1
|
Wu P, Wang W, Huang C, Sun L, Wu X, Xu L, Xiao P. A rapid and reliable targeted LC-MS/MS method for quantitative analysis of the Tryptophan-NAD metabolic network disturbances in tissues and blood of sleep deprivation mice. Anal Chim Acta 2024; 1328:343125. [PMID: 39266191 DOI: 10.1016/j.aca.2024.343125] [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: 04/20/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND TRY-NAD metabolic network includes TRY (tryptophan), 5-HT (5-hydroxytryptamine), KYN (kynurenine), and NAD (nicotinamide adenine dinucleotide) pathway, which plays a significant role in neurological diseases and ageing. It is important to monitor these metabolites for studying the pathological anatomy of disease and treatment of responses evaluation. Although previous studies have reported quantitative methods for several metabolites in the network, the bottlenecks of simultaneously quantifying the whole metabolic network are their similar structures, diverse physico-chemical properties, and instability. Standardized protocols for the whole metabolic network are still missing, which hinders the in-depth study of TRY-NAD metabolic network in laboratory research and clinical screening. RESULTS We developed a LC-MS/MS method for quantifying 28 metabolites in the TRY-NAD network simultaneously. Optimization was done for the mass spectral parameters, chromatographic conditions and sample pretreatment process. The developed method was fully validated in terms of standard curves, sensitivity, carryover, recovery, matrix effect, accuracy, precision, and stability. The pretreatment of 30 samples only takes 90 min, and the LC-MS/MS running time of one sample is only 13 min. With this method, we bring to light the chaos of global TRY-NAD metabolic network in sleep deprivation mice for the first time, including serum, clotted blood cells, hippocampus, cerebral cortex, and liver. NAD pathway levels in brain and blood decreased, whereas the opposite happened in the liver. The 5-HT pathway decreased and the concentration of KYN increased significantly in the brain. The concentration of many metabolites in KYN pathway (NAD+ de novo synthesis pathway) increased in the liver. SIGNIFICANCE This method is the first time to determine the metabolites of KYN, 5-HT and NAD pathway at the same time, and it is found that TRY-NAD metabolic network will be disordered after sleep deprivation. This work clarifies the importance of the pH of the extraction solution, the time and temperature control in pretreatment in standardized protocols building, and overcoming the problems of inconsistent sample pretreatment, separation, matrix effect interference and potential metabolite degradation. This method exhibits great prospects in providing more information on metabolic disturbances caused by sleep deprivation as well as neurological diseases and ageing.
Collapse
Affiliation(s)
- Peiling Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Wenjie Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Chuan Huang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Le Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Xiaoli Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Lijia Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China; Key Laboratory of Resources Conservation and Development of Southern Medicine of Hainan Province & Key Laboratory of State Administration of Traditional Chinese Medicine for Agarwood Sustainable Utilization, Hainan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Haikou, 570311, China.
| | - Peigen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
2
|
Miller VK, Broadie K. Experience-dependent serotonergic signaling in glia regulates targeted synapse elimination. PLoS Biol 2024; 22:e3002822. [PMID: 39352884 PMCID: PMC11444420 DOI: 10.1371/journal.pbio.3002822] [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: 04/22/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
The optimization of brain circuit connectivity based on initial environmental input occurs during critical periods characterized by sensory experience-dependent, temporally restricted, and transiently reversible synapse elimination. This precise, targeted synaptic pruning mechanism is mediated by glial phagocytosis. Serotonin signaling has prominent, foundational roles in the brain, but functions in glia, or in experience-dependent brain circuit synaptic connectivity remodeling, have been relatively unknown. Here, we discover that serotonergic signaling between glia is essential for olfactory experience-dependent synaptic glomerulus pruning restricted to a well-defined Drosophila critical period. We find that experience-dependent serotonin signaling is restricted to the critical period, with both (1) serotonin production and (2) 5-HT2A receptors specifically in glia, but not neurons, absolutely required for targeted synaptic glomerulus pruning. We discover that glial 5-HT2A receptor signaling limits the experience-dependent synaptic connectivity pruning in the critical period and that conditional reexpression of 5-HT2A receptors within adult glia reestablishes "critical period-like" experience-dependent synaptic glomerulus pruning at maturity. These results reveal an essential requirement for glial serotonergic signaling mediated by 5-HT2A receptors for experience-dependent synapse elimination.
