1
|
Tanabe M, Kunisawa K, Saito I, Kosuge A, Tezuka H, Kawai T, Kon Y, Yoshidomi K, Kagami A, Hasegawa M, Kubota H, Ojika H, Fujii T, Tochio T, Hirooka Y, Saito K, Nabeshima T, Mouri A. Adolescent social isolation decreases colonic goblet cells and impairs spatial cognition through the reduction of cystine. Mol Psychiatry 2025; 30:2137-2151. [PMID: 39613916 PMCID: PMC12014494 DOI: 10.1038/s41380-024-02826-9] [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: 04/22/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
Negative experiences during adolescence, such as social isolation (SI), bullying, and abuse, increase the risk of psychiatric diseases in adulthood. However, the pathogenesis of psychiatric diseases induced by these factors remain poorly understood. In adolescents, stress affects the intestinal homeostasis in the gut-brain axis. This study determined whether adolescent SI induces behavioral abnormalities by disrupting colonic function. Adolescent mice exposed to SI exhibit spatial cognitive deficits and microglial activation in the hippocampus (HIP). SI decreased the differentiation of mucin-producing goblet cells, which was accompanied by alterations in the composition of the gut microbiota, particularly the depletion of mucin-feeding bacteria. Treatment with rebamipide, which promotes goblet cell differentiation in the colon, attenuated SI-induced spatial cognitive deficits and microglial activation in the HIP and decreased cystine, a downstream metabolite of homocysteine. Treatment with cystine ameliorated SI-induced spatial cognitive deficits and increased microglial C-C motif chemokine ligand 7 (CCL7) levels in the HIP. Inhibition of CCL7 receptors by antagonists of CC motif chemokine receptors 2 (CCR2) and 3 (CCR3) in the HIP prevented spatial cognitive deficits induced by SI. Infusion of CCL7 into the HIP following microglial ablation with clodronate liposome induced spatial cognitive deficits. These findings suggest that adolescent SI decreases serum cystine levels by damaging the colonic goblet cells, resulting in spatial cognitive deficits by triggering microglial activation in the HIP. Our results indicate that increased CCL7 expression in hippocampal microglia may contribute to spatial cognitive deficits by activating CCR2 and CCR3.
Collapse
Affiliation(s)
- Moeka Tanabe
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan.
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan.
| | - Imari Saito
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Aika Kosuge
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hiroyuki Tezuka
- Department of Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Tomoki Kawai
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yuki Kon
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Koyo Yoshidomi
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Akari Kagami
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
| | - Haruto Ojika
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Tadashi Fujii
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Takumi Tochio
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiki Hirooka
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Kuniaki Saito
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research (J-DO), Nagoya, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan.
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan.
- Japanese Drug Organization of Appropriate Use and Research (J-DO), Nagoya, Aichi, Japan.
| |
Collapse
|
2
|
Tanabe M, Kunisawa K, Saito I, Ojika H, Saito K, Nabeshima T, Mouri A. High-Cellulose Diet Ameliorates Cognitive Impairment by Modulating Gut Microbiota and Metabolic Pathways in Mice. J Nutr 2025:S0022-3166(25)00187-7. [PMID: 40216297 DOI: 10.1016/j.tjnut.2025.04.004] [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: 12/13/2024] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Nutrition is a key factor in cognitive function, and safe dietary interventions are promising to prevent cognitive impairment in pediatric psychiatric disorders. We previously demonstrated that childhood social isolation (SI) stress affects colonic function, leading to cognitive impairment. Cellulose, an insoluble dietary fiber, shows benefits to intestinal health, but its potential impact on cognitive impairment has not been explored. OBJECTIVES This study investigated whether a high-cellulose diet ameliorates cognitive impairment induced by SI through modulation of gut microbiota and metabolic pathways. METHODS C57BL/6J male mice (3 wk old; n = 10-15/group) were randomly divided into 2 groups: individually housed (SI) group and housed 5 mice per cage (group-housed) group. Each group received either a normal diet (5% cellulose) or a high-cellulose diet (30% cellulose) for 5 wk daily until the end of the behavioral testing. We evaluated behavior abnormalities, gut microbiota composition, and metabolites, and performed 2-way analysis of variance. RESULTS Intake of a high-cellulose diet ameliorated cognitive impairment, including decreased time spent in a novel location of SI mice in novel object location test (NOLT; +30%; P < 0.01) with reduction of Iba-1 positive cells, microglia, in the hippocampus (-33%; P < 0.05). The high-cellulose diet indicated a significant difference in gut microbiota clustering plots (P < 0.01) and enhanced the variation in malate-aspartate shuttle pathways in SI mice (P < 0.01). Notably, fecal microbiota transplantation (FMT) from SI mice fed a high-cellulose diet after antibiotic treatment, replicated amelioration of cognitive impairment in NOLT (+46%; P < 0.01). Additionally, the FMT replicated a decrease of Iba-1 positive cells indicating suppressed hippocampal microglial activation (-52%; P < 0.01), and enhanced the variation in malate-aspartate shuttle pathways (P < 0.01). CONCLUSIONS These findings suggest that a high-cellulose diet may ameliorate pediatric-specific cognitive impairment through modulation of the gut microbiota and metabolic pathways.
Collapse
Affiliation(s)
- Moeka Tanabe
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan; Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan; International Center for Brain Science, Fujita Health University, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan.
| | - Imari Saito
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Haruto Ojika
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Kuniaki Saito
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Aichi, Japan; Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Aichi, Japan; International Center for Brain Science, Fujita Health University, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan; International Center for Brain Science, Fujita Health University, Aichi, Japan; Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan.
