, Gómez-Riera R, Fanlo-Ucar H, Yap DS, Martínez De Lagrán M, Fernández-Blanco Á, Rodríguez-Marin C, Permanyer J, Fölsz O, Dominguez-Sala E, Sierra C, Legutko D, Wojnacki J, Musoles Lleo JL, Cosma MP, Muñoz FJ, Blencowe BJ, Herrera E, Dierssen M, Irimia M. A highly conserved neuronal microexon in DAAM1 controls actin dynamics, RHOA/ROCK signaling, and memory formation. Nat Commun 2025; 16:4210. [PMID: 40328765 PMCID: PMC12056172 DOI: 10.1038/s41467-025-59430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Actin cytoskeleton dynamics is essential for proper nervous system development and function. A conserved set of neuronal-specific microexons influences multiple aspects of neurobiology; however, their roles in regulating the actin cytoskeleton are unknown. Here, we study a microexon in DAAM1, a formin-homology-2 (FH2) domain protein involved in actin reorganization. Microexon inclusion extends the linker region of the DAAM1 FH2 domain, altering actin polymerization. Genomic deletion of the microexon leads to neuritogenesis defects and increased calcium influx in differentiated neurons. Mice with this deletion exhibit postsynaptic defects, fewer immature dendritic spines, impaired long-term potentiation, and deficits in memory formation. These phenotypes are associated with increased RHOA/ROCK signaling, which regulates actin-cytoskeleton dynamics, and are partially rescued by treatment with a ROCK inhibitor. This study highlights the role of a conserved neuronal microexon in regulating actin dynamics and cognitive functioning.
Collapse
Affiliation(s)
- Patryk Poliński
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Marta Miret Cuesta
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Federica Mantica
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Gerard Cantero-Recasens
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Carlotta Viana
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Davide Normanno
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- Institute of Human Genetics, Univ Montpellier, CNRS, Montpellier, France
| | - Luis P Iñiguez
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | | | - Sophie Bonnal
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Raúl Gómez-Riera
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Dominic S Yap
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Álvaro Fernández-Blanco
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Jon Permanyer
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Orsolya Fölsz
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Eduardo Dominguez-Sala
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- TecnoCampus, Universitat Pompeu Fabra, Department of Health Sciences, Mataró, Spain
| | - Cesar Sierra
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Diana Legutko
- Nencki Institute of Experimental Biology, BRAINCITY, Warsaw, Poland
| | - José Wojnacki
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Juan Luis Musoles Lleo
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria Pia Cosma
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | | | | | - Mara Dierssen
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
- Universitat Pompeu Fabra, Barcelona, Spain.
- Biomedical Research Networking Center for Rare Diseases (CIBERER), Barcelona, Spain.
| | - Manuel Irimia
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
- Universitat Pompeu Fabra, Barcelona, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
17
|
Zuo X, Bai H, Qiu J, Li R, Kuang X, Zhao Y, Tuo J, Zhao Q, Zhao X, Feng X. 17β-Trenbolone modulates anxiety-related synaptic plasticity by affecting the interaction between hippocampal TACR3 and systemic testosterone in male mice. ENVIRONMENTAL RESEARCH 2025; 279:121796. [PMID: 40340005 DOI: 10.1016/j.envres.2025.121796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Anxiety Disorder is a common neurological disorder for which ubiquitous environmental endocrine disruptors may be risk factors. 17β-trenbolone (17-TB) has been recognized as a potential environmental endocrine disruptor and its neurotoxic effects have attracted attention. Tachykinin receptor 3 (TACR3), a G protein-coupled receptor involved in pubertal anxiety, and its underlying neural mechanisms remain enigmatic. Therefore, this study investigated the possible relationship between 17-TB and TACR3, testosterone (T), and synaptic plasticity. Our results showed that adolescent male Balb/c mice developed significant anxiety-like behavior after four weeks of exposure to environmentally relevant concentrations of 17-TB (100 μg/kg/day). Transcriptomic RNA-Seq results showed that 17-TB affected abnormal synaptic transmission signals in the hippocampus. Electrophysiological results showed that 17-TB reduced the activity of hippocampal dentate gyrus (DG) neurons and led to the downregulation of hippocampal TACR3 and T levels. In addition, Western blot, immunohistochemistry, ELISA, and qRT-PCR showed that 17-TB exposure led to the downregulation of key hippocampal synaptic proteins PSD95, Gephyrin, and Syn, and induced a metabolic imbalance in glutamate (Glu)/GABA signaling, affecting dopamine signaling. Interestingly, testosterone supplementation (10 mg/kg/day) was effective in ameliorating the above phenomena and alleviating anxiety-like behaviors. These results suggest that 17-TB modulates anxiety-related synaptic plasticity by regulating hippocampal TACR3 and T interactions in vivo. In conclusion, our study contributes to understanding the neurobehavioral and potential mechanistic effects of environmental 17-TB exposure in mammals. It alerts the public to the health risks of chemicals to mammals and suggests new research directions and potential therapeutic targets.
Collapse
Affiliation(s)
- Xiang Zuo
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Huijuan Bai
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Jinyu Qiu
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Ruimin Li
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Xiaochen Kuang
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Yudi Zhao
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Jingyi Tuo
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qili Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Xin Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China.
| | - Xizeng Feng
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
18
|
Kruse MS, Coirini H, Rey M. Early Consumption of Stevia rebaudiana Bertoni on Rat Females: Actions on Their Fertility, Progeny, and Behavior. J Nutr 2025:S0022-3166(25)00268-8. [PMID: 40324524 DOI: 10.1016/j.tjnut.2025.04.023] [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: 10/03/2024] [Revised: 03/27/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND The growing interest in healthy diets has led to increased consumption of natural products such as stevia. Pregnant females, who often consume high amounts of non-caloric sweeteners, are particularly vulnerable. Given the limited health guidelines on stevia, evaluating its effects on this population and their offspring is essential. OBJECTIVES This study assessed the impact of early, continuous exposure to an aqueous extract of Stevia rebaudiana Bertoni (Asteraceae) on female rat reproductive health, anxiety, sexual behavior, and progeny outcomes. METHODS Female Sprague-Dawley rats (PND21) were assigned to a control group (CON, water) or a stevia group (STE). In experiment 1, estrous cycles were monitored via vaginal smears (CON, n = 6/assay; STE, n = 14/assay). Anxiety-like behavior was assessed using the elevated plus maze test, and sexual behavior was evaluated through the partner preference test (CON, n = 4/assay; STE, n = 4/assay). Pregnancy rate, duration, litter size, sex ratio, and offspring survival were also assessed. In experiment 2, stevia administration began 5, 10, 15, or 50 d before mating, and the same reproductive parameters were evaluated. Statistical analyses were performed using a Student's t-test, repeated measures analysis of variance (RM-ANOVA), and one-way analysis of variance (ANOVA) with Fisher's Protected Least Significant Difference post-hoc test. RESULTS The STE group exhibited disrupted estrous cycles, reduced pregnancy rates, and smaller litters (with notably fewer males). Body weight was significantly lower in the STE group during the third week of pregnancy. There were no significant differences in food or beverage intake. Behaviorally, the STE group showed decreased sexual partner preference, lower anxiety, and increased risk assessment behavior. Prolonged premating exposure to stevia was associated with extended pregnancy duration, smaller litters, and higher pup weights at PND1. CONCLUSIONS Stevia disrupted fertility, reducing pregnancy rates, litter size, and male offspring, while extending gestation. Prolonged exposure worsened these effects, highlighting stevia's impact on reproduction and prenatal development. These findings underscore the need for improved health guidelines for vulnerable populations.
Collapse
Affiliation(s)
- María Sol Kruse
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental-CONICET, Ciudad Autónoma de Buenos Aires C1428ADN, Argentina
| | - Héctor Coirini
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental-CONICET, Ciudad Autónoma de Buenos Aires C1428ADN, Argentina
| | - Mariana Rey
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental-CONICET, Ciudad Autónoma de Buenos Aires C1428ADN, Argentina.
| |
Collapse
|
19
|
Bouteldja A, Marceau L, Srivastava L, Cermakian N. Early-Life Ventral Hippocampal Lesion and Circadian Disruption Result in Altered Behavior in Adult Mice in a Sex-Dependent Manner. Eur J Neurosci 2025; 61:e70134. [PMID: 40356274 PMCID: PMC12069966 DOI: 10.1111/ejn.70134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/23/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025]
Abstract
Schizophrenia is believed to arise because of the interaction of early abnormal neurodevelopment with environmental insults during key developmental stages later in life. Furthermore, disrupted circadian rhythms are reported in patients, and circadian disruption is associated with increased symptom severity, hinting at its role as a risk factor. Using the neonatal ventral hippocampal lesion mouse model, we aimed to assess the interaction between disrupted ventral hippocampal development with circadian disruption during adolescence in affecting behavior in male and female C57BL/6N mice. After conducting a series of behavioral tests, we found that the neonatal ventral hippocampal lesion and chronic jet lag during adolescence synergistically led to increased anxiety-like behavior in males. In females, the lesion prevented increased social preference caused by chronic jet lag and led to increased anxiety-like behavior. Mice were then moved to running wheel cages to measure their locomotor activity rhythms. We found that the lesioned male mice exposed to chronic jet lag exhibited fragmented rhythms under constant darkness. Moreover, lesioned male and female mice, especially those exposed to chronic jet lag, had reduced activity counts under constant light. These findings highlight that the interaction of abnormal neurodevelopment in areas relevant to schizophrenia with circadian disruption during adolescence results in lasting behavioral changes in a sex-dependent manner in mice.
