1
|
Akande AO, Carter ZA, Stokes KY, Nam HW. Endothelial Neurogranin Regulates Blood-Brain Barrier Permeability via Modulation of the AKT Pathway. Mol Neurobiol 2025; 62:3991-4007. [PMID: 39367201 PMCID: PMC11880131 DOI: 10.1007/s12035-024-04522-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024]
Abstract
Neurogranin (Ng) expression is a biomarker for Alzheimer's disease. A loss of brain Ng and an increase in CSF Ng positively correlate with cognitive decline. Ng is known to regulate neuronal calcium-calmodulin binding and synaptic plasticity, which are critical for learning/memory. Interestingly, we discovered that Ng is also expressed in mouse and human blood-brain barrier (BBB). However, the role of Ng expression in brain vasculature remains largely undefined. In this study, we investigated the role of Ng expression on neurovascular structure and function using Ng null mice and human cerebral microvascular endothelial (hCMEC/D3) cells. We performed brain clearing and immunolabeling of blood vessels from whole brains and brain slices. Deletion of Ng significantly decreases neurovascular density in mice. Using in vivo permeability assays, we found increased neurovascular permeability in Ng null mice. We also observed significant changes in the expression of tight junction proteins using western blot and immunofluorescent staining. To identify the molecular pathways involved, we carried out label-free proteomics on brain lysates from endothelial-specific Ng knockout mice. Ingenuity Pathway Analysis indicated that the AKT pathway is attenuated in the vasculature of endothelial-specific Ng knockout mice. To validate these in vivo findings, we pharmacologically manipulated AKT signaling in hCMEC/D3 cells and observed that inhibition of AKT activation causes increased permeability. Our results indicate that the loss of Ng expression alters neurovascular structure and permeability, potentially contributing to neurological dysfunction. Therefore, modulating Ng expression in the BBB may offer a novel therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Adesewa O Akande
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - Zachary A Carter
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - Karen Y Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA.
| |
Collapse
|
2
|
Zheng J, Wang H, Wu W, Wang L, Qin M, Zhu L, Liu Z, Chen Y, Yu Y. Role of FPR2 antagonism in alleviating social isolation-induced depression and protecting blood-brain barrier integrity. J Neuroinflammation 2025; 22:79. [PMID: 40083006 PMCID: PMC11907847 DOI: 10.1186/s12974-025-03408-4] [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: 10/21/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Social isolation (SI) is a prevalent issue in modern society, particularly exacerbated during the COVID-19 pandemic, and it is a significant contributor to depressive disorders. Inflammation-related markers are upregulated in patients with major depressive disorder (MDD) unresponsive to first-line selective serotonin reuptake inhibitor (SSRI) antidepressants. This study investigates the role of formyl peptide receptor 2 (FPR2), a G-protein coupled receptor expressed in central and peripheral immune cells, in SI-induced depression. We developed a mouse model of SI by housing mice individually for three weeks. SI mice exhibited increased capillary-associated microglia (CAMs) with upregulated FPR2 expression in the prefrontal cortex (PFC) and hippocampus compared to group-housed controls. Notably, subcutaneous administration of the FPR2 antagonist WRW4 alleviated depressive and anxiety-like behaviors in SI mice, reducing microglial activation and neuronal damage. WRW4 treatment decreased CAM numbers and their FPR2 expression. RNA sequencing revealed that SI primarily induced changes in genes associated with blood-brain barrier (BBB) function, followed by alterations in genes related to hormone activity, immune activation, and neuronal function. Transcriptomic changes in brain endothelial cells from SI mice resembled those observed in animal models of several neurological disorders and in MDD patients. WRW4 treatment partially reversed these transcriptomic alterations and restored compromised BBB integrity. Additionally, intracerebroventricular (ICV) injection of WRW4 also alleviated depressive and anxiety-like behaviors in SI mice. Finally, our analysis of public transcriptome databases indicates FPR2 upregulation in the orbital ventral PFC of MDD patients and peripheral blood mononuclear cells of those in severe depressive episodes. These findings suggest that the pharmacological targeting of FPR2 may rescue SI-induced pathology in mice by protecting BBB integrity.