| |
Collapse
|
3
|
Ali A, Milman S, Weiss EF, Gao T, Napolioni V, Barzilai N, Zhang ZD, Lin J. Genetic variants associated with age-related episodic memory decline implicate distinct memory pathologies. Alzheimers Dement 2025; 21:e14379. [PMID: 39559945 PMCID: PMC11775541 DOI: 10.1002/alz.14379] [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: 05/10/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Approximately 40% of people aged ≥ 65 experience memory loss, particularly in episodic memory. Identifying the genetic basis of episodic memory decline is crucial for uncovering its underlying causes. METHODS We investigated common and rare genetic variants associated with episodic memory decline in 742 (632 for rare variants) Ashkenazi Jewish individuals (mean age 75) from the LonGenity study. All-atom molecular dynamics simulations were performed to uncover mechanistic insights underlying rare variants associated with episodic memory decline. RESULTS In addition to the common polygenic risk of Alzheimer's disease, we identified and replicated rare variant associations in ITSN1 and CRHR2. Structural analyses revealed distinct memory pathologies mediated by interfacial rare coding variants such as impaired receptor activation of corticotropin releasing hormone and dysregulated L-serine synthesis. DISCUSSION Our study uncovers novel risk loci for episodic memory decline. The identified underlying mechanisms point toward heterogenous memory pathologies mediated by rare coding variants. HIGHLIGHTS We demonstrated the contribution of the common polygenic risk of Alzheimer's disease to episodic memory decline. We discovered and replicated two risk genes associated with episodic memory decline implicated by rare variants, were discovered and replicated. We demonstrated molecular mechanisms and potential novel memory pathologies underlying interfacial rare coding variants. Molecular dynamics simulations were performed to understand the downstream effects of risk rare coding variants.
Collapse
Affiliation(s)
- Amanat Ali
- Department of MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Sofiya Milman
- Department of MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
- Department of GeneticsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Erica F. Weiss
- Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Tina Gao
- Department of MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Valerio Napolioni
- School of Biosciences and Veterinary MedicineUniversity of CamerinoCamerinoItaly
| | - Nir Barzilai
- Department of MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
- Department of GeneticsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Zhengdong D. Zhang
- Department of GeneticsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jhih‐Rong Lin
- Department of GeneticsAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
4
|
Zoubovsky SP, Muglia LJ. Transplacental signals involved in the programming effects of prenatal psychosocial stress on neurodevelopment. Neurotoxicol Teratol 2025; 107:107424. [PMID: 39755178 DOI: 10.1016/j.ntt.2025.107424] [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: 06/30/2024] [Revised: 12/03/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Exposure to psychosocial stress during pregnancy has been associated with the emergence of neurodevelopmental and neuropsychiatric disorders in offspring. The placenta is known to orchestrate various functions that are essential for normal fetal development, including the brain. It has therefore been postulated that alterations in such functions, and downstream signaling, have the potential to dramatically affect brain developmental trajectories and contribute to adverse neurodevelopmental outcomes. This review will focus on discussing various placental functions that have been proposed to be affected by exposure to prenatal psychosocial stress and the implications of such disruptions on long-term neurodevelopmental programming.
Collapse
Affiliation(s)
- Sandra P Zoubovsky
- Department of Pediatrics, University of Colorado, Denver, CO, United States
| | - Louis J Muglia
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Office of the President, Burroughs Wellcome Fund, Research Triangle Park, Durham, NC, United States.
| |
Collapse
|
5
|
Gupta P, Galimberti M, Liu Y, Beck S, Wingo A, Wingo T, Adhikari K, Kranzler HR, Stein MB, Gelernter J, Levey DF. A genome-wide investigation into the underlying genetic architecture of personality traits and overlap with psychopathology. Nat Hum Behav 2024; 8:2235-2249. [PMID: 39134740 PMCID: PMC11576509 DOI: 10.1038/s41562-024-01951-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/09/2024] [Indexed: 08/21/2024]
Abstract
Personality is influenced by both genetic and environmental factors and is associated with other psychiatric traits such as anxiety and depression. The 'big five' personality traits, which include neuroticism, extraversion, agreeableness, conscientiousness and openness, are a widely accepted and influential framework for understanding and describing human personality. Of the big five personality traits, neuroticism has most often been the focus of genetic studies and is linked to various mental illnesses, including depression, anxiety and schizophrenia. Our knowledge of the genetic architecture of the other four personality traits is more limited. Here, utilizing the Million Veteran Program cohort, we conducted a genome-wide association study in individuals of European and African ancestry. Adding other published data, we performed genome-wide association study meta-analysis for each of the five personality traits with sample sizes ranging from 237,390 to 682,688. We identified 208, 14, 3, 2 and 7 independent genome-wide significant loci associated with neuroticism, extraversion, agreeableness, conscientiousness and openness, respectively. These findings represent 62 novel loci for neuroticism, as well as the first genome-wide significant loci discovered for agreeableness. Gene-based association testing revealed 254 genes showing significant association with at least one of the five personality traits. Transcriptome-wide and proteome-wide analysis identified altered expression of genes and proteins such as CRHR1, SLC12A5, MAPT and STX4. Pathway enrichment and drug perturbation analyses identified complex biology underlying human personality traits. We also studied the inter-relationship of personality traits with 1,437 other traits in a phenome-wide genetic correlation analysis, identifying new associations. Mendelian randomization showed positive bidirectional effects between neuroticism and depression and anxiety, while a negative bidirectional effect was observed for agreeableness and these psychiatric traits. This study improves our comprehensive understanding of the genetic architecture underlying personality traits and their relationship to other complex human traits.
Collapse
Affiliation(s)
- Priya Gupta
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Marco Galimberti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Yue Liu
- Department of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sarah Beck
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Aliza Wingo
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA
| | - Thomas Wingo
- Department of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Keyrun Adhikari
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Henry R Kranzler
- Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Murray B Stein
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA, USA
- Departments of Psychiatry, School of Medicine, and Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, CA, USA
| | - Joel Gelernter
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Daniel F Levey
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA.