Collapse
Affiliation(s)
- Ahmed A. Bouteldja
- Douglas Mental Health University InstituteMontréalQuébecCanada
- Integrated Program in NeuroscienceMcGill UniversityMontréalQuébecCanada
| | | | - Lalit K. Srivastava
- Douglas Mental Health University InstituteMontréalQuébecCanada
- Department of PsychiatryMcGill UniversityMontréalQuébecCanada
| | - Nicolas Cermakian
- Douglas Mental Health University InstituteMontréalQuébecCanada
- Department of PsychiatryMcGill UniversityMontréalQuébecCanada
| |
Collapse
|
20
|
Dragon J, Gołyszny M, Obuchowicz E. Co-treatment with cannabidiol and escitalopram in ineffective doses induces antidepressant effect in maternally separated male adolescent rats. Pharmacol Biochem Behav 2025; 250:174000. [PMID: 40090594 DOI: 10.1016/j.pbb.2025.174000] [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: 11/20/2024] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Due to low efficacy and delayed therapeutic effect of drugs currently used in the therapy of depression in adolescent population, a lot of effort has been put into finding new substances using alternative target points that could support treatment with traditional antidepressive drugs. Cannabidiol, compound derived from Cannabis sativa may have therapeutic potential in depressive disorders. This study aimed to investigate whether combined administration of escitalopram with cannabidiol in ineffective doses, will provide better or similar effects in behavioral tests compared to escitalopram in an effective dose in adolescent maternally separated rats. Maternal separation has been used as a form of early life adversity. The pups were separated from their dams for 360 min daily from postnatal day (PND) 2 until PND 15. Later, escitalopram (15 or 5 mg/kg) or vehicle were administered ip. in a subacute manner in mid-adolescent male rats. Cannabidiol (15 mg/kg) or vehicle were injected ip. in a single dose about 1 h prior to behavioral assessment. Three standard behavioral tests were performed: the elevated plus maze and the open field test on PND 42 and the forced swimming test on PND 43-44 on the subsequent groups of rats. The combined treatment with escitalopram and cannabidiol in ineffective doses did not induce anxiolytic-like effects but successfully relieved despair behavior in the forced swimming test showing similar efficacy as treatment with escitalopram in effective dose. This result might be the basis for future research and the development of new therapeutic strategies for treatment of adolescent depression.
Collapse
Affiliation(s)
- Jonasz Dragon
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Katowice, Poland.
| | - Miłosz Gołyszny
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Katowice, Poland.
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Katowice, Poland.
| |
Collapse
|
21
|
Nasanbuyan N, Yoshida M, Inutsuka A, Takayanagi Y, Kato S, Hidema S, Nishimori K, Kobayashi K, Onaka T. Differential Functions of Oxytocin Receptor-Expressing Neurons in the Ventromedial Hypothalamus in Social Stress Responses: Induction of Adaptive and Maladaptive Coping Behaviors. Biol Psychiatry 2025; 97:874-886. [PMID: 39343339 DOI: 10.1016/j.biopsych.2024.09.015] [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: 12/26/2023] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND The flexibility to adjust actions and attitudes in response to varying social situations is a fundamental aspect of adaptive social behavior. Adaptive social behaviors influence an individual's vulnerability to social stress. While it has been proposed that oxytocin is a facilitator of active coping behaviors during social stress, the exact mechanisms remain unknown. METHODS Using a social defeat stress paradigm in male mice, we identified the distribution of oxytocin receptor (OXTR)-expressing neurons in the ventrolateral part of the ventromedial hypothalamus (vlVMH) that are activated during stress by detection of c-Fos protein expression. We then investigated the role of vlVMH OXTR-expressing neurons in social defeat stress responses by chemogenetic methods or deletion of local OXTRs. The social defeat posture was measured for quantification of adaptive social behavior during repeated social stress. RESULTS Social defeat stress activated OXTR-expressing neurons rather than estrogen receptor 1-expressing neurons in the rostral vlVMH. OXTR-expressing neurons in the vlVMH were glutamatergic. Chemogenetic activation of vlVMH OXTR-expressing neurons facilitated exhibition of the social defeat posture during exposure to social stress, while local OXTR deletion suppressed it. In contrast, overactivation of vlVMH-OXTR neurons induced generalized social avoidance after exposure to chronic social defeat stress. Neural circuits for the social defeat posture centered on OXTR-expressing neurons were identified by viral tracers and c-Fos mapping. CONCLUSIONS vlVMH OXTR-expressing neurons are a functionally unique population of neurons that promote active coping behavior during social stress, but their excessive and repetitive activation under chronic social stress impairs subsequent social behavior.
Collapse
Affiliation(s)
- Naranbat Nasanbuyan
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan
| | - Masahide Yoshida
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan.
| | - Ayumu Inutsuka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan
| | - Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shizu Hidema
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuhiko Nishimori
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
22
|
Schipper L, van Heijningen S, Karapetsas G, Olivier JDA, van Dijk G. Social housing conditions, hierarchical status and testing order affect behavioral test outcomes of male C57BL6/J mice. Physiol Behav 2025; 293:114859. [PMID: 39999904 DOI: 10.1016/j.physbeh.2025.114859] [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: 10/16/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 02/27/2025]
Abstract
Male mice are frequently used for behavioral neuroscience research, but outcomes of behavioral tests are often variable across studies, contributing to poor reproducibility. Social housing conditions, social hierarchical status and within-cage order of testing are factors that likely influence behavioral outcomes, but it is unknown to what extent and if these factors interact. For this purpose, behavior of socially and individually housed male C57BL6/J mice was studied upon subjection to the open field test, the elevated plus-maze test and the three-chamber social test. In socially housed animals, effects of social hierarchical status and within-cage testing order were evaluated. We show that the differences in behavior outcomes between individually and socially housed mice depend on the social hierarchical status and the test order of the socially housed mice. Careful consideration of these factors in the design, analysis and interpretation of behavioral experiments with socially housed mice can lead to more precise results and more reliable research outcomes.
Collapse
Affiliation(s)
- Lidewij Schipper
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands; Danone Research and Innovation, Utrecht, The Netherlands.
| | - Steffen van Heijningen
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Giorgio Karapetsas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Jocelien D A Olivier
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Gertjan van Dijk
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Sarfi M, Elahdadi Salmani M, Lashkarbolouki T, Goudarzi I. Divergent effects of noradrenergic activation and orexin receptor 1 blockade on hippocampal structure, anxiety-like behavior, and social interaction following chronic stress. Pharmacol Biochem Behav 2025; 250:173997. [PMID: 40073949 DOI: 10.1016/j.pbb.2025.173997] [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: 12/18/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Chronic stress (Ch.S) has detrimental effects on the brain's structure and function, particularly in the hippocampus. The noradrenergic and orexinergic systems play crucial roles in the stress response and regulation of stress-related behaviors. This study aimed to investigate the interaction between noradrenergic activation and orexin receptor 1 inhibition on chronic stress-induced hippocampal alterations. The study conducted experiments on male Wistar rats, subjected to Ch.S, OXr1 blocking, noradrenergic activation, or a combination of these treatments. Plasma corticosterone level was measured using a fluorometric method. Behavioral assessment of social maze, elevated plus maze (EPM) and novel object recognition (NOR) test were performed. Then, the expression of prepro-orexin, OXr1, and glucocorticoid receptor (GR) was analyzed using semiquantitative RT-PCR. Neuronal populations were quantified through Nissl staining. The data revealed that all stress and yohimbine groups had elevated plasma corticosterone levels. Ch.S significantly altered behavior, impairing social interaction, disrupting object recognition memory and increasing anxiety-like responses in the EPM. OXr1 blocking reversed these stress-induced behavioral deficits, while yohimbine did not improve these behavioral outcomes. Chronic stress led to a significant increase in prepro-orexin, OXr1, and GR expression. While blocking OXr1 helped counteract these stress-induced changes, yohimbine failed to restore the expression levels. Ch.S reduced hippocampal neuronal populations, while OXr1 blocking partially reversed this effect, and yohimbine further recovered the reversal. These findings indicate that blocking hippocampal OXr1 can mitigate the adverse effects of chronic stress on both hippocampal structure and anxiety-like behaviors, while noradrenergic signaling appears to have differential effects on behavioral and cellular measures.
Collapse
Affiliation(s)
| | | | | | - Iran Goudarzi
- School of Biology, Damghan University, Damghan, Iran.
| |
Collapse
|
24
|
Leung WL, Shad A, Perucca P, O'Brien TJ, Semple BD, Casillas-Espinosa PM. Chronic outcomes after mild-moderate traumatic brain injury in adult seizure-prone (FAST) and seizure-resistant (SLOW) rats: A model for understanding genetic contributions to acquired epileptogenesis? Epilepsy Behav 2025; 166:110347. [PMID: 40022952 DOI: 10.1016/j.yebeh.2025.110347] [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: 09/25/2024] [Revised: 01/31/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Post-traumatic epilepsy (PTE) is a common, serious, long-term complication of traumatic brain injury (TBI). However, only a minority of individuals will develop epilepsy after a TBI, and the contribution of genetic predisposition to the risk of acquired epilepsy warrants further exploration. In this study, we examined whether innate, genetically determined differences in seizure susceptibility between seizure-prone FAST and seizure-resistant SLOW rat strains would influence chronic behavioral and PTE outcomes after experimental TBI. We hypothesized that FAST rats would show increased vulnerability to PTE and poorer neurobehavioral outcomes. Using the lateral fluid percussion injury model, we first determined the optimal injury parameters to generate a mild-moderate TBI in young adult FAST rats, which had previously shown high mortality to severe TBI. Then, FAST and SLOW rats underwent TBI or sham surgery, and a series of behavioral tests were performed either acutely (within 4 weeks) or chronically (more than 22 weeks) post-injury. Acutely, FAST rats showed an increased physiological response to TBI with a longer apnea duration, delayed pain response, and delayed self-righting, as well as increased acute seizure-like behavior compared to SLOW rats. Conversely, SLOW rats showed greater neuromotor deficits and weight loss sub-acutely compared to FAST rats. Chronically, while strain-specific phenotypes were observed (e.g., FAST rats showing increased anxiety-like behavior, altered nociceptive responses, and polydipsia), no TBI effects were detected. Analysis of continuous video-electroencephalographic recordings over a 1-month period starting at 6 months post-TBI did not reveal any spontaneous seizures. However, periodic epileptiform discharges were only found in FAST rats that had a TBI. Together, these findings reflect fundamental differences in chronic behavior and epileptiform discharges as a result of innate distinctions in epileptogenic susceptibility in FAST versus SLOW rats. However, a lack of spontaneous seizure activity or chronic neurobehavioral deficits in TBI animals confounded our ability to address the initial hypothesis, such that alternative injury models may be more suitable to study genetic contributions to the development of PTE.