Collapse
Affiliation(s)
- Jiayi Zheng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hanqi Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanning Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Linlin Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Meizhen Qin
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, China
| | - Lingfeng Zhu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhen Liu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yijun Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
3
|
She K, Yuan N, Huang M, Zhu W, Tang M, Ma Q, Chen J. Emerging role of microglia in the developing dopaminergic system: perturbation by early life stress. Neural Regen Res 2024; 21:01300535-990000000-00587. [PMID: 39589170 PMCID: PMC12094535 DOI: 10.4103/nrr.nrr-d-24-00742] [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: 08/06/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Early life stress correlates with a higher prevalence of neurological disorders, including autism, attention-deficit/hyperactivity disorder, schizophrenia, depression, and Parkinson's disease. These conditions, primarily involving abnormal development and damage of the dopaminergic system, pose significant public health challenges. Microglia, as the primary immune cells in the brain, are crucial in regulating neuronal circuit development and survival. From the embryonic stage to adulthood, microglia exhibit stage-specific gene expression profiles, transcriptome characteristics, and functional phenotypes, enhancing the susceptibility to early life stress. However, the role of microglia in mediating dopaminergic system disorders under early life stress conditions remains poorly understood. This review presents an up-to-date overview of preclinical studies elucidating the impact of early life stress on microglia, leading to dopaminergic system disorders, along with the underlying mechanisms and therapeutic potential for neurodegenerative and neurodevelopmental conditions. Impaired microglial activity damages dopaminergic neurons by diminishing neurotrophic support (e.g., insulin-like growth factor-1) and hinders dopaminergic axon growth through defective phagocytosis and synaptic pruning. Furthermore, blunted microglial immunoreactivity suppresses striatal dopaminergic circuit development and reduces neuronal transmission. Furthermore, inflammation and oxidative stress induced by activated microglia can directly damage dopaminergic neurons, inhibiting dopamine synthesis, reuptake, and receptor activity. Enhanced microglial phagocytosis inhibits dopamine axon extension. These long-lasting effects of microglial perturbations may be driven by early life stress-induced epigenetic reprogramming of microglia. Indirectly, early life stress may influence microglial function through various pathways, such as astrocytic activation, the hypothalamic-pituitary-adrenal axis, the gut-brain axis, and maternal immune signaling. Finally, various therapeutic strategies and molecular mechanisms for targeting microglia to restore the dopaminergic system were summarized and discussed. These strategies include classical antidepressants and antipsychotics, antibiotics and anti-inflammatory agents, and herbal-derived medicine. Further investigations combining pharmacological interventions and genetic strategies are essential to elucidate the causal role of microglial phenotypic and functional perturbations in the dopaminergic system disrupted by early life stress.
Collapse
Affiliation(s)
- Kaijie She
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Naijun Yuan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- Shenzhen People’s Hospital, The 2 Clinical Medical College, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Shenzhen, Guangdong Province, China
| | - Minyi Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Wenjun Zhu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Manshi Tang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Farinha-Ferreira M, Magalhães DM, Neuparth-Sottomayor M, Rafael H, Miranda-Lourenço C, Sebastião AM. Unmoving and uninflamed: Characterizing neuroinflammatory dysfunction in the Wistar-Kyoto rat model of depression. J Neurochem 2024; 168:2443-2460. [PMID: 38430009 DOI: 10.1111/jnc.16083] [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: 10/25/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Reductionistic research on depressive disorders has been hampered by the limitations of animal models. Recently, it has been hypothesized that neuroinflammation is a key player in depressive disorders. The Wistar-Kyoto (WKY) rat is an often-used animal model of depression, but no information so far exists on its neuroinflammatory profile. As such, we compared male young adult WKY rats to Wistar (WS) controls, with regard to both behavioral performance and brain levels of key neuroinflammatory markers. We first assessed anxiety- and depression-like behaviors in a battery consisting of the Elevated Plus Maze (EPM), the Novelty Suppressed Feeding (NSFT), Open Field (OFT), Social Interaction (SIT), Forced Swim (FST), Sucrose Preference (SPT), and Splash tests (ST). We found that WKY rats displayed increased NSFT feeding latency, decreased OFT center zone permanence, decreased EPM open arm permanence, decreased SIT interaction time, and increased immobility in the FST. However, WKY rats also evidenced marked hypolocomotion, which is likely to confound performance in such tests. Interestingly, WKY rats performed similarly, or even above, to WS levels in the SPT and ST, in which altered locomotion is not a significant confound. In a separate cohort, we assessed prefrontal cortex (PFC), hippocampus and amygdala levels of markers of astrocytic (GFAP, S100A10) and microglial (Iba1, CD86, Ym1) activation status, as well as of three key proinflammatory cytokines (IL-1β, IL-6, TNF-α). There were no significant differences between strains in any of these markers, in any of the regions assessed. Overall, results highlight that behavioral data obtained with WKY rats as a model of depression must be carefully interpreted, considering the marked locomotor activity deficits displayed. Furthermore, our data suggest that, despite WKY rats replicating many depression-associated neurobiological alterations, as shown by others, this is not the case for neuroinflammation-related alterations, thus representing a novel limitation of this model.