| |
Collapse
|
6
|
Joshi A, Giorgi FM, Sanna PP. Transcriptional Patterns in Stages of Alzheimer's Disease Are Cell-Type-Specific and Partially Converge with the Effects of Alcohol Use Disorder in Humans. eNeuro 2024; 11:ENEURO.0118-24.2024. [PMID: 39299805 PMCID: PMC11485264 DOI: 10.1523/eneuro.0118-24.2024] [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: 02/28/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
Advances in single-cell technologies have led to the discovery and characterization of new brain cell types, which in turn lead to a better understanding of the pathogenesis of Alzheimer's disease (AD). Here, we present a detailed analysis of single-nucleus (sn)RNA-seq data for three stages of AD from middle temporal gyrus and compare it with snRNA-seq data from the prefrontal cortices from individuals with alcohol use disorder (AUD). We observed a significant decrease in both inhibitory and excitatory neurons, in general agreement with previous reports. We observed several cell-type-specific gene expressions and pathway dysregulations that delineate AD stages. Endothelial and vascular leptomeningeal cells showed the greatest degree of gene expression changes. Cell-type-specific evidence of neurodegeneration was seen in multiple neuronal cell types particularly in somatostatin and Layer 5 extratelencephalic neurons, among others. Evidence of inflammatory responses was seen in non-neuronal cells, particularly in intermediate and advanced AD. We observed common perturbations in AD and AUD, particularly in pathways, like transcription, translation, apoptosis, autophagy, calcium signaling, neuroinflammation, and phosphorylation, that imply shared transcriptional pathogenic mechanisms and support the role of excessive alcohol intake in AD progression. Major AUD gene markers form and perturb a network of genes significantly associated with intermediate and advanced AD. Master regulator analysis from AUD gene markers revealed significant correlation with advanced AD of transcription factors that have implications in intellectual disability, neuroinflammation, and other neurodegenerative conditions, further suggesting a shared nexus of transcriptional changes between AD and AUD.
Collapse
Affiliation(s)
- Arpita Joshi
- The Scripps Research Institute, San Diego, California 92117
| | - Federico Manuel Giorgi
- The Scripps Research Institute, San Diego, California 92117
- University of Bologna, Bologna 40136, Italy
| | | |
Collapse
|
7
|
González-Gil A, Sánchez-Maldonado B, Rojo C, Flor-García M, Queiroga FL, Ovalle S, Ramos-Ruiz R, Fuertes-Recuero M, Picazo RA. Proneurogenic actions of follicle-stimulating hormone on neurospheres derived from ovarian cortical cells in vitro. BMC Vet Res 2024; 20:372. [PMID: 39160565 PMCID: PMC11334536 DOI: 10.1186/s12917-024-04203-8] [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: 01/26/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Neural stem and progenitor cells (NSPCs) from extra-neural origin represent a valuable tool for autologous cell therapy and research in neurogenesis. Identification of proneurogenic biomolecules on NSPCs would improve the success of cell therapies for neurodegenerative diseases. Preliminary data suggested that follicle-stimulating hormone (FSH) might act in this fashion. This study was aimed to elucidate whether FSH promotes development, self-renewal, and is proneurogenic on neurospheres (NS) derived from sheep ovarian cortical cells (OCCs). Two culture strategies were carried out: (a) long-term, 21-days NS culture (control vs. FSH group) with NS morphometric evaluation, gene expression analyses of stemness and lineage markers, and immunolocalization of NSPCs antigens; (b) NS assay to demonstrate FSH actions on self-renewal and differentiation capacity of NS cultured with one of three defined media: M1: positive control with EGF/FGF2; M2: control; and M3: M2 supplemented with FSH. RESULTS In long-term cultures, FSH increased NS diameters with respect to control group (302.90 ± 25.20 μm vs. 183.20 ± 7.63 on day 9, respectively), upregulated nestin (days 15/21), Sox2 (day 21) and Pax6 (days 15/21) and increased the percentages of cells immunolocalizing these proteins. During NS assays, FSH stimulated NSCPs proliferation, and self-renewal, increasing NS diameters during the two expansion periods and the expression of the neuron precursor transcript DCX during the second one. In the FSH-group there were more frequent cell-bridges among neighbouring NS. CONCLUSIONS FSH is a proneurogenic hormone that promotes OCC-NSPCs self-renewal and NS development. Future studies will be necessary to support the proneurogenic actions of FSH and its potential use in basic and applied research related to cell therapy.
Collapse
Affiliation(s)
- Alfredo González-Gil
- Department of Physiology, School of Veterinary Medicine, Complutense University of Madrid, Avda. Puerta de Hierro SN, Madrid, 28040, Spain.
| | - Belén Sánchez-Maldonado
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, Complutense University of Madrid, Madrid, 28040, Spain
| | - Concepción Rojo
- Department of Anatomy and Embriology, School of Veterinary Medicine, University Complutense of Madrid, Madrid, 28040, Spain
| | - Miguel Flor-García
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Spanish Research Council (CSIC)-Universidad Autónoma de Madrid, Madrid, 28049, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, 28049, Spain
| | - Felisbina Luisa Queiroga
- Centre for the Study of Animal Science, CECA-ICETA, University of Porto, Porto, Portugal.
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Quinta dos Prados, Vila Real, 5000-801, Portugal.
| | - Susana Ovalle
- Genomic Unit Cantoblanco, Fundación Parque Científico de Madrid. C/ Faraday 7, Madrid, 28049, Spain
| | - Ricardo Ramos-Ruiz
- Genomic Unit Cantoblanco, Fundación Parque Científico de Madrid. C/ Faraday 7, Madrid, 28049, Spain
| | - Manuel Fuertes-Recuero
- Department of Physiology, School of Veterinary Medicine, Complutense University of Madrid, Avda. Puerta de Hierro SN, Madrid, 28040, Spain
| | - Rosa Ana Picazo
- Department of Physiology, School of Veterinary Medicine, Complutense University of Madrid, Avda. Puerta de Hierro SN, Madrid, 28040, Spain
| |
Collapse
|
8
|
Chiani F, Mastrorilli V, Marchetti N, Macioce A, Nappi C, Strimpakos G, Pasquini M, Gambadoro A, Battistini JI, Cutuli D, Petrosini L, Marinelli S, Scardigli R, Farioli Vecchioli S. Essential role of p21 Waf1/Cip1 in the modulation of post-traumatic hippocampal Neural Stem Cells response. Stem Cell Res Ther 2024; 15:197. [PMID: 38971774 PMCID: PMC11227726 DOI: 10.1186/s13287-024-03787-0] [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: 10/24/2023] [Accepted: 06/07/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Traumatic Brain Injury (TBI) represents one of the main causes of brain damage in young people and the elderly population with a very high rate of psycho-physical disability and death. TBI is characterized by extensive cell death, tissue damage and neuro-inflammation with a symptomatology that varies depending on the severity of the trauma from memory loss to a state of irreversible coma and death. Recently, preclinical studies on mouse models have demonstrated that the post-traumatic adult Neural Stem/Progenitor cells response could represent an excellent model to shed light on the neuro-reparative role of adult neurogenesis following damage. The cyclin-dependent kinase inhibitor p21Waf1/Cip1 plays a pivotal role in modulating the quiescence/activation balance of adult Neural Stem Cells (aNSCs) and in restraining the proliferation progression of progenitor cells. Based on these considerations, the aim of this work is to evaluate how the conditional ablation of p21Waf1/Cip1 in the aNSCS can alter the adult hippocampal neurogenesis in physiological and post-traumatic conditions. METHODS We designed a novel conditional p21Waf1/Cip1 knock-out mouse model, in which the deletion of p21Waf1/Cip1 (referred as p21) is temporally controlled and occurs in Nestin-positive aNSCs, following administration of Tamoxifen. This mouse model (referred as p21 cKO mice) was subjected to Controlled Cortical Impact to analyze how the deletion of p21 could influence the post-traumatic neurogenic response within the hippocampal niche. RESULTS The data demonstrates that the conditional deletion of p21 in the aNSCs induces a strong increase in activation of aNSCs as well as proliferation and differentiation of neural progenitors in the adult dentate gyrus of the hippocampus, resulting in an enhancement of neurogenesis and the hippocampal-dependent working memory. However, following traumatic brain injury, the increased neurogenic response of aNSCs in p21 cKO mice leads to a fast depletion of the aNSCs pool, followed by declined neurogenesis and impaired hippocampal functionality. CONCLUSIONS These data demonstrate for the first time a fundamental role of p21 in modulating the post-traumatic hippocampal neurogenic response, by the regulation of the proliferative and differentiative steps of aNSCs/progenitor populations after brain damage.