Collapse
Affiliation(s)
- Vanessa Kay Miller
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
3
|
Butler JJ, Ricci D, Aman C, Beyeler A, De Deurwaerdère P. Classical psychedelics' action on brain monoaminergic systems. Int J Biochem Cell Biol 2024; 176:106669. [PMID: 39332625 DOI: 10.1016/j.biocel.2024.106669] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
The study of the mechanism of action of classical psychedelics has gained significant interest due to their clinical potential in the treatment of several psychiatric conditions, including major depressive and anxiety disorders. These drugs bind 5-hydroxytryptamine receptors (5-HTR) including 5-HT1AR, 5-HT2AR, 5-HT2BR, and/or 5-HT2CR, as well as other targets. 5-HTRs regulate the activity of ascending monoaminergic neurons, a mechanism primarily involved in the action of classical antidepressant drugs, antipsychotics, and drugs of abuse. Sparse neurochemical data have been produced on the control of monoaminergic neuron activity in response to classical psychedelics. Here we review the available data in order to determine whether classical psychedelics have specific neurochemical effects on serotonergic, dopaminergic, and noradrenergic neurons. The data show that these drugs have disparate effects on each monoaminergic system, demonstrating a complex response with state-dependent and region-specific effects. For instance, several psychedelics inhibit the firing of serotonergic neurons, although this is not necessarily associated with a decrease in serotonin release in all regions. Noradrenergic neuron spontaneous activity also appears to be inhibited by psychedelics, also not necessarily associated with a decrease in noradrenaline release in all regions. Psychedelics influence on dopaminergic systems is also complex as the above-mentioned 5-HTRs may have opposing effects on dopaminergic neuron activity, in a state-dependent manner. There is an apparent lack of clear neuronal signature induced by psychedelics on monoaminergic neuron activity despite specific recurrent mechanisms. This review provides a current summary of the action of psychedelics on monoamine neuromodulators serotonin, dopamine and noradrenaline, compiling reoccurring and contradictory findings demonstrating that a monoamine signature of psychedelics, if applicable, would be state- and region-dependant.
Collapse
Affiliation(s)
- Jasmine Jade Butler
- University of Bordeaux, France; Centre National de la Recherche Scientifique (CNRS), unit 5287, Institut des Neurosciences Intégratives et Cognitives d'Aquitaine (INCIA), Bordeaux, France
| | - Daria Ricci
- University of Bordeaux, France; Institut National pour la Santé et la Recherche Médicale (INSERM), unit 1215, Neurocentre Magendie, Bordeaux, France
| | - Chloé Aman
- University of Bordeaux, France; Centre National de la Recherche Scientifique (CNRS), unit 5287, Institut des Neurosciences Intégratives et Cognitives d'Aquitaine (INCIA), Bordeaux, France
| | - Anna Beyeler
- University of Bordeaux, France; Institut National pour la Santé et la Recherche Médicale (INSERM), unit 1215, Neurocentre Magendie, Bordeaux, France.
| | - Philippe De Deurwaerdère
- University of Bordeaux, France; Centre National de la Recherche Scientifique (CNRS), unit 5287, Institut des Neurosciences Intégratives et Cognitives d'Aquitaine (INCIA), Bordeaux, France.