Collapse
Affiliation(s)
- Miguel Farinha-Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mariana Neuparth-Sottomayor
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Hugo Rafael
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
5
|
Adkins AM, Luyo ZNM, Gibbs AJ, Boden AF, Heerbrandt RS, Gotthold JD, Britten RA, Wellman LL, Sanford LD. Alterations in Blood-Brain Barrier Integrity and Lateral Ventricle Differ in Rats Exposed to Space Radiation and Social Isolation. Life (Basel) 2024; 14:636. [PMID: 38792656 PMCID: PMC11122575 DOI: 10.3390/life14050636] [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: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The proposed Mars missions will expose astronauts to long durations of social isolation (SI) and space radiation (SR). These stressors have been shown to alter the brain's macrostructure and microenvironment, including the blood-brain barrier (BBB). Breakdown of the BBB is linked to impaired executive functions and physical deficits, including sensorimotor and neurocognitive impairments. However, the precise mechanisms mediating these effects remain unknown. Additionally, the synergistic effects of combined exposure to SI and SR on the structural integrity of the BBB and brain remain unknown. We assessed the BBB integrity and morphology in the brains of male rats exposed to ground-based analogs of SI and SR. The rats exposed to SR had enlarged lateral ventricles and increased BBB damage associated with a loss of astrocytes and an increased number of leaky vessels. Many deficits observed in SR-treated animals were attenuated by dual exposure to SI (DFS). SI alone did not show BBB damage but did show differences in astrocyte morphology compared to the Controls. Thus, determining how single and combined inflight stressors modulate CNS structural integrity is crucial to fully understand the multiple pathways that could impact astronaut performance and health, including the alterations to the CNS structures and cell viability observed in this study.
Collapse
Affiliation(s)
- Austin M. Adkins
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Zachary N. M. Luyo
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Alayna J. Gibbs
- Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
| | - Alea F. Boden
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Riley S. Heerbrandt
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Justin D. Gotthold
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Richard A. Britten
- Center for Integrative Neuroscience and Inflammatory Diseases, Radiation Oncology, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
| | - Laurie L. Wellman
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| | - Larry D. Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (A.M.A.); (Z.N.M.L.); (A.F.B.); (R.S.H.); (J.D.G.); (L.L.W.)
| |
Collapse
|
6
|
Aburto MR, Cryan JF. Gastrointestinal and brain barriers: unlocking gates of communication across the microbiota-gut-brain axis. Nat Rev Gastroenterol Hepatol 2024; 21:222-247. [PMID: 38355758 DOI: 10.1038/s41575-023-00890-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 02/16/2024]
Abstract
Crosstalk between gut and brain has long been appreciated in health and disease, and the gut microbiota is a key player in communication between these two distant organs. Yet, the mechanisms through which the microbiota influences development and function of the gut-brain axis remain largely unknown. Barriers present in the gut and brain are specialized cellular interfaces that maintain strict homeostasis of different compartments across this axis. These barriers include the gut epithelial barrier, the blood-brain barrier and the blood-cerebrospinal fluid barrier. Barriers are ideally positioned to receive and communicate gut microbial signals constituting a gateway for gut-microbiota-brain communication. In this Review, we focus on how modulation of these barriers by the gut microbiota can constitute an important channel of communication across the gut-brain axis. Moreover, barrier malfunction upon alterations in gut microbial composition could form the basis of various conditions, including often comorbid neurological and gastrointestinal disorders. Thus, we should focus on unravelling the molecular and cellular basis of this communication and move from simplistic framing as 'leaky gut'. A mechanistic understanding of gut microbiota modulation of barriers, especially during critical windows of development, could be key to understanding the aetiology of gastrointestinal and neurological disorders.
Collapse
Affiliation(s)
- María R Aburto
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
7
|
López-Otín C, Kroemer G. The missing hallmark of health: psychosocial adaptation. Cell Stress 2024; 8:21-50. [PMID: 38476764 PMCID: PMC10928495 DOI: 10.15698/cst2024.03.294] [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: 01/11/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
The eight biological hallmarks of health that we initially postulated (Cell. 2021 Jan 7;184(1):33-63) include features of spatial compartmentalization (integrity of barriers, containment of local perturbations), maintenance of homeostasis over time (recycling & turnover, integration of circuitries, rhythmic oscillations) and an array of adequate responses to stress (homeostatic resilience, hormetic regulation, repair & regeneration). These hallmarks affect all eight somatic strata of the human body (molecules, organelles, cells, supracellular units, organs, organ systems, systemic circuitries and meta-organism). Here we postulate that mental and socioeconomic factors must be added to this 8×8 matrix as an additional hallmark of health ("psychosocial adaptation") and as an additional stratum ("psychosocial interactions"), hence building a 9×9 matrix. Potentially, perturbation of each of the somatic hallmarks and strata affects psychosocial factors and vice versa. Finally, we discuss the (patho)physiological bases of these interactions and their implications for mental health improvement.