Collapse
Affiliation(s)
- Francesco Chiani
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Monterotondo, Rome, Italy
| | | | - Nicole Marchetti
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Monterotondo, Rome, Italy
- PhD Course in Sciences of Nutrition, Aging, Metabolism and Gender Pathologies, Catholic University of Roma, 00100, Rome, Italy
| | - Andrea Macioce
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Monterotondo, Rome, Italy
| | - Chiara Nappi
- Instituto de Neurosciencias, Universidad Miguel-Hernandez, Alicante, Spain
| | - Georgios Strimpakos
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Monterotondo, Rome, Italy
| | - Miriam Pasquini
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Monterotondo, Rome, Italy
| | - Alessia Gambadoro
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Monterotondo, Rome, Italy
| | | | - Debora Cutuli
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185, Rome, Italy
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179, Rome, Italy
| | - Laura Petrosini
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179, Rome, Italy
| | - Sara Marinelli
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Monterotondo, Rome, Italy
| | - Raffaella Scardigli
- European Brain Research Institute (EBRI), Viale Regine Elena, 00161, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | | |
Collapse
|
9
|
Nakos Bimpos M, Karali K, Antoniou C, Palermos D, Fouka M, Delis A, Tzieras I, Chrousos GP, Koutmani Y, Stefanis L, Polissidis A. Alpha-synuclein-induced stress sensitivity renders the Parkinson's disease brain susceptible to neurodegeneration. Acta Neuropathol Commun 2024; 12:100. [PMID: 38886854 PMCID: PMC11181569 DOI: 10.1186/s40478-024-01797-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/12/2024] [Indexed: 06/20/2024] Open
Abstract
A link between chronic stress and Parkinson's disease (PD) pathogenesis is emerging. Ample evidence demonstrates that the presynaptic neuronal protein alpha-synuclein (asyn) is closely tied to PD pathogenesis. However, it is not known whether stress system dysfunction is present in PD, if asyn is involved, and if, together, they contribute to neurodegeneration. To address these questions, we assess stress axis function in transgenic rats overexpressing full-length wildtype human asyn (asyn BAC rats) and perform multi-level stress and PD phenotyping following chronic corticosterone administration. Stress signaling, namely corticotropin-releasing factor, glucocorticoid and mineralocorticoid receptor gene expression, is also examined in post-mortem PD patient brains. Overexpression of human wildtype asyn leads to HPA axis dysregulation in rats, while chronic corticosterone administration significantly aggravates nigrostriatal degeneration, serine129 phosphorylated asyn (pS129) expression and neuroinflammation, leading to phenoconversion from a prodromal to an overt motor PD phenotype. Interestingly, chronic corticosterone in asyn BAC rats induces a robust, twofold increase in pS129 expression in the hypothalamus, the master regulator of the stress response, while the hippocampus, both a regulator and a target of the stress response, also demonstrates elevated pS129 asyn levels and altered markers of stress signalling. Finally, defective hippocampal stress signalling is mirrored in human PD brains and correlates with asyn expression levels. Taken together, our results link brain stress system dysregulation with asyn and provide evidence that elevated circulating glucocorticoids can contribute to asyn-induced neurodegeneration, ultimately triggering phenoconversion from prodromal to overt PD.
Collapse
Affiliation(s)
- Modestos Nakos Bimpos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Katerina Karali
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Christine Antoniou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Dionysios Palermos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Maria Fouka
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Anastasios Delis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Iason Tzieras
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - George Panagiotis Chrousos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- University Research Institute on Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Yassemi Koutmani
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- 1St Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece.
- Department of Science and Mathematics, ACG-Research Center, Deree - American College of Greece, 15342, Athens, Greece.
| |
Collapse
|
10
|
Ali A, Milman S, Weiss EF, Gao T, Napolioni V, Barzilai N, Zhang ZD, Lin JR. Rare genetic coding variants associated with age-related episodic memory decline implicate distinct memory pathologies in the hippocampus. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.21.24307692. [PMID: 38826255 PMCID: PMC11142267 DOI: 10.1101/2024.05.21.24307692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Approximately 40% of people aged 65 or older experience memory loss, particularly in episodic memory. Identifying the genetic basis of episodic memory decline is crucial for uncovering its underlying causes. Methods We investigated common and rare genetic variants associated with episodic memory decline in 742 (632 for rare variants) Ashkenazi Jewish individuals (mean age 75) from the LonGenity study. All-atom MD simulations were performed to uncover mechanistic insights underlying rare variants associated with episodic memory decline. Results In addition to the common polygenic risk of Alzheimer's Disease (AD), we identified and replicated rare variant association in ITSN1 and CRHR2 . Structural analyses revealed distinct memory pathologies mediated by interfacial rare coding variants such as impaired receptor activation of corticotropin releasing hormone and dysregulated L-serine synthesis. Discussion Our study uncovers novel risk loci for episodic memory decline. The identified underlying mechanisms point toward heterogeneous memory pathologies mediated by rare coding variants.