Collapse
Affiliation(s)
- Wai Lam Leung
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Ali Shad
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia; Department of Immunology & Pathology, Monash University, Melbourne, VIC 3004, Australia
| | - Piero Perucca
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia; Epilepsy Research Centre, Department of Medicine (Austin Health), The University of Melbourne, Heidelberg, VIC 3084, Australia; Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Heidelberg, VIC 3084, Australia; Department of Neurology, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia; Department of Neurology, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC 3050, Australia
| | - Bridgette D Semple
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC 3050, Australia.
| |
Collapse
|
25
|
Akkaya A, Aykan D, Gencturk S, Unal G. Intermittent environmental enrichment induces behavioral despair, while intermittent social isolation impairs spatial learning in rats. Pharmacol Biochem Behav 2025; 250:174001. [PMID: 40118218 DOI: 10.1016/j.pbb.2025.174001] [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: 05/28/2024] [Revised: 03/01/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Environmental enrichment and social isolation constitute two well-studied experimental manipulations that result in several behavioral, neural, and molecular changes in rodents. Enrichment is linked to enhanced cognitive performance, and mitigation of different nervous system injuries and disorders. In contrast, social isolation or impoverished environment often induce negative effects on cognitive and affective systems. Both manipulations are typically examined with a short-term or chronic exposure, which cannot capture the actual human experiences. In this study, we explored the behavioral and neural alterations led by intermittent environmental enrichment or social isolation in adult Wistar rats. Animals were assigned to an enriched condition (EC), isolation/impoverished condition (IC), or standard condition (SC). The differential housing protocol involved transferring the animals to their respective cages for two days at the end of each five-day standard housing period for 8 weeks. Enriched animals exhibited behavioral despair in the forced swim test without differential overall locomotor activity. In the Morris water maze, impoverished animals displayed a slower learning rate compared to the SC and EC groups. In line with this, the IC group had fewer parvalbumin (PV) immunopositive (+) cells in the CA1 and dentate gyrus. No differences were observed in PV+ cell levels in the amygdala, while the IC group had more c-Fos+ cells in the same region following acute restraint stress. These findings implicate that intermittent isolation or enrichment are sufficient to trigger distinct behavioral changes at the cognitive and affective domains, and pinpoint PV as a biomarker for environmentally induced alterations in hippocampal memory performance.
Collapse
Affiliation(s)
- Aybuke Akkaya
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342 Istanbul, Turkey
| | - Deren Aykan
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342 Istanbul, Turkey
| | - Sinem Gencturk
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342 Istanbul, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342 Istanbul, Turkey.
| |
Collapse
|
26
|
Carnevali L, Barbetti M, Fotio Y, Ferlenghi F, Vacondio F, Mor M, Piomelli D, Sgoifo A. Enhancement of peripheral fatty acyl ethanolamide signaling prevents stress-induced social avoidance and anxiety-like behaviors in male rats. Psychopharmacology (Berl) 2025; 242:997-1009. [PMID: 37932554 PMCID: PMC12043783 DOI: 10.1007/s00213-023-06473-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/25/2023] [Indexed: 11/08/2023]
Abstract
RATIONALE Exposure to traumatic events can lead to alterations in social and anxiety-related behaviors. Emerging evidence suggests that peripheral host-defense processes are implicated in the expression of stress-induced behavioral responses and may be targeted to mitigate the negative sequalae of stress exposure. OBJECTIVES In this study, we used the peripherally restricted FAAH inhibitor URB937 to investigate the effects of the fatty acyl ethanolamide (FAE) family of lipid mediators - which include the endocannabinoid anandamide and the endogenous PPAR-α agonists, oleoylethanolamide and palmitoylethanolamide - on behavioral and peripheral biochemical responses to two ethologically distinct rat models of stress. METHODS Male adult rats were exposed to acute social defeat, a model of psychological stress (Experiment 1), or to the predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT), a test of innate predator-evoked fear (Experiment 2), and subsequently treated with URB937 (1 or 3 mg/kg, intraperitoneal) or vehicle. Behavioral analyses were conducted 24 h (Experiment 1) or 7 days (Experiment 2) after exposure. RESULTS URB937 administration prevented the emergence of both social avoidance behavior after social defeat stress and anxiety-related behaviors after TMT exposure. Further, URB937 administration blocked social defeat-induced transient increase in plasma concentrations of pro-inflammatory cytokines and the elevation in plasma corticosterone levels observed 24 h after social defeat CONCLUSIONS: Enhancement of peripheral FAAH-regulated lipid signaling prevents the emergence of stress-induced social avoidance and anxiety-like behaviors in male rats through mechanisms that may involve an attenuation of peripheral cytokine release induced by stress exposure.
Collapse
Affiliation(s)
- Luca Carnevali
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
| | - Margherita Barbetti
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Yannick Fotio
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | | | | | - Marco Mor
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
- Department of Biological Chemistry, University of California, Irvine, CA, 92697, USA
| | - Andrea Sgoifo
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| |
Collapse
|
27
|
Raddatz MC, Newson CM, Stott M, Campbell C, Bobeck EN. GPR171 is necessary for normal physiological functions and mood-related behaviors in males, but not females. Behav Brain Res 2025; 490:115618. [PMID: 40318809 DOI: 10.1016/j.bbr.2025.115618] [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: 12/19/2024] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
GPR171 is a recently deorphanized G protein-coupled receptor that has been implicated in feeding, mood regulation, and pain. However, the role of this receptor in other physiological functions and in female mice is largely unexplored. Using a novel genetic GPR171 knockout mouse model, we examined general physiological behaviors and mood-related behaviors in both sexes. In male GPR171 knockout mice, we observed increased feeding, reduced water intake, heightened ataxia, and decreased anxiety-like behaviors. Male GPR171 knockout mice showed greater depressive-like behaviors compared to female knockout mice, but this did not correspond to a change in cFos activity in the basolateral amygdala. No significant changes were detected in female GPR171 knockout mice in other behaviors measured. These findings corroborate previous studies linking GPR171 to feeding and mood regulation in males, but highlight notable sex differences that were previously unknown. These sex differences indicate that future studies should focus on elucidating the behavioral effects of GPR171 in females to provide a more comprehensive understanding of its physiological roles.
Collapse
Affiliation(s)
- Megan C Raddatz
- Department of Biology, Utah State University, Logan, UT, United States; Interdisciplinary Neuroscience Program, Utah State University, Logan, UT, United States
| | - Callie M Newson
- Department of Biology, Utah State University, Logan, UT, United States; Interdisciplinary Neuroscience Program, Utah State University, Logan, UT, United States
| | - Mitchel Stott
- Department of Biology, Utah State University, Logan, UT, United States
| | - Colton Campbell
- Department of Biology, Utah State University, Logan, UT, United States
| | - Erin N Bobeck
- Department of Biology, Utah State University, Logan, UT, United States; Interdisciplinary Neuroscience Program, Utah State University, Logan, UT, United States.
| |
Collapse
|
28
|
Xu X, Gong Q, Wang XD. MK-801 attenuates one-trial tolerance in the elevated plus maze via the thalamic nucleus reuniens. Neuropharmacology 2025; 268:110318. [PMID: 39842626 DOI: 10.1016/j.neuropharm.2025.110318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/04/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Anxiety, a future-oriented negative emotional state, is characterized by heightened arousal and vigilance. The elevated plus maze (EPM) test is a widely used assay of anxiety-related behaviors in rodents and shows a phenomenon where animals with prior test experience tend to avoid open arms in retest sessions. While this one-trial tolerance (OTT) phenomenon limits the reuse of the EPM test, the potential mechanisms remain unsolved. Here, we found that neither anxiogenic factors like acute restraint stress nor anxiolytic factors like diazepam (2 mg/kg) influenced the emergence of the OTT phenomenon in mice in the EPM test. In contrast, OTT was markedly attenuated by MK-801 (0.1 mg/kg), a non-competitive N-methyl-D-aspartate receptor antagonist. Through the use of c-fos mapping, MK-801 was found to increase neuronal activation in the thalamic nucleus reuniens (Re). Moreover, chemogenetic inactivation of Re neurons could prevent the effects of MK-801. Our findings suggest the Re as a crucial brain region in emotional adaptation in the EPM and shed light on the experimental design optimization and mechanistic investigation of anxiety-related behaviors.
Collapse
Affiliation(s)
- Xue Xu
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China; Lingang Laboratory, Shanghai, 200031, China.
| | - Qian Gong
- Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Xiao-Dong Wang
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China; Department of Psychiatry of Sir Run Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, 310058, Hangzhou, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
29
|
Lima JLS, Amaral AR, Cavalcante AMDO, Chagas AKO, Oliveira DN, Melo JC, Leite GDO, Sessle BJ, Campos AR. Anxiety- and nociception-like behaviours in mature adult mice induced by audiovisual overstimulation during infancy. Brain Res Bull 2025; 224:111314. [PMID: 40127727 DOI: 10.1016/j.brainresbull.2025.111314] [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: 12/11/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVES To evaluate the behavioural effects in adult mice previously subjected to audiovisual overstimulation during infancy and adolescence. METHODS Mice aged 21, 26 and 36 days (p21, p26 and p36) underwent auditory (70 db) and visual (flashing lights) stimulation for 2 or 6 h per day until p64; naive animals were used as controls. At p200, tests assessed respectively motor activity (open field test), depression (forced swimming and splash tests), anxiety (hole board, plus maze and marble burying tests, aggression (resident-intruder test), and nociception (von Frey and hot plate tests). RESULTS There were no significant (ANOVA, p > 0.05) behavioural changes in forced swimming, splash, hole board, or marble burying tests between overstimulated and naive groups. However, the p21 group showed significantly (ANOVA, p < 0.05) increased anxiety-like behaviour (2 h) in the elevated plus maze test and altered nociceptive behaviour in the von Frey test (2 and 6 h). The p26 group (2 h) displayed significantly reduced rearing behaviours, fewer entries in the plus maze test, and faster reaction times to noxious thermal stimulation (2 h). CONCLUSION Audiovisual overstimulation during early development can promote anxiety-like behaviour and affect nociceptive behaviour in adult mice.