Collapse
Affiliation(s)
- Carlos López-Otín
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
8
|
Magalhães DM, Mampay M, Sebastião AM, Sheridan GK, Valente CA. Age-related impact of social isolation in mice: Young vs middle-aged. Neurochem Int 2024; 174:105678. [PMID: 38266657 DOI: 10.1016/j.neuint.2024.105678] [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: 10/04/2023] [Revised: 01/14/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Social isolation is a chronic mild stressor and a significant risk factor for mental health disorders. Herein we explored the impact of social isolation on depression- and anxiety-like behaviours, as well as spatial memory impairments, in middle-aged male mice compared to post-weaning mice. We aimed to quantify and correlate social isolation-induced behaviour discrepancies with changes in hippocampal glial cell reactivity and pro-inflammatory cytokine levels. Post-weaning and middle-aged C57BL7/J6 male mice were socially isolated for a 3-week period and behavioural tests were performed on the last five days of isolation. We found that 3 weeks of social isolation led to depressive-like behaviour in the forced swim test, anxiety-like behaviour in the open field test, and spatial memory impairment in the Morris water maze paradigm in middle-aged male mice. These behavioural alterations were not observed in male mice after post-weaning social isolation, indicating resilience to isolation-mediated stress. Increased Iba-1 expression and NLRP3 priming were both observed in the hippocampus of socially isolated middle-aged mice, suggesting a role for microglia and NLRP3 pathway in the detrimental effects of social isolation on cognition and behaviour. Young socially isolated mice also demonstrated elevated NLRP3 priming compared to controls, but no differences in Iba-1 levels and no significant changes in behaviour. Ageing-induced microglia activation and enhancement of IL-1β, TNF-α and IL-6 proinflammatory cytokines, known signs of a chronic low-grade inflammatory state, were also detected. Altogether, data suggest that social isolation, in addition to inflammaging, contributes to stress-related cognitive impairment in middle-aged mice.
Collapse
Affiliation(s)
- Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; School of Applied Sciences, University of Brighton, Brighton, UK
| | - Myrthe Mampay
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | | | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
9
|
Mohd Sahini SN, Mohd Nor Hazalin NA, Srikumar BN, Jayasingh Chellammal HS, Surindar Singh GK. Environmental enrichment improves cognitive function, learning, memory and anxiety-related behaviours in rodent models of dementia: Implications for future study. Neurobiol Learn Mem 2024; 208:107880. [PMID: 38103676 DOI: 10.1016/j.nlm.2023.107880] [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: 04/25/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Environmental enrichment (EE) is a process of brain stimulation by modifying the surroundings, for example, by changing the sensory, social, or physical conditions. Rodents have been used in such experimental strategies through exposure to diverse physical, social, and exploration conditions. The present study conducted an extensive analysis of the existing literature surrounding the impact of EE on dementia rodent models. The review emphasised the two principal aspects that are very closely related to dementia: cognitive function (learning and memory) as well as psychological factors (anxiety-related behaviours such as phobias and unrealistic worries). Also highlighted were the mechanisms involved in the rodent models of dementia showing EE effects. Two search engines, PubMed and Science Direct, were used for data collection using the following keywords: environmental enrichment, dementia, rodent model, cognitive performance, and anxiety-related behaviour. Fifty-five articles were chosen depending on the criteria for inclusion and exclusion. The rodent models with dementia demonstrated improved learning and memory in the form of hampered inflammatory responses, enhanced neuronal plasticity, and sustained neuronal activity. EE housing also prevented memory impairment through the prevention of amyloid beta (Aβ) seeding formation, an early stage of Aβ plaque formation. The rodents subjected to EE were observed to present increased exploratory activity and exert less anxiety-related behaviour, compared to those in standard housing. However, some studies have proposed that EE intervention through exercise would be too mild to counteract the anxiety-related behaviour and risk assessment behaviour deficits in the Alzheimer's disease rodent model. Future studies should be conducted on old-aged rodents and the duration of EE exposure that would elicit the greatest benefits since the existing studies have been conducted on a range of ages and EE durations. In summary, EE had a considerable effect on dementia rodent models, with the most evident being improved cognitive function.
Collapse
Affiliation(s)
- Siti Norhafizah Mohd Sahini
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Outpatient Pharmacy Department, Hospital Raja Permaisuri Bainun, 30450 Ipoh, Perak, Malaysia
| | - Nurul Aqmar Mohd Nor Hazalin
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Integrative Pharmacogenomics Institute (iPROMiSE), Level 7, FF3, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | - Bettadapura N Srikumar
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru - 560029, India
| | - Hanish Singh Jayasingh Chellammal
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Brain Degeneration and Therapeutics Group, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia
| | - Gurmeet Kaur Surindar Singh
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Brain Degeneration and Therapeutics Group, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia.