Collapse
|
11
|
Gupta P, Galimberti M, Liu Y, Beck S, Wingo A, Wingo T, Adhikari K, Stein MB, Gelernter J, Levey DF. A genome-wide investigation into the underlying genetic architecture of personality traits and overlap with psychopathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.17.24301428. [PMID: 38293137 PMCID: PMC10827244 DOI: 10.1101/2024.01.17.24301428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Personality is influenced by both genetic and environmental factors and is associated with other psychiatric traits such as anxiety and depression. The "Big Five" personality traits, which include neuroticism, extraversion, agreeableness, conscientiousness, and openness, are a widely accepted and influential framework for understanding and describing human personality. Of the big five personality traits, neuroticism has most often been the focus of genetic studies and is linked to various mental illnesses including depression, anxiety, and schizophrenia. Our knowledge of the genetic architecture of the other four personality traits is more limited. Utilizing the Million Veteran Program (MVP) cohort we conducted a genome-wide association study (GWAS) in individuals of European and African ancestry. Adding other published data, we performed GWAS meta-analysis for each of the five personality traits with sample sizes ranging from 237,390 to 682,688. We identified 158, 14, 3, 2, and 7 independent genome-wide significant (GWS) loci associated with neuroticism, extraversion, agreeableness, conscientiousness, and openness, respectively. These findings represent 55 novel loci for neuroticism, as well as the first GWS loci discovered for extraversion and agreeableness. Gene-based association testing revealed 254 genes showing significant association with at least one of the five personality traits. Transcriptome-wide and proteome-wide analysis identified altered expression of genes and proteins such as CRHR1, SLC12A5, MAPT, and STX4. Pathway enrichment and drug perturbation analyses identified complex biology underlying human personality traits. We also studied the inter-relationship of personality traits with 1,437 other traits in a phenome-wide genetic correlation analysis, identifying new associations. Mendelian randomization showed positive bidirectional effects between neuroticism and depression and anxiety while a negative bidirectional effect was observed for agreeableness and these psychiatric traits. This study improves our comprehensive understanding of the genetic architecture underlying personality traits and their relationship to other complex human traits.
Collapse
Affiliation(s)
- Priya Gupta
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Marco Galimberti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Yue Liu
- Department of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, USA
| | - Sarah Beck
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Aliza Wingo
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, USA
- Atlanta Veterans Affairs Medical Center, USA
| | - Thomas Wingo
- Department of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, USA
| | - Keyrun Adhikari
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Murray B Stein
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA
- Departments of Psychiatry, School of Medicine, and Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, CA
| | - Joel Gelernter
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Daniel F Levey
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| |
Collapse
|
12
|
Bhuiyan P, Sun Z, Chen Y, Qian Y. Peripheral surgery triggers mast cells activation: Focusing on neuroinflammation. Behav Brain Res 2023; 452:114593. [PMID: 37499912 DOI: 10.1016/j.bbr.2023.114593] [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: 03/14/2023] [Revised: 06/12/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Peripheral surgery can lead to a systemic aseptic inflammatory response comprising several mediators aiming at restoring tissue homeostasis. It induces inflammatory mechanisms through neuroimmune interaction between the periphery and to brain which also plays a critical role in causing cognitive impairments. Accumulating scientific evidence revealed that acute neuroinflammation of the brain triggered by peripheral surgery that causes peripheral inflammation leads to transmitting signals into the brain through immune cells. Mast cells (MCs) play an important role in the acute neuroinflammation induced by peripheral surgical trauma. After peripheral surgery, brain-resident MCs can be rapidly activated followed by releasing histamine, tryptase, and other inflammatory mediators. These mediators then interact with other immune cells in the peripheral and amplify the signal into the brain by disrupting BBB and activating principle innate immune cells of brain including microglia, astrocytes, and vascular endothelial cells, which release abundant inflammatory mediators and in turn accelerate the activation of brain MCs, amplify the cascade effect of neuroinflammatory response. Surgical stress may induce HPA axis activation by releasing corticotropin-releasing hormone (CRH) subsequently influence the activation of brain MCs, thus resulting in impaired synaptic plasticity. Herein, we discuss the better understating of MCs mediated neuroinflammation mechanisms after peripheral surgery and potential therapeutic targets for controlling inflammatory cascades.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| | - Zhaochu Sun
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| | - Yinan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China.
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
13
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
14
|
Nikolopoulos D, Manolakou T, Polissidis A, Filia A, Bertsias G, Koutmani Y, Boumpas DT. Microglia activation in the presence of intact blood-brain barrier and disruption of hippocampal neurogenesis via IL-6 and IL-18 mediate early diffuse neuropsychiatric lupus. Ann Rheum Dis 2023; 82:646-657. [PMID: 36898766 PMCID: PMC10176423 DOI: 10.1136/ard-2022-223506] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/26/2023] [Indexed: 03/12/2023]
Abstract
INTRODUCTION Inflammatory mediators are detected in the cerebrospinal fluid of systemic lupus erythematosus patients with central nervous system involvement (NPSLE), yet the underlying cellular and molecular mechanisms leading to neuropsychiatric disease remain elusive. METHODS We performed a comprehensive phenotyping of NZB/W-F1 lupus-prone mice including tests for depression, anxiety and cognition. Immunofluorescence, flow cytometry, RNA-sequencing, qPCR, cytokine quantification and blood-brain barrier (BBB) permeability assays were applied in hippocampal tissue obtained in both prenephritic (3-month-old) and nephritic (6-month-old) lupus mice and matched control strains. Healthy adult hippocampal neural stem cells (hiNSCs) were exposed ex vivo to exogenous inflammatory cytokines to assess their effects on proliferation and apoptosis. RESULTS At the prenephritic stage, BBB is intact yet mice exhibit hippocampus-related behavioural deficits recapitulating the human diffuse neuropsychiatric disease. This phenotype is accounted by disrupted hippocampal neurogenesis with hiNSCs exhibiting increased proliferation combined with decreased differentiation and increased apoptosis in combination with microglia activation and increased secretion of proinflammatory cytokines and chemokines. Among these cytokines, IL-6 and IL-18 directly induce apoptosis of adult hiNSCs ex vivo. During the nephritic stage, BBB becomes disrupted which facilitates immune components of peripheral blood, particularly B-cells, to penetrate into the hippocampus further augmenting inflammation with locally increased levels of IL-6, IL-12, IL-18 and IL-23. Of note, an interferon gene signature was observed only at nephritic-stage. CONCLUSION An intact BBB with microglial activation disrupting the formation of new neurons within the hippocampus represent early events in NPSLE. Disturbances of the BBB and interferon signature are evident later in the course of the disease.