Collapse
Affiliation(s)
- Jessica L S Lima
- Experimental Biology Center, University of Fortaleza, Fortaleza, Brazil
| | - Amanda R Amaral
- Graduate Program in Medical Sciences, University of Fortaleza, Fortaleza, Brazil
| | | | | | | | - Juliana C Melo
- Medical School, University of Fortaleza, Fortaleza, Brazil
| | | | - Barry J Sessle
- Faculty of Dentistry and Temerty Faculty of Medicine Department of Physiology, and Centre for the Study of Pain, University of Toronto, Toronto, Canada
| | - Adriana R Campos
- Experimental Biology Center, University of Fortaleza, Fortaleza, Brazil; Graduate Program in Medical Sciences, University of Fortaleza, Fortaleza, Brazil; Medical School, University of Fortaleza, Fortaleza, Brazil.
| |
Collapse
|
30
|
Gilfarb RA, Ranade S, Smail M, Wangler L, Stewart M, Rajesh A, Lenz KM, Leuner B. Hormonal contraceptives during adolescence impact the female brain and behavior in a rat model. Horm Behav 2025; 171:105725. [PMID: 40188588 DOI: 10.1016/j.yhbeh.2025.105725] [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: 10/15/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/08/2025]
Abstract
Millions of people take hormonal contraceptives (HCs), often starting during adolescence when ovarian hormones influence brain and behavioral maturation. However, there is a fundamental lack of information about the neurobehavioral consequences of hormonal alterations via adolescent HC use. To begin addressing this gap, we validated a rodent model of adolescent HC administration and characterized its impact on endocrine, transcriptional, and behavioral endpoints. Cohorts of intact post-pubertal female Sprague-Dawley rats received daily subcutaneous injections of either vehicle or HC [10 μg ethinyl estradiol (EE) + 20 μg levonorgestrel (LNG)] for the duration of adolescence from postnatal day (PND) 35 to PND56. Blood and brain tissue was collected at PND57. Other cohorts received daily injections of vehicle or HC from PND35 until behavioral assays were completed on PND57-64. HC treatment was effective, as vaginal lavage indicated disrupted estrous cycling and ELISA indicated suppressed serum luteinizing hormone in HC-treated rats. Liquid chromatography-mass spectrometry analysis showed EE and LNG in serum and brain as well as diminished serum and brain levels of allopregnanolone and testosterone in HC-treated rats. NanoString nCounter analysis indicated that adolescent HC administration impacted expression of genes related to synapses, white matter, neuroimmune, monoamine, and hormone signaling in the hypothalamus and medial prefrontal cortex. While no effects of HCs were seen on sociability in the social preference test or stress coping behavior in the forced swim test, adolescent HC administration diminished risk-assessment behaviors in the novelty-induced hypophagia paradigm and altered anxiety-like behavior in the open field test and elevated plus maze. Overall, these data suggest that exposure to contraceptive hormones during the critical developmental period of adolescence may shape the brain and behavior.
Collapse
Affiliation(s)
- Rachel A Gilfarb
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Sanjana Ranade
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Marissa Smail
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Lynde Wangler
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Meredith Stewart
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Abhishek Rajesh
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 370 W. 9th Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 370 W. 9th Avenue, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
31
|
Odendaal‐Gambrell CP, O'Brien C, Cairns M, Maarman GJ, Joseph DE, Smith C, Rautenbach F, Marnewick JL, Essop MF. Chronic stress elicits sex-specific mitochondrial respiratory functional changes in the rat heart. Physiol Rep 2025; 13:e70371. [PMID: 40356314 PMCID: PMC12069860 DOI: 10.14814/phy2.70371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Although chronic psychosocial stress is linked to cardiovascular diseases, the underlying mechanisms remain elusive. For this study, we focused on the mitochondrion as a putative mediator of stress-related cardiac pathologies in a sex-dependent manner. Male and female Wistar rats were subjected to chronic stress for 4 weeks (mimicking an anxious phenotype) versus matched controls. Cardiac redox status, mitochondrial respiration parameters, and expression levels of proteins involved in mitochondrial oxidative phosphorylation, dynamics, and biogenesis were evaluated. Despite limited changes in behavior and circulating stress hormones (both sexes), stressed males exhibited altered cardiac oxidative phosphorylation via β-oxidation- and glucose oxidation-linked respiratory pathways together with increased myocardial antioxidant capacity and decreased lipid peroxidation. Conversely, stressed females exhibited a protective and resilient phenotype by displaying augmented levels of major mitochondrial respiratory complexes (complex I, III, and ATP synthase) and a fusion marker (mitofusin-2 [Mfn2]), together with attenuated expression of a fission marker (dynamin-related protein-1 [Drp1]) despite decreased estradiol levels. In contrast, stressed males displayed increased cardiac ATP synthase levels together with diminished peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α) expression versus controls. These findings indicate that male mitochondria are more prone to stress-related functional changes, while females exhibited a more protective and resilient phenotype.
Collapse
Affiliation(s)
- Caitlin P. Odendaal‐Gambrell
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Cassidy O'Brien
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Megan Cairns
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Gerald J. Maarman
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Danzil E. Joseph
- Centre for Cardiometabolic Research in Africa (CARMA), Department of Physiological Sciences, Faculty of ScienceStellenbosch UniversityStellenboschSouth Africa
| | - Carine Smith
- Experimental Medicine Unit, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch UniversityStellenboschSouth Africa
| | - Fanie Rautenbach
- Applied Microbial and Health Biotechnology InstituteCape Peninsula University of TechnologyCape TownSouth Africa
| | - Jeanine L. Marnewick
- Applied Microbial and Health Biotechnology InstituteCape Peninsula University of TechnologyCape TownSouth Africa
| | - M. Faadiel Essop
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| |
Collapse
|
32
|
Valeeva EV, Nikitin DO, Nikiforova LS, Semina II, Ahmetov II. Effects of Pharmacological Treatment on Telomere Length and the Expression of Telomerase/Shelterin-Related Genes in Rat Models of Autism. J Mol Neurosci 2025; 75:55. [PMID: 40272729 PMCID: PMC12021733 DOI: 10.1007/s12031-025-02353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 04/16/2025] [Indexed: 04/27/2025]
Abstract
Telomeres are increasingly recognized for their potential role in the etiology of autism spectrum disorder (ASD) due to their involvement in cellular aging and telomerase-shelterin function. Although shorter telomeres have been observed in individuals with ASD, studies linking telomere dynamics in blood cells and brain regions remain limited. Using the valproic acid (VPA, 500 mg/kg) rodent model, this study aimed to assess the impact of three drugs commonly used in ASD treatment (amitriptyline, risperidone, and nooclerin) on telomere length and the expression of telomerase/shelterin-related genes (Dkc1, Gar1, Pot1a, Pot1b, Tep1, Terc, Terf2ip, Tert, Tinf2, Tnks, Tpp1, Trf1, and Trf2) in blood cells, the prefrontal cortex, and hippocampus of VPA-exposed Wistar rats. Telomere length and gene expression were measured using quantitative PCR. Risperidone treatment in VPA males resulted in telomere elongation and increased expression of Tnks in blood cell and Trf1, Trf2 genes in prefrontal cortex. Nooclerin treatment also showed beneficial effects on telomere length of blood cell in males, alongside increased Trf1 expression. Long telomeres in male blood cells were associated with reduced anxiety, while a positive correlation was found between Tpp1 expression and stereotypical behavior in both male and female VPA rats. These findings suggest that nooclerin and risperidone influence telomere length and gene expression related to the telomere-telomerase complex in a sex-dependent manner, offering insights into the neurobiological mechanisms underlying ASD.
Collapse
Affiliation(s)
- Elena V Valeeva
- Central Research Laboratory, Kazan State Medical University, Kazan, 420012, Russia
- Laboratory of Genetics of Aging and Longevity, Kazan State Medical University, Kazan, 420012, Russia
| | - Dmitry O Nikitin
- Pharmacology Department, Kazan State Medical University, Kazan, 420012, Russia
| | - Lubov S Nikiforova
- Central Research Laboratory, Kazan State Medical University, Kazan, 420012, Russia
| | - Irina I Semina
- Central Research Laboratory, Kazan State Medical University, Kazan, 420012, Russia
- Pharmacology Department, Kazan State Medical University, Kazan, 420012, Russia
| | - Ildus I Ahmetov
- Central Research Laboratory, Kazan State Medical University, Kazan, 420012, Russia.
- Laboratory of Genetics of Aging and Longevity, Kazan State Medical University, Kazan, 420012, Russia.
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
33
|
Ge Q, Zhou S, Porras J, Fu P, Wang T, Du J, Li K. Investigating post-infection anxiety- and depression-like behaviors in a SARS-CoV-2 mouse model. Theranostics 2025; 15:5738-5755. [PMID: 40365287 PMCID: PMC12068287 DOI: 10.7150/thno.102752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 03/27/2025] [Indexed: 05/15/2025] Open
Abstract
Rationale: The COVID-19 pandemic, driven by SARS-CoV-2, has resulted in a wide range of neuropsychiatric symptoms associated with post-acute sequelae (PASC). However, the mechanisms by which SARS-CoV-2 impacts the brain and leads to persistent behavioral changes remain poorly understood. We hypothesize that SARS-CoV-2 exposure induces neuroinflammation and microglial activation, leading to anxiety- and depression-like behaviors in mice. Methods: We established a SARS-CoV-2 mouse model using the virulent SARS2-N501YMA30 strain to investigate its impact on the central nervous system (CNS). We assessed neuroinvasion via immunostaining of dsRNA and markers for neuronal, astrocyte, and microglia in brain slices. Behavioral changes were evaluated at 2 weeks, 2 months, and 4 months post-infection. Molecular and cellular analyses included bulk RNA-seq, Golgi-Cox staining, field excitatory postsynaptic potential (fEPSP) recordings, immunofluorescence, and quantitative real-time PCR (qRT-PCR) to assess gene expression, neuronal morphology, and microglial activation in the brain. Results: We demonstrated that intranasal inoculation of SARS2-N501YMA30 results in viral dissemination to multiple brain regions, including the amygdala and the prefrontal cortex (PFC). Behavioral assays indicated a marked elevation in anxiety- and depression-like behaviors post-infection. A comparative analysis of RNA expression profiles disclosed alterations in the post-infected brains. Additionally, we observed dendritic spine remodeling on neurons within the amygdala after infection. Infection with SARS2-N501YMA30 was associated with microglial activation and a subsequent increase in microglia-dependent neuronal activity in the amygdala. Transcriptomic analysis of infected brains revealed the upregulation of inflammatory and cytokine-related pathways, implicating neuroinflammation in the pathogenesis of neuronal hyperactivity and behavioral abnormality. Conclusion: Our findings provide evidence that SARS-CoV-2 neuroinvasion plays a critical role in the development of lasting behavioral sequelae observed in PASC. These data provide critical insights into the neurological consequences of SARS-CoV-2 infection and underscore microglia as a potential therapeutic target for ameliorating virus-induced neurobehavioral abnormalities.