| |
Collapse
|
10
|
Tang L, Zhao HQ, Yang H, Hu C, Ma SJ, Xiao WZ, Qing YH, Yang L, Zhou RR, Liu J, Zhang SH. Spectrum-effect relationship combined with bioactivity evaluation to discover the main anxiolytic active components of Baihe Dihuang decoction. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117090. [PMID: 37640258 DOI: 10.1016/j.jep.2023.117090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/06/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anxiety disorders leads to a decline in quality of life and increased risk of morbidity and mortality. The Baihe Dihuang decoction (BDD) is a classic Chinese medical formula that has been widely used to treat anxiety disorders for thousands of years in China. However, the pharmacodynamic material that is responsible for the antianxiety of BDD remains unclear. AIM OF THE STUDY To screen the main ingredients of anti-anxiety in BDD based on the establishment of spectrum-effect relationship and verified experiment. METHODS The UPLC-Q-TOF/MS technique was utilized to establish fingerprints of various fractions of BDD and identify the main compounds. The anti-anxiety effects of BDD were comprehensively evaluated through multiple assessments, including the open field test, elevated plus maze test, and neurotransmitters tests. Then, the spectrum-effect relationship was established through Pearson correlation analysis, gray correlation analysis, orthogonal partial least squares regression analysis. The spectrum-effect relationship results were confirmed through various measures on an anxiety condition cell model, induced by a corticosterone and lipopolysaccharide intervention. These measures included assessing neuronal cell viability, morphology, apoptosis, synaptic damage, and the expression of neurotransmitters and inflammatory factors. RESULTS In the UPLC-Q-TOF-MS fingerprint, 46 common peaks were identified. The pharmacological results indicated that different fractions of BDD have strong effects on improving anxiety-like behavior and regulating neurotransmitters. Among them, butanol fraction has the highest comprehensive evaluation score of anti-anxiety efficacy, which is main active fraction of BDD for anti-anxiety. The analysis of the spectrum-effect relationship revealed that the 46 peaks exhibited varying degrees of correlation with the anti-anxiety efficacy indicators of BDD. Among them, 14 components have a high correlation with the anti-anxiety efficacy indicators, which may be the potential anti-anxiety efficacy components of BDD. The in vitro activity verification of active components verified our prediction, regaloside A, B, C, D, H, acteoside, and isoacteoside improved neuronal cell viability, cell morphology, apoptosis, and synaptic damage. Additionally, regaloside A, B, C, D, H and acteoside regulated the neurotransmitter levels, while regaloside A, B, C, D, acteoside and isoacteoside inhibited the levels of inflammatory cytokines. CONCLUSION The butanol fraction was found to be the main active fraction of BDD, and 14 compounds were the major anxiolytic active components. The results of verifying the major active components were consistent with the predicted results of the spectrum-effect analysis. The developed spectrum-effect analysis in this study demonstrates high accuracy and reliability for screening active components in TCMs.
Collapse
Affiliation(s)
- Lin Tang
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hong-Qing Zhao
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hui Yang
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Chao Hu
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Si-Jing Ma
- Hunan Academy of Chinese Medicine, Changsha, Hunan Province, China
| | - Wang-Zhong Xiao
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Yu-Hui Qing
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Lei Yang
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Rong-Rong Zhou
- Hunan Academy of Chinese Medicine, Changsha, Hunan Province, China.
| | - Jian Liu
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Shui-Han Zhang
- Hunan Academy of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
11
|
Fong H, Zhou B, Feng H, Luo C, Bai B, Zhang J, Wang Y. Recapitulation of Structure-Function-Regulation of Blood-Brain Barrier under (Patho)Physiological Conditions. Cells 2024; 13:260. [PMID: 38334652 PMCID: PMC10854731 DOI: 10.3390/cells13030260] [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: 10/30/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
The blood-brain barrier (BBB) is a remarkable and intricate barrier that controls the exchange of molecules between the bloodstream and the brain. Its role in maintaining the stability of the central nervous system cannot be overstated. Over the years, advancements in neuroscience and technology have enabled us to delve into the cellular and molecular components of the BBB, as well as its regulation. Yet, there is a scarcity of comprehensive reviews that follow a logical framework of structure-function-regulation, particularly focusing on the nuances of BBB regulation under both normal and pathological conditions. This review sets out to address this gap by taking a historical perspective on the discovery of the BBB and highlighting the major observations that led to its recognition as a distinct brain barrier. It explores the intricate cellular elements contributing to the formation of the BBB, including endothelial cells, pericytes, astrocytes, and neurons, emphasizing their collective role in upholding the integrity and functionality of the BBB. Furthermore, the review delves into the dynamic regulation of the BBB in physiological states, encompassing neural, humoral, and auto-regulatory mechanisms. By shedding light on these regulatory processes, a deeper understanding of the BBB's response to various physiological cues emerges. This review also investigates the disruption of the BBB integrity under diverse pathological conditions, such as ischemia, infection, and toxin exposure. It elucidates the underlying mechanisms that contribute to BBB dysfunction and explores potential therapeutic strategies that aim to restore the BBB integrity and function. Overall, this recapitulation provides valuable insights into the structure, functions, and regulation of the BBB. By integrating historical perspectives, cellular elements, regulatory mechanisms, and pathological implications, this review contributes to a more comprehensive understanding of the BBB and paves the way for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Hin Fong