Collapse
Affiliation(s)
- Dionysis Nikolopoulos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece .,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Manolakou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Anastasia Filia
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - George Bertsias
- Laboratory of Autoimmunity-Inflammation, Institute of Molecular Biology and Biotechnology, Heraklion, Greece.,Rheumatology, Clinical Immunology and Allergy Department, Medical School University of Crete, Heraklion, Greece
| | | | - Dimitrios T Boumpas
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece .,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Medical School, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
15
|
Kwon M, Lee JH, Yoon Y, Pleasure SJ, Yoon K. The CRHR1/CREB/REST signaling cascade regulates mammalian embryonic neural stem cell properties. EMBO Rep 2023; 24:e55313. [PMID: 36413000 PMCID: PMC9900344 DOI: 10.15252/embr.202255313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Growing evidence suggests that the corticotropin-releasing hormone (CRH) signaling pathway, mainly known as a critical initiator of humoral stress responses, has a role in normal neuronal physiology. However, despite the evidence of CRH receptor (CRHR) expression in the embryonic ventricular zone, the exact functions of CRH signaling in embryonic brain development have not yet been fully determined. In this study, we show that CRHR1 is required for the maintenance of neural stem cell properties, as assessed by in vitro neurosphere assays and cell distribution in the embryonic cortical layers following in utero electroporation. Identifying the underlying molecular mechanisms of CRHR1 action, we find that CRHR1 functions are accomplished through the increasing expression of the master transcription factor REST. Furthermore, luciferase reporter and chromatin immunoprecipitation assays reveal that CRHR1-induced CREB activity is responsible for increased REST expression at the transcriptional level. Taken together, these findings indicate that the CRHR1/CREB/REST signaling cascade plays an important role downstream of CRH in the regulation of neural stem cells during embryonic brain development.
Collapse
Affiliation(s)
- Mookwang Kwon
- Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwonSouth Korea
| | - Ju Hyun Lee
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| | - Youngik Yoon
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| | - Samuel J Pleasure
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Keejung Yoon
- Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwonSouth Korea
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| |
Collapse
|
16
|
Butkevich IP, Mikhailenko VA, Vershinina EA. Sexual Dimorphism in the Effect of Neonatal Inflammatory Pain on Stress Reactivity of Hormonal Response and Cognitive Functions in Adult Rats. J EVOL BIOCHEM PHYS+ 2022; 58:353-363. [PMID: 35599637 PMCID: PMC9109674 DOI: 10.1134/s0022093022020053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 11/23/2022]
Abstract
The effect of moderate neonatal stress induced by inflammatory
pain in rat pups of both sexes on the hormonal response and cognitive
processes in adult animals was studied in the Morris water maze.
No significant differences in spatial learning and memory were found
in experimental rats exposed to neonatal inflammatory pain vs. control
animals. However, experimental rats exhibited sex differences in
long-term spatial memory whose efficiency was higher in males vs.
females. After long-term memory testing, stress responsiveness of
the hypothalamic-pituitary-adrenocortical axis, as assessed by the
plasma corticosterone level in the formalin test, was higher in
experimental males vs. females. Only experimental females exhibited
differences between short-term and long-term memory, with the efficiency
being higher in the former. Thus, sexual dimorphism was found in
the effect of neonatal nociceptive stress on long-term spatial memory
in adult rats: experimental males vs. females demonstrated more
effective long-term memory combined with a higher stress reactivity
of the hormonal response.
Collapse
Affiliation(s)
- I. P. Butkevich
- Pavlov Institute of Physiology,
Russian Academy of Sciences, St. Petersburg, Russia
| | - V. A. Mikhailenko
- Pavlov Institute of Physiology,
Russian Academy of Sciences, St. Petersburg, Russia
| | - E. A. Vershinina
- Pavlov Institute of Physiology,
Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
17
|
Roque A, Valles Méndez KM, Ruiz R, Pineda E, Lajud N. Early life stress induces a transient increase in hippocampal corticotropin-releasing hormone in rat neonates that precedes the effects on hypothalamic neuropeptides. Eur J Neurosci 2022; 55:2108-2121. [PMID: 33745155 DOI: 10.1111/ejn.15193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
Early life stress (ELS) programs hypothalamus-pituitary-adrenal (HPA) axis activity and affects synaptic plasticity and cognitive performance in adults; however, the effects of ELS during the temporal window of vulnerability are poorly understood. This study aimed to thoroughly characterize the effects of ELS in the form of periodic maternal separation (MS180) during the time of exposure to stress. Hippocampal corticotropin-releasing hormone (CRH) gene expression and baseline HPA axis activity were analyzed at postnatal (P) days 6, 12, 15, and 21, and in adulthood (P75); these factors were correlated with plasticity markers and adult behavior. Our results indicate that MS180 induces an increase in hippocampal CRH expression at P9, P12, and P15, whereas an increase in hypothalamic CRH expression was observed from P12 to P21. Increased arginine-vasopressin expression and corticosterone levels were observed only at P21. Moreover, MS180 caused transient alterations in hypothalamic synaptophysin expression during early life. As adults, MS180 rats showed a passive coping strategy in the forced swimming test, cognitive impairments in the object location test, increased hypothalamic CRH expression, and decreased oxytocin (OXT) expression. Spearman's analysis indicated that cognitive impairments correlated with CRH and OXT expression. In conclusion, our data indicate that MS180 induces a transient increase in hippocampal CRH expression in neonates that precedes the effects on hypothalamic neuropeptides, confirming the role of increased CRH during the temporal window of vulnerability as a mediator of some of the detrimental effects of ELS on brain development and adult behavior.