Collapse
Affiliation(s)
- Qian Ge
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Shan Zhou
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL 34987, USA
| | - Jose Porras
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL 34987, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, 34987, USA
| | - Jianyang Du
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kun Li
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL 34987, USA
| |
Collapse
|
34
|
Xue R, Liu K, Hu X, Ma X, Li S, Deng Z, Zhong K, Yang J, Zhu X, Liu S, Shi Z, Zhou M, Tang Y. Longitudinal observation of radiation-induced cognitive impairment and emotional dysfunction: Based on animal model and clinical cohort. Neuroscience 2025; 572:171-181. [PMID: 39921023 DOI: 10.1016/j.neuroscience.2025.02.010] [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: 10/24/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Radiation-induced cognitive impairment (RICI) and emotional disorder (RIED) are comorbidities that seriously affect the quality of life in patients with radiation-induced brain injury (RIBI). A longitudinal study was conducted to observe the occurrence and development of RICI and RIED in an RIBI animal model and a clinical cohort following intensity modulated radiotherapy (IMRT). RIBI mice and sham controls were subjected to three cycles of behavior tests for cognitive and emotional function at post-irradiation 1 week (1w), 5 weeks (5w), and 9 weeks (9w) corresponding to early, middle and late stage after radiotherapy. Additionally, 139 patients who underwent IMRT after nasopharyngeal carcinoma and were firstly diagnosed with radiation-induced brain injury were enrolled. Pre-treatment and follow-up neuropsychological assessments of cognition, anxiety and depression were completed. Compared with control, significant declines in working memory, object recognition memory and social memory were observed in RIBI mice at post-irradiation 5w and 9w. Longitudinal observations revealed that memory impairment predominantly occurred in the middle stage and persisted into the late stage. Anxiety-like behaviors were only observed at post-irradiation 9w. In the clinical cohort, RICI exhibited a parallel cumulative incidence curve and a similar median onset to RIBI. RICI predominantly occurred 2-6 years post-IMRT 2-6 and progressively deteriorated beyond 6 years while RIED gradually increased beyond 6 years after IMRT. During two-year follow-up visits, half of the patients with RICI combined with RIBI benefited from drug treatment, achieving stable or improved cognition, while the other half showed no response or experienced cognition aggravation. In summary, RICI predominantly occurred in the middle stage post-irradiation and progressed to the late stage while RIED mostly emerged in the late stage in RIBI. Consistency in the development process of RICI and RIED was observed in the animal model and the clinical cohort.
Collapse
Affiliation(s)
- Ruiqi Xue
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kejia Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xia Hu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xueying Ma
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shaojian Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhenhong Deng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ke Zhong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingwen Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoqiu Zhu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Brain Research Center, Department of Neurology, Vice President, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
35
|
Fernández PR, Gaydou L, Schumacher R, Rossetti MF, García AP, Sabella A, Ramos JG, Canesini G, Stoker C. Early overfeeding and adult anhedonia: Impact of neonatal nutrition on hedonic food regulation in male rats. J Nutr Biochem 2025; 143:109933. [PMID: 40254039 DOI: 10.1016/j.jnutbio.2025.109933] [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: 12/16/2024] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
The aim of our study was to analyze the impact of early-life overnutrition and the exposure in adulthood to a cafeteria diet (CAF) on eating behavior and on the expression of key genes involved in the regulation of food intake. Male Wistar rats were raised in small (SL, 4 pups/dam) or normal litters (NL, 10 pups/dam), fed a control diet (CON) until postnatal day (PND) 90. Then, they received CON or CAF for 11 weeks (NL-CON, NL-CAF, SL-CON, SL-CAF; 12±2 rats/group). Body weight, food intake and behavioral tests (Elevated Plus Maze: EPM, Sensory-specific satiety: SSS) were assessed. At PND167, the rats were euthanized to obtain brain, blood and fat pads. Ventral tegmental area (VTA), Nucleus Accumbens (NAc) and Arcuate Nucleus (Arc), were isolated by micropunch technique for qPCR analysis. Early overfeeding alone had the ability to alter long-term SSS. CAF groups showed increased body weight, adiposity and energy intake; sweet food preference and altered SSS. SL-CAF showed hypophagia, basal hyperglycemia, altered SSS and anxiety-like behavior. Both NL-CAF and SL-CAF showed antidopaminergic effects, but through different pathways: NL-CAF reduced dopamine (DA) production in VTA via decreased tyrosine hydroxylase (TH) expression, while SL-CAF exhibited an increase in dopamine active transporter (DAT) expression in NAc enhancing clearance. SL decreased Neuropeptide Y (NPY) expression in the Arc in adulthood, which has been proposed to be the link between homeostatic and hedonic systems. Our research reveals a key link between early-life overnutrition and adult hedonic feeding control, emphasizing its lasting impact on eating behavior and the potential for innovative therapeutics to combat obesity.
Collapse
Affiliation(s)
- Pamela Rocío Fernández
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Cátedra de Nutrición en Situaciones Patológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Luisa Gaydou
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Rocío Schumacher
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina
| | - María Florencia Rossetti
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina
| | - Ana Paula García
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina
| | - Agustina Sabella
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina
| | - Jorge Guillermo Ramos
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Guillermina Canesini
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Cátedra de Nutrición en Situaciones Patológicas, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Cora Stoker
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
36
|
Paul S, Chandran R, Vijayan DK, Bhardwaj J, Singh P, Shetty P, Cheruku S, Meleveetil S, Balachandran Krishnamma B. A Cdk5 inhibitor restores cognitive function and alleviates type 2 diabetes in mice. iScience 2025; 28:112200. [PMID: 40224020 PMCID: PMC11986975 DOI: 10.1016/j.isci.2025.112200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/29/2025] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disorder commonly linked with cognitive decline, increasing patients' susceptibility to dementia. Alzheimer's disease (AD) has a strong connection with hyperglycemia and insulin dysregulation. Interestingly, certain anti-diabetic drugs have shown potential in reducing T2D-induced cognitive impairment. Previous studies, including ours, have highlighted the dysregulation of cyclin-dependent kinase 5 (Cdk5) activity in both T2D and AD, which may contribute to pathological changes in these conditions. Thus, targeting the Cdk5 kinase could offer a therapeutic approach for T2D and cognitive deterioration. Our research identifies Cdk5 as a key link between T2D and cognitive decline. By screening the KINACore library, we discovered two new brain-penetrant Cdk5 inhibitors, BLINK11 and BLINK15. In a high-fat diet-induced T2D model, these inhibitors improved blood glucose levels, obesity, and cognitive function. BLINK11, in particular, shows promise as a therapeutic candidate for treating cognitive impairment associated with T2D.
Collapse
Affiliation(s)
- Sangita Paul
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Remya Chandran
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Dileep K. Vijayan
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Juhi Bhardwaj
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Praveen Singh
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Poornima Shetty
- Srinivasa Engineering College, Mukka, Mangalore 574146, India
| | - Srinivas Cheruku
- Department of Chemistry, Manasa Gangotri, Mysore University, Mysuru 570005, India
| | - Sajith Meleveetil
- Department of Chemistry, SSIT, Sri Siddhartha Academy of Higher Education, Tumkur 572107, Karnataka, India
| | - Binukumar Balachandran Krishnamma
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
37
|
Reiszadeh Jahromi S, Keikhosravi Z, SoukhakLari R, Moosavi M. Curcumin mitigates memory deficits induced by subcutaneous aluminum nanoparticle administration through modulation of hippocampal brain-derived neurotrophic factor and Akt signaling pathways. Behav Pharmacol 2025:00008877-990000000-00130. [PMID: 40293979 DOI: 10.1097/fbp.0000000000000825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Research has indicated a strong link between exposure to aluminum (Al) and the development of Alzheimer's disease (AD). Given the rising use of Al nanoparticles, which are far more neurotoxic than Al, it is noteworthy to investigate the possible protective properties of natural substances. Curcumin, an important component of turmeric, has demonstrated neuroprotective effects in some animal studies. The main objective of this study was to examine the protective effects of curcumin on the memory deficit induced by subcutaneous aluminum oxide nanoparticles (Al-NP) administration in mice. Additionally, considering the roles of the hippocampal brain-derived neurotrophic factor (BDNF) and Akt pathway in AD pathology, their levels were evaluated. Adult male Swiss mice (SWR/J) were administered Al-NP (10 mg/kg/s.c.) with or without curcumin (2.5, or 25 mg/kg/P.O) for 10 days. Memory and anxiety-like behavior were assessed using passive avoidance and elevated plus maze tasks, respectively. Western blot analysis was employed to measure hippocampal BDNF and Akt proteins in the hippocampus. The findings revealed that Al-NP induced memory impairment in mice, whereas curcumin at 25 mg/kg prevented this memory deficit. Additionally, Al-NP significantly reduced the hippocampal BDNF and phosphorylated Akt levels, while curcumin increased BDNF and phosphorylated Akt to a nonsignificant level compared to the control group. These results not only suggest the neuroprotective properties of curcumin but also suggest a possible association between hippocampal BDNF and Akt signaling in the neuroprotective mechanism of this compound against Al-NP toxicity.