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - Botao Zhou
- Department of Physiology, Basic Medical and Public Health School, Jinan University, Guangzhou 510632, China;
| | - Haixiao Feng
- Gies College of Business, University of Illinois Urbana-Champaign, Urbana-Champaign, IL 61801, USA;
| | - Chuoying Luo
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - Boren Bai
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - John Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA;
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuechun Wang
- Department of Physiology, Basic Medical and Public Health School, Jinan University, Guangzhou 510632, China;
| |
Collapse
|
12
|
Laighneach A, Kelly JP, Desbonnet L, Holleran L, Kerr DM, McKernan D, Donohoe G, Morris DW. Social isolation-induced transcriptomic changes in mouse hippocampus impact the synapse and show convergence with human genetic risk for neurodevelopmental phenotypes. PLoS One 2023; 18:e0295855. [PMID: 38127959 PMCID: PMC10735045 DOI: 10.1371/journal.pone.0295855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Early life stress (ELS) can impact brain development and is a risk factor for neurodevelopmental disorders such as schizophrenia. Post-weaning social isolation (SI) is used to model ELS in animals, using isolation stress to disrupt a normal developmental trajectory. We aimed to investigate how SI affects the expression of genes in mouse hippocampus and to investigate how these changes related to the genetic basis of neurodevelopmental phenotypes. BL/6J mice were exposed to post-weaning SI (PD21-25) or treated as group-housed controls (n = 7-8 per group). RNA sequencing was performed on tissue samples from the hippocampus of adult male and female mice. Four hundred and 1,215 differentially-expressed genes (DEGs) at a false discovery rate of < 0.05 were detected between SI and control samples for males and females respectively. DEGS for both males and females were significantly overrepresented in gene ontologies related to synaptic structure and function, especially the post-synapse. DEGs were enriched for common variant (SNP) heritability in humans that contributes to risk of neuropsychiatric disorders (schizophrenia, bipolar disorder) and to cognitive function. DEGs were also enriched for genes harbouring rare de novo variants that contribute to autism spectrum disorder and other developmental disorders. Finally, cell type analysis revealed populations of hippocampal astrocytes that were enriched for DEGs, indicating effects in these cell types as well as neurons. Overall, these data suggest a convergence between genes dysregulated by the SI stressor in the mouse and genes associated with neurodevelopmental disorders and cognitive phenotypes in humans.
Collapse
Affiliation(s)
- Aodán Laighneach
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| | - John P. Kelly
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Lieve Desbonnet
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Laurena Holleran
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| | - Daniel M. Kerr
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Declan McKernan
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| | - Derek W. Morris
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, Galway, Ireland
| |
Collapse
|
13
|
Guimarães DM, Valério-Gomes B, Vianna-Barbosa RJ, Oliveira W, Neves GÂ, Tovar-Moll F, Lent R. Social isolation leads to mild social recognition impairment and losses in brain cellularity. Brain Struct Funct 2023; 228:2051-2066. [PMID: 37690044 DOI: 10.1007/s00429-023-02705-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Chronic social stress is a significant risk factor for several neuropsychiatric disorders, mainly major depressive disorder (MDD). In this way, patients with clinical depression may display many symptoms, including disrupted social behavior and anxiety. However, like many other psychiatric diseases, MDD has a very complex etiology and pathophysiology. Because social isolation is one of the multiple depression-inducing factors in humans, this study aims to understand better the link between social stress and MDD using an animal model based on social isolation after weaning, which is known to produce social stress in mice. We focused on cellular composition and white matter integrity to establish possible links with the abnormal social behavior that rodents isolated after weaning displayed in the three-chamber social approach and recognition tests. We used the isotropic fractionator method to assess brain cellularity, which allows us to robustly estimate the number of oligodendrocytes and neurons in dissected brain regions. In addition, diffusion tensor imaging (DTI) was employed to analyze white matter microstructure. Results have shown that post-weaning social isolation impairs social recognition and reduces the number of neurons and oligodendrocytes in important brain regions involved in social behavior, such as the anterior neocortex and the olfactory bulb. Despite the limitations of animal models of psychological traits, evidence suggests that behavioral impairments observed in patients might have similar biological underpinnings.