Collapse
Affiliation(s)
- Angélica Roque
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Kinberli Marcela Valles Méndez
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Roberto Ruiz
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Edel Pineda
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| | - Naima Lajud
- Laboratorio de Neurobiología del Desarrollo, División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, México
| |
Collapse
|
18
|
Kassotaki I, Valsamakis G, Mastorakos G, Grammatopoulos DK. Placental CRH as a Signal of Pregnancy Adversity and Impact on Fetal Neurodevelopment. Front Endocrinol (Lausanne) 2021; 12:714214. [PMID: 34408727 PMCID: PMC8366286 DOI: 10.3389/fendo.2021.714214] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 11/13/2022] Open
Abstract
Early life is a period of considerable plasticity and vulnerability and insults during that period can disrupt the homeostatic equilibrium of the developing organism, resulting in adverse developmental programming and enhanced susceptibility to disease. Fetal exposure to prenatal stress can impede optimum brain development and deranged mother's hypothalamic-pituitary-adrenal axis (HPA axis) stress responses can alter the neurodevelopmental trajectories of the offspring. Corticotropin-releasing hormone (CRH) and glucocorticoids, regulate fetal neurogenesis and while CRH exerts neuroprotective actions, increased levels of stress hormones have been associated with fetal brain structural alterations such as reduced cortical volume, impoverishment of neuronal density in the limbic brain areas and alterations in neuronal circuitry, synaptic plasticity, neurotransmission and G-protein coupled receptor (GPCR) signalling. Emerging evidence highlight the role of epigenetic changes in fetal brain programming, as stress-induced methylation of genes encoding molecules that are implicated in HPA axis and major neurodevelopmental processes. These serve as molecular memories and have been associated with long term modifications of the offspring's stress regulatory system and increased susceptibility to psychosomatic disorders later in life. This review summarises our current understanding on the roles of CRH and other mediators of stress responses on fetal neurodevelopment.
Collapse
Affiliation(s)
- Ifigeneia Kassotaki
- Department of Internal Medicine, 2nd Internal Medicine Clinic, Venizeleio Pananeio General Hospital, Heraklion, Greece
| | - Georgios Valsamakis
- Second University Department of Obs and Gynae, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Translational Medicine, Warwick Medical School, Coventry, United Kingdom
| | - George Mastorakos
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris K. Grammatopoulos
- Translational Medicine, Warwick Medical School, Coventry, United Kingdom
- Institute of Precision Diagnostics and Translational Medicine, Pathology, University Hospitals Coventry and Warwickshire (UHCW) NHS Trust, Coventry, United Kingdom
| |
Collapse
|
19
|
Butkevich IP, Mikhailenko VA, Vershinina EA, Barr GA. The Long-Term Effects of Neonatal Inflammatory Pain on Cognitive Function and Stress Hormones Depend on the Heterogeneity of the Adolescent Period of Development in Male and Female Rats. Front Behav Neurosci 2021; 15:691578. [PMID: 34366805 PMCID: PMC8334561 DOI: 10.3389/fnbeh.2021.691578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/21/2021] [Indexed: 11/21/2022] Open
Abstract
Exposure to stress at an early age programs the HPA axis which can lead to cognitive deficits in adults. However, it is not known whether these deficits emerge in adulthood or are expressed earlier in life. The aims of the study were to investigate (1) the immediate effects of early injury-induced stress in one-day-old (P1) and repeated stress on at P1 and P2 rat pups on plasma corticosterone levels; and (2) examine the subsequent long-term effects of this early stress on spatial learning and memory, and stress reactivity in early P26-34 and late P45-53 adolescent male and female rats. Intra-plantar injection of formalin induced prolonged and elevated levels of corticosterone in pups and impaired spatial learning and short- and long-term memory in late adolescent males and long-term memory in early adolescent females. There were sex differences in late adolescence in both learning and short-term memory. Performance on the long-term memory task was better than that on the short-term memory task for all early adolescent male and female control and stressed animals. Short-term memory was better in the late age control rats of both sexes and for formalin treated females as compared with the early age rats. These results are consistent with an impaired function of structures involved in memory (the hippocampus, amygdala, prefrontal cortex) after newborn pain. However, activation of the HPA axis by neonatal pain did not directly correlate with spatial learning and memory outcomes and the consequences of neonatal pain remain are likely multi-determined.
Collapse
Affiliation(s)
- Irina P. Butkevich
- Laboratory of Ontogenesis of the Nervous System, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Viktor A. Mikhailenko
- Laboratory of Ontogenesis of the Nervous System, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Elena A. Vershinina
- Department of Information Technologies and Mathematical Modeling, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Gordon A. Barr
- Department of Anesthesiology and Critical Care Medicine, The Children’s Hospital of Philadelphia and the Perelman School of Medicine, Philadelphia, PA, United States
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
20
|
Neurogenic and Neuroprotective Potential of Stem/Stromal Cells Derived from Adipose Tissue. Cells 2021; 10:cells10061475. [PMID: 34208414 PMCID: PMC8231154 DOI: 10.3390/cells10061475] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
Currently, the number of stem-cell based experimental therapies in neurological injuries and neurodegenerative disorders has been massively increasing. Despite the fact that we still have not obtained strong evidence of mesenchymal stem/stromal cells’ neurogenic effectiveness in vivo, research may need to focus on more appropriate sources that result in more therapeutically promising cell populations. In this study, we used dedifferentiated fat cells (DFAT) that are proven to demonstrate more pluripotent abilities in comparison with standard adipose stromal cells (ASCs). We used the ceiling culture method to establish DFAT cells and to optimize culture conditions with the use of a physioxic environment (5% O2). We also performed neural differentiation tests and assessed the neurogenic and neuroprotective capability of both DFAT cells and ASCs. Our results show that DFAT cells may have a better ability to differentiate into oligodendrocytes, astrocytes, and neuron-like cells, both in culture supplemented with N21 and in co-culture with oxygen–glucose-deprived (OGD) hippocampal organotypic slice culture (OHC) in comparison with ASCs. Results also show that DFAT cells have a different secretory profile than ASCs after contact with injured tissue. In conclusion, DFAT cells constitute a distinct subpopulation and may be an alternative source in cell therapy for the treatment of nervous system disorders.