Collapse
Affiliation(s)
| | - Zahra Keikhosravi
- Department of Biology, University of Sistan and Balouchestan, Zahedan, Iran
- Shiraz Neuroscience Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Moosavi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Messore F, Narayanan Therpurakal R, Dufour JP, Hoerder-Suabedissen A, Guidi L, Korrell K, Mueller M, Abuelem M, Lak A, Bannerman DM, Mann EO, Molnár Z. An orexin-sensitive subpopulation of layer 6 neurons regulates cortical excitability and anxiety behaviour. Transl Psychiatry 2025; 15:147. [PMID: 40229262 PMCID: PMC11997144 DOI: 10.1038/s41398-025-03350-2] [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: 10/11/2024] [Revised: 02/26/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
Cortical layer 6 neurons are the only projection neuron population in the cortical mantle known to electrophysiologically respond to orexin-a neuropeptide involved in cortical arousal and emotive behaviour. These neurons exhibit extensive intercortical and thalamic projections, yet the exact mechanisms underlying these responses are not fully understood. We hypothesize that cortical circuits activated by orexin sensitive L6 neurons in the medial prefrontal cortex (mPFC) are responsible for detecting salient features of sensory stimuli and are therefore involved in regulating emotional states. Here, we show that Drd1a-Cre+ neurons in the mPFC are selectively sensitive to orexin and gate the activation of the prefrontal network in vivo. Moreover, we demonstrated that chronically "silencing" this subpopulation of L6 neurons (Drd1a-Cre+/+:Snap25fl/fl) across the cortical mantle from birth abolishes the orexin-induced prefrontal activation. Consequently, the chronic silencing of these neurons had strong anxiolytic effects on several anxiety-related behavioural paradigms, indicating that orexin-responsive L6 neurons modulate emotional states and may be a substrate for anxiety regulation.
Collapse
Affiliation(s)
- Fernando Messore
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Rajeevan Narayanan Therpurakal
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jean-Philippe Dufour
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | | | - Luiz Guidi
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Kim Korrell
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Marissa Mueller
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Mohammed Abuelem
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Armin Lak
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Edward O Mann
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA.
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA.
| |
Collapse
|
39
|
de Brito MC, Costa BY, Souza Lima TAD, Camarini R. Environmental enrichment induces depressive- and anxiety-like behaviors in male Balb/C mice. Behav Brain Res 2025; 483:115462. [PMID: 39892654 DOI: 10.1016/j.bbr.2025.115462] [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: 11/06/2024] [Revised: 12/19/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Depression and anxiety disorders are prevalent neuropsychiatric conditions worldwide that impose substantial economic and social burdens worldwide. Environmental enrichment (EE) has been employed to investigate how the environment can influence these disorders. While EE is known to mitigate depressive and anxiety phenotypes across various mouse strains, the Balb/C strain exhibits greater sensitivity to different environmental stimuli. In this study, we aimed to assess the long-term effects of EE introduced after weaning on emotional behaviors in adulthood. Balb/C mice were weaned on postnatal day (PND) 21 and exposed to chronic EE for 3, 12, or 24 hours daily until PND 66. Depressive- and anxiety-like behaviors were assessed using the open field, elevated plus maze, and tail suspension tests, along with measurements of corticosterone plasma levels. EE exposure induced emotional dysregulation, evidenced by an increase in anxiety- and depressive-like behaviors. Shorter length of EE (3 h) had less impact on these behaviors compared to longer periods (12 and 24 h). These findings highlight the need for caution when employing the Balb/C strain in EE models, particularly in studies exploring emotional behaviors.
Collapse
Affiliation(s)
- Malcon Carneiro de Brito
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil
| | - Beatriz Yamada Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil
| | - Thiago Amorim de Souza Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil
| | - Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil.
| |
Collapse
|
40
|
Hernández-Fuentes GA, Sanchez-Ramirez CA, Cortes-Alvarez SI, Rodriguez-Hernández A, Cabrera-Medina AO, Moy-López NA, Guzman-Muñiz J, Garza-Veloz I, Rodriguez-Sanchez IP, Martinez-Fierro ML, Álvarez-Barajas JJ, Cortes-Alvarez NY, Ceballos-Magaña SG, Meza-Robles C, Delgado-Enciso I. Moringa oleifera Leaf Infusion as a Functional Beverage: Polyphenol Content, Antioxidant Capacity, and Its Potential Role in the Prevention of Metabolopathies. Life (Basel) 2025; 15:636. [PMID: 40283190 PMCID: PMC12028896 DOI: 10.3390/life15040636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Moringa oleifera (MO) leaf infusion has gained attention for its potential therapeutic effects, particularly in metabolic health, due to its rich content of bioactive compounds, including polyphenols. The study evaluates the antioxidant properties and metabolic effects of the prophylactic administration of MO infusion in a high-fat diet (HFD)-induced murine model. First, polyphenol content (0.45 mg/g) and antioxidant activity (45.39%) were determined using Folin-Ciocalteu, DPPH, phosphomolybdenum, ferrocyanide, and anti-browning assays. In the in vivo phase, BALB/c mice were divided into three groups: a balanced diet group, a negative control group, and an HFD group supplemented with MO infusion. Over eight months, biochemical analyses, psychomotor tests, glucose tolerance assessments, and liver histopathology were conducted. MO infusion significantly reduced food intake, weight gain, lipid profiles, and liver inflammation compared to the negative control group, while promoting a metabolic profile similar to that of the balanced diet group. Additionally, it positively influenced psychomotor performance, reinforcing its neuroactive potential. These findings suggest that MO leaf infusion may serve as a functional beverage with protective effects against metabolic disorders, offering a promising natural strategy for managing obesity-related health issues.
Collapse
Affiliation(s)
- Gustavo A. Hernández-Fuentes
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (S.I.C.-A.); (A.R.-H.); (A.O.C.-M.)
- Colima State Institute of Cancerology, IMSS-Bienestar, Colima 28085, Mexico;
- Faculty of Chemical Sciences, University of Colima, Coquimatlan 28400, Mexico;
| | - Carmen A. Sanchez-Ramirez
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (S.I.C.-A.); (A.R.-H.); (A.O.C.-M.)
| | - Salma I. Cortes-Alvarez
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (S.I.C.-A.); (A.R.-H.); (A.O.C.-M.)
- Colima State Institute of Cancerology, IMSS-Bienestar, Colima 28085, Mexico;
| | - Alejandrina Rodriguez-Hernández
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (S.I.C.-A.); (A.R.-H.); (A.O.C.-M.)
| | - Ana O. Cabrera-Medina
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (S.I.C.-A.); (A.R.-H.); (A.O.C.-M.)
| | - Norma A. Moy-López
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, Mexico; (N.A.M.-L.); (J.G.-M.); (N.Y.C.-A.)
| | - Jorge Guzman-Muñiz
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, Mexico; (N.A.M.-L.); (J.G.-M.); (N.Y.C.-A.)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (I.G.-V.); (M.L.M.-F.)
| | - Iram P. Rodriguez-Sanchez
- Molecular and Structural Physiology Laboratory, School of Biological Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico;
| | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (I.G.-V.); (M.L.M.-F.)
| | | | - Nadia Y. Cortes-Alvarez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, Mexico; (N.A.M.-L.); (J.G.-M.); (N.Y.C.-A.)
- Department of Nursing and Midwifery, Division of Natural and Exact Sciences, University of Guanajuato, Guanajuato 36259, Mexico
| | | | - Carmen Meza-Robles
- Colima State Institute of Cancerology, IMSS-Bienestar, Colima 28085, Mexico;
| | - Iván Delgado-Enciso
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (S.I.C.-A.); (A.R.-H.); (A.O.C.-M.)
- Colima State Institute of Cancerology, IMSS-Bienestar, Colima 28085, Mexico;
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
41
|
Kilic A, Ipek BE, Tatonyan S, Kilic K, Demirci H, Atalar F, Ustunova S, Dariyerli N. Alamandine enhanced spatial memory in rats by reducing neuroinflammation and altering BDNF levels in the hippocampus and prefrontal cortex. Sci Rep 2025; 15:12205. [PMID: 40204820 PMCID: PMC11982245 DOI: 10.1038/s41598-025-95683-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Our study aims to determine the effects of alamandine, the newest component of the renin-angiotensin system, on cognitive functions, neuroinflammation, and oxidative stress in the pathophysiology of depression. 35 male Sprague dawley rats, three months old, weighing between 300 and 350 g, were used. The chronic, unpredictable mild stress model of depression was performed. Experimental animals were divided into five groups: control (C), depression (D), alamandine (50 µg/kg, ip) (D + ALA), A779 (300 µg/kg, ip) (D + A779), and both alamandine and A779 treatment groups (D + ALA + A779). After confirming the development of depression through behavioral tests, the animals' learning and memory performances were measured using the Morris water maze test. At the end of the experiment, the animals' prefrontal cortex, hippocampus, and blood samples were isolated for biochemical studies and gene expression analyses. The sucrose preference, open field, elevated plus maze, tail suspension, and forced swimming tests were performed to determine the animals' anxiety levels. There was a significant increase in anxiety-like behaviors in the D group and the A779-treated group, while alamandine exhibited an anxiolytic effect. Moreover, improvements in cognitive skills observed in the Morris water maze test were paralleled by molecular changes, including an increase in BDNF protein levels and NMDA receptor expression and a decrease in GABA levels. In addition, the levels of TNF-α, IL-1β, IL-6, and oxidative stress markers were increased in the depression groups while significantly decreased with alamandine treatment. It was concluded that alamandine has an anxiolytic effect and facilitates spatial memory by reducing neuroinflammation and oxidative stress.