Collapse
Affiliation(s)
- Daniel Menezes Guimarães
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Robarts Research Institute, University of Western Ontario, London, Canada.
| | - Bruna Valério-Gomes
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Washington Oliveira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilda Ângela Neves
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Roberto Lent
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- D'Or Institute of Research and Education, Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Hong H, Guo C, Liu X, Yang L, Ren W, Zhao H, Li Y, Zhou Z, Lam SM, Mi J, Zuo Z, Liu C, Wang GD, Zhuo Y, Zhang YP, Li Y, Shui G, Zhang YQ, Xiong Y. Differential effects of social isolation on oligodendrocyte development in different brain regions: insights from a canine model. Front Cell Neurosci 2023; 17:1201295. [PMID: 37538851 PMCID: PMC10393781 DOI: 10.3389/fncel.2023.1201295] [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: 04/06/2023] [Accepted: 06/07/2023] [Indexed: 08/05/2023] Open
Abstract
Social isolation (SI) exerts diverse adverse effects on brain structure and function in humans. To gain an insight into the mechanisms underlying these effects, we conducted a systematic analysis of multiple brain regions from socially isolated and group-housed dogs, whose brain and behavior are similar to humans. Our transcriptomic analysis revealed reduced expression of myelin-related genes specifically in the white matter of prefrontal cortex (PFC) after SI during the juvenile stage. Despite these gene expression changes, myelin fiber organization in PFC remained unchanged. Surprisingly, we observed more mature oligodendrocytes and thicker myelin bundles in the somatosensory parietal cortex in socially isolated dogs, which may be linked to an increased expression of ADORA2A, a gene known to promote oligodendrocyte maturation. Additionally, we found a reduced expression of blood-brain barrier (BBB) structural components Aquaporin-4, Occludin, and Claudin1 in both PFC and parietal cortices, indicating BBB disruption after SI. In agreement with BBB disruption, myelin-related sphingolipids were increased in cerebrospinal fluid in the socially isolated group. These unexpected findings show that SI induces distinct alterations in oligodendrocyte development and shared disruption in BBB integrity in different cortices, demonstrating the value of dogs as a complementary animal model to uncover molecular mechanisms underlying SI-induced brain dysfunction.
Collapse
Affiliation(s)
- Huilin Hong
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Chao Guo
- Division of Life Sciences and Medicine, School of Life Sciences, University of Science and Technology of China, Hefei, China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xueru Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Liguang Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei Ren
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hui Zhao
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yuan Li
- Beijing Sinogene Biotechnology Co., Ltd., Beijing, China
| | - Zhongyin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Sin Man Lam
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Jidong Mi
- Beijing Sinogene Biotechnology Co., Ltd., Beijing, China
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Cirong Liu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Guo-Dong Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yan Zhuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yixue Li
- Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guanghou Shui
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yong Q. Zhang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Xiong
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Wang G, Cheng Z, Zhou Y, Duan J, Kan Q, Tang S. The effect of childhood social isolation on behavioral cognition in Chinese middle-aged and older adults: The moderating effect of family support. Arch Gerontol Geriatr 2023; 113:105060. [PMID: 37270941 DOI: 10.1016/j.archger.2023.105060] [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: 02/10/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/06/2023]
Abstract
INTRODUCTION This study aimed to analyze the association of childhood social isolation and behavioral cognition, and moderating effect of family support in middle-aged and older adults. METHODS The study samples were from the China Health and Retirement Longitudinal Study (CHARLS) of 2014 and 2018. The dependent variable was behavioral cognitive ability, measured by episodic memory and mental state; The independent variable was childhood social isolation, and the moderating variable was family support. The baseline OLS regression model was used to explore the correlation among independent variables, dependent variables and moderating variables, the least square regression model was used to analyze the moderating effect of family support, and the replacement model and the method of replacing characteristic variables were used for the robustness test. Hierarchical regression heterogeneity analysis was performed to further verify the results of the moderating effect. RESULTS In this study, 3459 samples were selected for analysis. OLS baseline regression results showed that the deepening of childhood social isolation was significantly correlated with the decline of behavioral cognitive ability in middle-aged and elderly people (β=-0.9664, t = 0.0893). After adding all covariates, we found that childhood social isolation was significantly negatively correlated with behavioral cognitive ability in middle-aged and elderly people (β=-0.4118, t = 0.0785). Among the moderating variables of family support, we found that there is a moderating effect on female guardians' efforts of caring in early parental support (β=0.0948, t = 0.0320) and the frequency of children's visits in late children support (β=0.0073, t = 0.0036). Finally, through the heterogeneity test, we found that there were differences in the relationship between childhood social isolation and behavioral cognitive ability among middle-aged and elderly people of different ages, genders and places of residence. Moreover, there are significant differences in the moderating effect of female guardian's effort of caring and the frequency of children's visits in heterogeneous groups. CONCLUSIONS The greater the degree of childhood social isolation experienced by middle-aged and elderly people, the worse their behavioral cognitive ability. Female guardian's effort of caring and the frequency of children's visits have a moderating effect to suppress this negative effect.