Collapse
|
21
|
Weir K, Kim DW, Blackshaw S. A potential role for somatostatin signaling in regulating retinal neurogenesis. Sci Rep 2021; 11:10962. [PMID: 34040115 PMCID: PMC8155210 DOI: 10.1038/s41598-021-90554-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Neuropeptides have been reported to regulate progenitor proliferation and neurogenesis in the central nervous system. However, these studies have typically been conducted using pharmacological agents in ex vivo preparations, and in vivo evidence for their developmental function is generally lacking. Recent scRNA-Seq studies have identified multiple neuropeptides and their receptors as being selectively expressed in neurogenic progenitors of the embryonic mouse and human retina. This includes Sstr2, whose ligand somatostatin is transiently expressed by immature retinal ganglion cells. By analyzing retinal explants treated with selective ligands that target these receptors, we found that Sstr2-dependent somatostatin signaling induces a modest, dose-dependent inhibition of photoreceptor generation, while correspondingly increasing the relative fraction of primary progenitor cells. These effects were confirmed by scRNA-Seq analysis of retinal explants but abolished in Sstr2-deficient retinas. Although no changes in the relative fraction of primary progenitors or photoreceptor precursors were observed in Sstr2-deficient retinas in vivo, scRNA-Seq analysis demonstrated accelerated differentiation of neurogenic progenitors. We conclude that, while Sstr2 signaling may act to negatively regulate retinal neurogenesis in combination with other retinal ganglion cell-derived secreted factors such as Shh, it is dispensable for normal retinal development.
Collapse
Affiliation(s)
- Kurt Weir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
22
|
Myospryn deficiency leads to impaired cardiac structure and function and schizophrenia-associated symptoms. Cell Tissue Res 2021; 385:675-696. [PMID: 34037836 DOI: 10.1007/s00441-021-03447-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/02/2021] [Indexed: 12/25/2022]
Abstract
The desmin-associated protein myospryn, encoded by the cardiomyopathy-associated gene 5 (CMYA5), is a TRIM-like protein associated to the BLOC-1 (Biogenesis of Lysosomes Related Organelles Complex 1) protein dysbindin. Human myospryn mutations are linked to both cardiomyopathy and schizophrenia; however, there is no evidence of a direct causative link of myospryn to these diseases. Therefore, we sought to unveil the role of myospryn in heart and brain. We have genetically inactivated the myospryn gene by homologous recombination and demonstrated that myospryn null hearts have dilated phenotype and compromised cardiac function. Ultrastructural analyses revealed that the sarcomere organization is not obviously affected; however, intercalated disk (ID) integrity is impaired, along with mislocalization of ID and sarcoplasmic reticulum (SR) protein components. Importantly, cardiac and skeletal muscles of myospryn null mice have severe mitochondrial defects with abnormal internal vacuoles and extensive cristolysis. In addition, swollen SR and T-tubules often accompany the mitochondrial defects, strongly implying a potential link of myospryn together with desmin to SR- mitochondrial physical and functional cross-talk. Furthermore, given the reported link of human myospryn mutations to schizophrenia, we performed behavioral studies, which demonstrated that myospryn-deficient male mice display disrupted startle reactivity and prepulse inhibition, asocial behavior, decreased exploratory behavior, and anhedonia. Brain neurochemical and ultrastructural analyses revealed prefrontal-striatal monoaminergic neurotransmitter defects and ultrastructural degenerative aberrations in cerebellar cytoarchitecture, respectively, in myospryn-deficient mice. In conclusion, myospryn is essential for both cardiac and brain structure and function and its deficiency leads to cardiomyopathy and schizophrenia-associated symptoms.
Collapse
|
23
|
Poulia N, Delis F, Brakatselos C, Polissidis A, Koutmani Y, Kokras N, Dalla C, Politis PK, Antoniou K. Detrimental effects of adolescent escalating low-dose Δ 9 -tetrahydrocannabinol leads to a specific bio-behavioural profile in adult male rats. Br J Pharmacol 2021; 178:1722-1736. [PMID: 33496341 DOI: 10.1111/bph.15394] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/27/2020] [Accepted: 01/18/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Adolescent cannabis use is associated with adult psychopathology. When Δ9 -tetrahydrocannabinol (THC), mainly in high doses, is administered to adolescence rats there are also long lasting effects in adults. This study aims to determine the specific adult bio-behavioural profile after adolescent low-dose THC, which better mirrors adolescent recreational cannabis use. EXPERIMENTAL APPROACH Adolescent male Sprague-Dawley rats were treated with escalating low-dose of THC. In adulthood, they were evaluated for their spontaneous locomotion, sensorimotor gating, higher order and spatial cognitive functions. Dopaminergic activity and cannabinoid receptor expression were measured in distinct brain regions. Hippocampal neurogenic activity of neural stem cells was determined and protein levels of neuroplasticity-related biomarkers were quantified. Adolescent low-dose THC exposure increased spontaneous open-field activity, without affecting prepulse inhibition and attentional set-shifting performance. Region-specific dopaminergic alterations and CB1 receptor up-regulation in the prefrontal cortex were observed. Impaired spatial memory, as assessed with the object location task and Morris water maze test, was associated with significantly decreased proliferative activity (SOX2-positive cells), neurogenic potential (decreased doublecortin-positive cells) in the adult hippocampus and defective neuroplasticity, including reduced BDNF expression in the hippocampus and prefrontal cortex. KEY RESULTS Our findings reveal the adverse impact of adolescent low-dose THC on the psychomotor profile, dopaminergic neurotransmission, compensatory cannabinoid receptor response, cognition-related neurobiological and behavioural functions. CONCLUSION AND IMPLICATIONS Our adolescent low-dose THC animal model does not induce tangible psychotic-like effects, such as those reported in high-dose THC studies, but it impairs cognitive functions and points to hippocampal vulnerability and disrupted neurogenesis.
Collapse
Affiliation(s)
- Nafsika Poulia
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Foteini Delis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Charalampos Brakatselos
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Yassemi Koutmani
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Nikolaos Kokras
- Department of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,First Department of Psychiatry, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis K Politis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Katerina Antoniou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| |
Collapse
|