Collapse
Affiliation(s)
- Aysu Kilic
- Department of Physiology, School of Medicine, Bezmialem Vakif University, Fatih, Istanbul, Turkey.
| | - Betul Esra Ipek
- Department of Physiology, School of Medicine, Bezmialem Vakif University, Fatih, Istanbul, Turkey
| | - Suzin Tatonyan
- Department of Immunology, Institute of Graduate Studies in Health Sciences, Istanbul University, Istanbul, Turkey
- Rare Diseases Research Laboratory, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Kubra Kilic
- School of Medicine, Istanbul University, Istanbul, Turkey
| | - Huri Demirci
- Department of Medical Biochemistry, School of Medicine, Biruni University, Istanbul, Turkey
| | - Fatmahan Atalar
- Department of Rare Diseases, Istanbul University, Child Health Institute, Istanbul, Turkey
| | - Savas Ustunova
- Department of Physiology, School of Medicine, Bezmialem Vakif University, Fatih, Istanbul, Turkey
| | - Nuran Dariyerli
- Department of Physiology, School of Medicine, Istanbul University - Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
42
|
Lu OD, White K, Raymond K, Liu C, Klein AS, Green N, Vaillancourt S, Gallagher A, Shindy L, Li A, Wallquist K, Li R, Zou M, Casey AB, Cameron LP, Pomrenze MB, Sohal V, Kheirbek MA, Gomez AM, Lammel S, Heifets BD, Malenka R. A multi-institutional investigation of psilocybin's effects on mouse behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.08.647810. [PMID: 40291657 PMCID: PMC12027077 DOI: 10.1101/2025.04.08.647810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Studies reporting novel therapeutic effects of psychedelic drugs are rapidly emerging. However, the reproducibility and reliability of these findings could remain uncertain for years. Here, we implemented a multi-institutional collaborative approach to define the robust and replicable effects of the psychedelic drug psilocybin on mouse behavior. Five laboratories performed the same experiments to test the acute and persistent effects of psilocybin (2 mg/kg, IP) on various behaviors that psychedelics have been proposed to affect, including anxiety-related approach-avoidance, exploration, sociability, depression-related behaviors, fear extinction, and social reward learning. Through this coordinated approach, we found that psilocybin had several robust and replicable acute effects on mouse behavior, including increased anxiety- and avoidance-related behaviors and decreased fear expression. Surprisingly, however, we found that psilocybin did not have replicable effects 24 hours post psilocybin administration on reducing anxiety- and depression-like behaviors or facilitating fear extinction learning. Additionally, we were unable to observe psilocybin-induced alterations in social preference or social reward learning. Overall, our comprehensive characterization of psilocybin's acute and persistent behavioral effects using ∼200 total male and female mice per experiment spread across five independent labs demonstrates with unique certainty several acute drug effects and suggests that psilocybin's persistent effects in mice may be more modest and inconsistent than previously suggested. We believe this unusual multi-laboratory, highly coordinated research effort serves as a model for facilitating the generation of replicable results and consequently will reduce efforts based on unreliable and spurious results.
Collapse
|
43
|
Kanzaki H, Suzuki S, Tabata T, Suzuki T, Seto Y, Kaneko K. Plant hormone jasmonic acid reduces anxiety behavior in mice. Sci Rep 2025; 15:11424. [PMID: 40181068 PMCID: PMC11968907 DOI: 10.1038/s41598-025-95689-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
Anxiety disorders are a leading cause of disability worldwide and major contributors to the global disease burden. In this study, we investigated the anxiolytic-like effects of plant-derived molecules in mice. Jasmonic acid (JA), a major plant hormone, has been identified as an injury response-related hormone in higher plants. We found that the oral, intraperitoneal, and intraventricular administration of JA in mice demonstrated anxiolytic-like effects in an elevated plus maze test. Additionally, JA exhibited anxiolytic-like effects in mice undergoing open field and novel environment feeding suppression tests. In addition, we found that the anxiolytic-like effects of JA were mediated by serotonin 5-HT1A receptors and central dopamine D1 receptor systems. Our findings reveal a novel role of JA in exerting anxiolytic-like effects in animals and suggest that plant hormones, such as JA, could serve as potential compounds for treating anxiety disorders.
Collapse
Affiliation(s)
- Hanako Kanzaki
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| | - Shiho Suzuki
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| | - Tomotaka Tabata
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| | - Taiki Suzuki
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| | - Yoshiya Seto
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| | - Kentaro Kaneko
- Department of Agricultural Chemistry, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan.
| |
Collapse
|
44
|
Cichon J, Joseph TT, Lu X, Wasilczuk AZ, Kelz MB, Mennerick SJ, Zorumski CF, Nagele P. Nitrous oxide activates layer 5 prefrontal neurons via SK2 channel inhibition for antidepressant effect. Nat Commun 2025; 16:2999. [PMID: 40180931 PMCID: PMC11968965 DOI: 10.1038/s41467-025-57951-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Nitrous oxide (N2O) induces rapid and durable antidepressant effects. The cellular and circuit mechanisms mediating this process are not known. Here we find that a single dose of inhaled N2O induces rapid and specific activation of layer V (L5) pyramidal neurons in the cingulate cortex of rodents exposed to chronic stress conditions. N2O-induced L5 activation rescues a stress-associated hypoactivity state, persists following exposure, and is necessary for its antidepressant-like activity. Although NMDA-receptor antagonism is believed to be a primary mechanism of action for N2O, L5 neurons activate even when NMDA-receptor function is attenuated through both pharmacological and genetic approaches. By examining different molecular and circuit targets, we identify N2O-induced inhibition of calcium-sensitive potassium (SK2) channels as a key molecular interaction responsible for driving specific L5 activity along with ensuing antidepressant-like effects. These results suggest that N2O-induced L5 activation is crucial for its fast antidepressant action and this effect involves novel and specific molecular actions in distinct cortical cell types.
Collapse
Affiliation(s)
- Joseph Cichon
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Thomas T Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrzej Z Wasilczuk
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max B Kelz
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven J Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| |
Collapse
|
45
|
Clunas H, Walpole S, Babic I, Nair M, May N, Huang XF, Solowij N, Newell KA, Weston-Green K. Improved recognition memory and reduced inflammation following β-caryophyllene treatment in the Wistar-Kyoto rodent model of treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111312. [PMID: 40049345 DOI: 10.1016/j.pnpbp.2025.111312] [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: 10/04/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/12/2025]
Abstract
Persistent low mood, anxiety and cognitive deficits are common symptoms of depression and highly efficacious treatments that address symptoms including cognitive dysfunction are still required. β-caryophyllene (BCP) is a terpene with anti-inflammatory and pro-cognitive properties; however, its efficacy on cognition in depression remains unclear. This study aimed to investigate acute and chronic BCP treatment effects on cognitive, depressive- and anxiety-like behaviours, and inflammation in male and female Wistar-Kyoto (WKY) rats, a rodent model of treatment-resistant depression. Rats were administered either BCP (50 mg/kg) or vehicle (control). Open field (OFT), social interaction, sucrose preference, novel object recognition (NOR) and elevated plus maze (EPM) tests were conducted after acute (1 h) and chronic (2 weeks) treatment. Peripheral plasma inflammatory cytokine levels were examined. BCP acutely increased locomotor activity in the OFT but did not improve social interaction, whereas chronic BCP prevented increased latency to first interaction in females (not males). BCP did not improve sucrose preference or prevent anxiety-like behaviours in the EPM. BCP significantly increased novel object discrimination in the NOR test in male and female WKY rats and reduced cytokine levels after chronic treatment. This study shows for the first time that chronic BCP treatment improved recognition memory and exerted anti-inflammatory properties in a rodent model of depressive-like behaviours. BCP did not significantly improve anxiety-like behaviours, social interaction or anhedonia in WKY rats of either sex. These findings demonstrate the pro-cognitive effects of BCP in a rodent model of treatment-resistant depression worthy of further investigation.
Collapse
Affiliation(s)
- Helen Clunas
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia
| | - Samara Walpole
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Ilijana Babic
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Mayank Nair
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Naomi May
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Xu-Feng Huang
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia
| | - Nadia Solowij
- Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia; School of Psychology, Faculty of the Arts, Social Sciences and Humanities, University of Wollongong, NSW 2522, Australia
| | - Kelly A Newell
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia
| | - Katrina Weston-Green
- Molecular Horizons and the School of Medical, Indigenous and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
46
|
Fleming SM, Scott S, Hamad EJ, Herman DE, Holden JG, Yan L, Linning-Duffy K, Kemp CJ, Patterson JR, Miller KM, Kubik M, Kuhn N, Stoll AC, Duffy MF, Steece-Collier K, Cole-Strauss A, Lipton JW, Luk KC, Sortwell CE. Intrastriatal injection of alpha-synuclein preformed fibrils to rats results in L-DOPA reversible sensorimotor impairments and alterations in non-motor function. Front Neurosci 2025; 19:1556447. [PMID: 40236948 PMCID: PMC11996896 DOI: 10.3389/fnins.2025.1556447] [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: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction The alpha-synuclein (α-syn) preformed fibril (PFF) model of Parkinson's disease (PD) is widely used in rodents to understand the mechanisms contributing to progression of pathology and neurodegeneration in the disorder. While the time course of pathology in the α-syn PFF rat model has been well characterized, it has been more challenging to determine reliable and reproducible behavior impairments. This is mainly due to α-syn PFF injections resulting in a partial nigrostriatal lesion that make motor anomalies more subtle and difficult to detect, just as in patients with PD. In the present study we sought to examine the effect of increased striatal distribution and injection quantity of α-syn PFFs in rats on accumulation of phosphorylated α-syn inclusions, nigrostriatal degeneration, sensorimotor behavior, and nonmotor function related to PD. Methods Male Fischer 344 rats were injected unilaterally in the striatum with a total of 24μg α-syn PFFs distributed into three sites, or an equal volume of phosphate buffered saline (PBS) as a control condition. Sensorimotor function was assessed using a battery of behavioral tests sensitive to varying degrees of nigrostriatal neurodegeneration. Non-motor testing included assays for olfaction, emotional reactivity, cognitive function, and sleep. Results At six months post injection, α-syn PFF rats displayed significant movement and somatosensory asymmetries compared with control rats. Time to initiate a forelimb step and time to contact an adhesive stimulus on the forepaw took significantly longer with the contralateral limb compared with the ipsilateral limb in α-syn PFF rats. Further, hindlimb stepping in the cylinder was significantly reduced in α-syn PFF-injected rats compared with controls. Cognitive function was also affected in the α-syn PFF rats, with investigation time significantly decreased in an object recognition test. Levodopa reversibility was observed in the movement initiation and cylinder tests. Postmortem analysis revealed a 55% loss of nigral tyrosine hydroxylase immunoreactive neurons and a 63% reduction in striatal dopamine content in α-syn PFF-injected rats. Conclusion Thus, using the present α-syn PFF surgical parameters, sufficient nigrostriatal degeneration can be achieved to manifest significant motor and non-motor deficits. These rat α-syn PFF surgical parameters will be important for preclinical assessment of novel diseasemodifying therapies.