Collapse
Affiliation(s)
- Gaoling Wang
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaopeng Cheng
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuqin Zhou
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Duan
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qianqian Kan
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaoliang Tang
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
16
|
Neelotpol S, Rezwan R, Shahriar M. Chemical analysis of calabash chalk and its effect on locomotor activities and behavior in Swiss albino mice. Heliyon 2023; 9:e14463. [PMID: 36994387 PMCID: PMC10040505 DOI: 10.1016/j.heliyon.2023.e14463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 01/29/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Continuous exposure of young women to calabash chalk, especially at child-bearing age, necessitated this study to analyze the chemical constituents of calabash chalk and to evaluate its effect on locomotor activities and behavior in Swiss albino mice. Dried cubes of calabash chalk were purchased and analyzed by atomic and flame atomic absorption spectrophotometer. Twenty-four Swiss-albino mice were taken and divided into four groups: control (1 ml distilled water) and three treated groups with the doses of 200 mg/kg, 400 mg/kg, and 600 mg/kg of calabash chalk suspension, respectively, by oral gavage. Hole Cross, Hole Board, and Open Field tests were performed to evaluate the locomotor activities, behavior, and anxiety along with measurement of body weight. The data were analyzed by SPSS-software. Chemical analysis of calabash chalk showed the presence of trace elements along with some heavy metals such as lead (19.26 ppm), Chromium (34.73 ppm), and arsenic (4.57 ppm). After 21 days of oral administration of calabash chalk, the study showed a significant decrease in body weight in the treated groups of mice (p < 0.01). Decreased locomotor activities were also observed in all three experiments. Significantly decreased locomotion and behaviors were also observed in the hole crossing, line crossing, head dipping, grooming, rearing, stretch attend, central square entry, central square duration, defecation, and urination, in a dose-dependent manner (p < 0.01). These effects also prove the anxiogenic behavior of calabash chalk in albino mice. Heavy metals are believed to be harmful to the brain and responsible for cognitive dysfunction and elevated anxiety. In this study, decreased body weight in mice may occur due to disorders in hunger and thirst centers of the brain by heavy metals. Therefore, heavy metals may be responsible for muscle weakness, decreased locomotor activities, and axiogenic effects of mice.
Collapse
|
17
|
Challa SR, Fornal CA, Wang BC, Boyineni J, DeVera RE, Unnam P, Song Y, Soares MB, Malchenko S, Gyarmati P, Veeravalli KK. The Impact of Social Isolation and Environmental Deprivation on Blood Pressure and Depression-Like Behavior in Young Male and Female Mice. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2023; 7:24705470231207010. [PMID: 37859939 PMCID: PMC10583512 DOI: 10.1177/24705470231207010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023]
Abstract
Background: Social isolation (SI) and loneliness are major adult and adolescent health concerns, particularly in the coronavirus disease 2019 (COVID-19) era. Recent prospective cohort studies indicate that older women who experienced both SI and loneliness had a significantly higher risk of cardiovascular disease (CVD). Hypertension, a well-established risk factor for CVD, is more prevalent in elderly women than men. Furthermore, a lack of social relationships is strongly associated with an increased risk of hypertension in middle-aged and elderly women compared to men. Although this has not been extensively studied, adolescents and young adults who experience loneliness or SI may also be at risk for CVD and depression. The purpose of this study was to examine the effect of SI on blood pressure and depression-like behavior in young male and female mice. Methods: Weaned C57BL/6 mice were randomly assigned (n = 6/group/sex) to either group housing (GH) or SI. Animals in the SI group were housed in individual cages for 8 weeks with no view of other animals. The cages were kept in ventilated racks to prevent pheromone exposure and socially isolated animals had no cage enrichment. Results: SI increased systolic, diastolic, and mean arterial blood pressure in females and elevated heart rate in both sexes. Body weight gain was dramatically increased in socially isolated females but tended to decrease in socially isolated males. In the forced swim test, which detects depression-like behavior, there was no difference between groups in total immobility time. The latency to immobility, however, was significantly decreased in socially isolated females. Serum concentrations of corticosterone and metanephrine did not differ between socially isolated and group-housed females, but corticosterone levels were significantly reduced in socially isolated males. Conclusions: Our results indicate that 8 weeks of SI leads to significant changes in blood pressure and heart rate and mild changes in depression-like behavior in young mice, with females affected more than males.
Collapse
Affiliation(s)
- Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
- Department of Pharmacology, KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, AP, India
| | - Casimir A. Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Billy C. Wang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
- Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Jerusha Boyineni
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Roberto E. DeVera
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Pavani Unnam
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Yajing Song
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Marcelo Bento Soares
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL, USA
- Department of Psychiatry and Behavioral Medicine, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Sergey Malchenko
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Peter Gyarmati
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
- Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL, USA
- Department of Neurology, University of Illinois College of Medicine Peoria, Peoria, IL, USA
| |
Collapse
|