Collapse
Affiliation(s)
- Sheila M. Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sophia Scott
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Edward J. Hamad
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Danielle E. Herman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - John G. Holden
- Department of Psychology, University of Cincinnati, Cincinnati, OH, United States
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI, United States
| | - Katrina Linning-Duffy
- Department of Psychology, Michigan State University, East Lansing, MI, United States
| | - Christopher J. Kemp
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Joseph R. Patterson
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kathryn M. Miller
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Michael Kubik
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Nathan Kuhn
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Anna C. Stoll
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Megan F. Duffy
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Allyson Cole-Strauss
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Jack W. Lipton
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Kelvin C. Luk
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Caryl E. Sortwell
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| |
Collapse
|
47
|
Hosaini M, Abbasnejad M, Kooshki R, Esmaeili-Mahani S, Raoof M, Naderi R, Aarab G, Lobbezoo F. The involvement of orexin-1 receptors in modulation of feeding and anxiety-like behavior in rats with complete Freund's adjuvant-induced temporomandibular joint disorder. Odontology 2025; 113:764-775. [PMID: 39843662 PMCID: PMC11950102 DOI: 10.1007/s10266-024-01021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 10/29/2024] [Indexed: 01/24/2025]
Abstract
Orexin-A (OXA), a neuropeptide produced in the hypothalamus, is recognized for its role in modulating orofacial nociception and regulating feeding behaviors, as well as its impact on psychophysiological responses. This study investigated the role of orexin-1 receptors (OX1R) in modulating nociceptive behaviors induced by noxious stimulation of the temporomandibular joint (TMJ) and the associated changes in mood and feeding behaviors in rats with complete Freund's adjuvant (CFA)-induced temporomandibular disorders (TMDs). Bilateral cannulation of the lateral ventricles was performed in rats. To induce nociception, CFA was injected unilaterally into the left TMJ of the rats. Nociceptive behaviors were assessed using the hot plate and tail flick tests, while anxiety-like behavior and food intake were evaluated using an elevated plus maze (EPM) and a food preference device, respectively. The results demonstrated a significant increase in nociceptive scores and anxiety-like behaviors, along with reductions in water and food consumption following CFA injection. However, post-treatment with OXA at concentrations of 50 and 100 pM/rat significantly decreased thermal nociceptive scores, alleviated anxiety-like behavior, and increased water and food intake. These beneficial effects were reversed when OXA was co-administered with SB-334867 (40 nM/rat), an OX1R antagonist. Collectively, our findings suggest that OX1R signaling plays a role in the modulation of anxiety-like behavior and abnormalities in food intake in CFA-treated rats. Understanding the involvement of OXA and its receptors in CFA-induced TMJ nociception and behavioral changes may pave the way for potential therapeutic interventions targeting OX1R signaling in the management of TMD-associated symptoms.
Collapse
Affiliation(s)
- Mojtaba Hosaini
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Razieh Kooshki
- Department of Biology, Faculty of Sciences, Lorestan University, Khorramabad, Iran
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Maryam Raoof
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Reyhaneh Naderi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Ghizlane Aarab
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frank Lobbezoo
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Orofacial Pain and Jaw Function, Faculty of Odontology, Malmö University, Malmö, Sweden
| |
Collapse
|
48
|
Stickling CP, Rosenkranz JA. Effects of repeated social stress on risk assessment behaviors and response to diazepam in the elevated plus maze in adult male rats. Behav Neurosci 2025; 139:60-73. [PMID: 39621391 PMCID: PMC11966652 DOI: 10.1037/bne0000612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Anxiety is highly common, and stress is a major trigger for anxiety. Anxiety includes heightened threat assessment and avoidance, but we do not fully understand which components are sensitive to stress. Rodents show a balance of exploration and avoidance that incorporates threat assessment prior to making the relatively risky decision to explore an open area. The purpose of this study was to determine if stress impacts risk assessment and if this is tied to the effects of stress on exploration. The present study used elevated plus maze (EPM) to test the effects of repeated social defeat stress (RSDS) on risk assessment behaviors in adult male rats. We then tested the effects of diazepam, an anxiolytic that reduces the impact of stress on EPM exploration, to further clarify the relationship between risk assessment and risky behavior in the EPM. We found that RSDS decreased time in the open arm, similar to prior studies. We also found that RSDS increased the likelihood of the primary risk assessment behavior, stretch and attend posture (SAP), increased SAP prior to entering an open arm, and decreased the likelihood that a rat would enter an open arm after SAP. Diazepam ameliorated the effects of RSDS on both SAP and exploratory behavior, further linking risk assessment and subsequent exploratory behaviors. These results suggest that increased risk assessment and reduced risky choices after risk assessment are tied to effects of stress on exploration and provide novel insight into how stress may increase avoidance by effects on risk assessment. (PsycInfo Database Record (c) 2025 APA, all rights reserved).
Collapse
Affiliation(s)
- Courtney P. Stickling
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - J. Amiel Rosenkranz
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
- Discipline of Cellular and Molecular Pharmacology, Department of Foundational Sciences and Humanities, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| |
Collapse
|
49
|
Shoae-Hagh P, Razavi BM, Sadeghnia HR, Mehri S, Karimi G, Hosseinzadeh H. Molecular and Behavioral Neuroprotective Effects of Clavulanic Acid and Crocin in Haloperidol-Induced Tardive Dyskinesia in Rats. Mol Neurobiol 2025; 62:5156-5182. [PMID: 39520654 DOI: 10.1007/s12035-024-04566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Clavulanic acid (ClvA), a beta-lactamase inhibitor, is being explored for its significant neuroprotective potential. The effects of ClvA were assessed both individually and in combination with crocin (Cr), an antioxidant derived from saffron, in the context of tardive dyskinesia (TD). In rat haloperidol (Hp)-induced-TD (1 mg/kg, i.p. 21 days), the effects of ClvA (50, 100, 150 mg/kg) and Cr (10, 20, 40 mg/kg) were assessed via vacuous chewing movements (VCM) and tongue protrusion (TP). Striatal malondialdehyde (MDA) and glutathione (GSH) were measured spectrophotometrically. Based on the results, ClvA (100 mg/kg) and Cr (10 mg/kg) were determined with sub-effective doses. Glutamate transporter-subtype1 (GLT1), dopamine active transporter (DAT), vesicular monoamine transporter-type2 (VMAT2), Bax/Bcl2, cleaved Caspase3, phosphorylated AKT/AKT, IL1β, and TNFα levels were quantified using western blotting in sub-effective doses and their combination. The behavioral results of catalepsy and orofacial dyskinesia demonstrated model establishment. Hp decreased GLT1 (p < 0.05), DAT (p < 0.01), VMAT2 (p < 0.001), GSH and pAKT/AKT (p < 0.0001); increased TNFα (p < 0.05), IL1β, cleaved Caspase3 (p < 0.001); MDA and Bax/Bcl2 (p < 0.0001). ClvA 100 mg/kg reversed the decreased GLT1 and VMAT2 (p < 0.01), alongside the increased MDA (p < 0.0001) and VCM (p < 0.05). It also increased AKT phosphorylation (p < 0.05). No effects were noted on DAT, GSH, Bax/Bcl2, or inflammatory factors. However, the combination with Cr at 10 mg/kg influenced ClvA on DAT (p < 0.01) and resulted in a significant increase in GSH (p < 0.0001). Additionally, there was a marked decrease in TNFα (p < 0.0001) and IL1β (p < 0.001), enhancing its effects on reducing MDA and increasing pAKT/AKT (p < 0.0001). The combination adversely affected GLT1. ClvA protects against TD via GLT1 and VMAT2; combined with Cr, it enhances antioxidant effects, improves DAT, and requires dose optimization for GLT1 disruption.
Collapse
Affiliation(s)
- Parisa Shoae-Hagh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hamid Reza Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
50
|
An L, Xiong Y, Yang Y, Lyu D, Yang Z, Zhang T. Re-socialization reduces social isolation-induced high alcohol preference and anxiety via possibly restoring dopamine-rewarding effects in the rat striatum. Pharmacol Biochem Behav 2025; 249:173981. [PMID: 39993506 DOI: 10.1016/j.pbb.2025.173981] [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: 10/11/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Social environmental factors frequently play an important role in early-life. It is reported that isolation increases vulnerability to develop alcohol use disorder. We investigated the effects of re-socialization on high alcohol preference and anxiety behaviors, induced by early-life social isolation (SI), and its possible underlying mechanism in male Wistar rats. On the 21st postnatal day, animals were either housed in groups of (CON) or isolated (SI-1) for the first stage (3 weeks). Afterwards, the SI-1 group were divided into two groups: re-socialization with socially housed rats (Re-SH) and isolation (SI-2) for a second stage (3 weeks). Both alcohol preference and behaviour tests were performed in these two stages. The ratio of dopamine content in striatum tissue was measured. The results showed that SI considerably induced the high alcohol preference and increased anxiety-like behaviors. However, during the 2nd stage, peer companionship significantly reduced the high alcohol preference and anxiety-like behaviors which were induced by early-life SI. Moreover, the striatal dopamine content was significantly enhanced by SI, but was evidently suppressed by re-socialization. Additionally, there was no statistical difference in body weight, anxiety-like behaviour, alcohol preference or dopamine content when the rats were only isolated during the SI-2 stage. It suggests that both the high alcohol preference and anxiety-like behaviors are able to be significantly reduced by re-socialization, which is possibly associated with regulating dopamine concentration.
Collapse
Affiliation(s)
- Lei An
- Department of Proctology, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Yuxiu Xiong
- College of Life Sciences, Nankai University, 300071 Tianjin, China
| | - Yang Yang
- Department of Neonatology, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Dan Lyu
- Department of Pain Management, Nankai University Affiliated Tianjin First Central Hospital, 300071 Tianjin, China
| | - Zhuo Yang
- College of Life Sciences, Nankai University, 300071 Tianjin, China
| | - Tao Zhang
- College of Life Sciences, Nankai University, 300071 Tianjin, China.
| |
Collapse
|