| |
Collapse
|
4
|
Tzitiridou-Chatzopoulou M, Kountouras J, Zournatzidou G. The Potential Impact of the Gut Microbiota on Neonatal Brain Development and Adverse Health Outcomes. CHILDREN (BASEL, SWITZERLAND) 2024; 11:552. [PMID: 38790548 PMCID: PMC11119242 DOI: 10.3390/children11050552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024]
Abstract
Over the past decade, microbiome research has significantly expanded in both scope and volume, leading to the development of new models and treatments targeting the gut-brain axis to mitigate the effects of various disorders. Related research suggests that interventions during the critical period from birth to three years old may yield the greatest benefits. Investigating the substantial link between the gut and brain during this crucial developmental phase raises fundamental issues about the role of microorganisms in human health and brain development. This underscores the importance of focusing on the prevention rather than the treatment of neurodevelopmental and neuropsychiatric disorders. The present review examines the gut microbiota from birth to age 3, with a particular focus on its potential relationship with neurodevelopment. This review emphasizes the immunological mechanisms underlying this relationship. Additionally, the study investigates the impact of the microbiome on cognitive development and neurobehavioral issues such as anxiety and autism. Importantly, it highlights the need to integrate mechanistic studies of animal models with epidemiological research across diverse cultures to better understand the role of a healthy microbiome in early life and the implications of dysbiosis. Furthermore, this review summarizes factors contributing to the transmission of gut microbiome-targeted therapies and their effects on neurodevelopment. Recent studies on environmental toxins known to impact neurodevelopment are also reviewed, exploring whether the microbiota may mitigate or modulate these effects.
Collapse
Affiliation(s)
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54 642 Thessaloniki, Greece;
| | - Georgia Zournatzidou
- Department of Business Administration, University of Western Macedonia, 50 100 Kozani, Greece
- Department of Accounting and Finance, Hellenic Mediterranean University, 71 410 Heraklion, Greece
| |
Collapse
|
5
|
Del Pozo A, Knox KM, Lehmann LM, Davidson S, Rho SL, Jayadev S, Barker-Haliski M. Chronic evoked seizures in young pre-symptomatic APP/PS1 mice induce serotonin changes and accelerate onset of Alzheimer's disease-related neuropathology. Prog Neurobiol 2024; 235:102591. [PMID: 38484965 PMCID: PMC11015961 DOI: 10.1016/j.pneurobio.2024.102591] [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: 10/31/2023] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVE Hyperexcitability is intimately linked to Alzheimer's disease (AD) pathology, but the precise timing and contributions of neuronal hyperexcitability to disease progression is unclear. Seizure induction in rodent AD models can uncover new therapeutic targets. Further, investigator-evoked seizures can directly establish how hyperexcitability and AD-associated risk factors influence neuropathological hallmarks and disease course at presymptomatic stages. METHODS Corneal kindling is a well-characterized preclinical epilepsy model that allows for precise control of seizure history to pair to subsequent behavioral assessments. 2-3-month-old APP/PS1, PSEN2-N141I, and transgenic control male and female mice were thus sham or corneal kindled for 2 weeks. Seizure-induced changes in glia, serotonin pathway proteins, and amyloid β levels in hippocampus and prefrontal cortex were quantified. RESULTS APP/PS1 females were more susceptible to corneal kindling. However, regardless of sex, APP/PS1 mice experienced extensive seizure-induced mortality versus kindled Tg- controls. PSEN2-N141I mice were not negatively affected by corneal kindling. Mortality correlated with a marked downregulation of hippocampal tryptophan hydroxylase 2 and monoamine oxidase A protein expression versus controls; these changes were not detected in PSEN2-N141I mice. Kindled APP/PS1 mice also exhibited soluble amyloid β upregulation and glial reactivity without plaque deposition. SIGNIFICANCE Evoked convulsive seizures and neuronal hyperexcitability in pre-symptomatic APP/PS1 mice promoted premature mortality without pathological Aβ plaque deposition, whereas PSEN2-N141I mice were unaffected. Disruptions in serotonin pathway metabolism in APP/PS1 mice was associated with increased glial reactivity without Aβ plaque deposition, demonstrating that neuronal hyperexcitability in early AD causes pathological Aβ overexpression and worsens long-term outcomes through a serotonin-related mechanism.
Collapse
Affiliation(s)
- Aaron Del Pozo
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Kevin M Knox
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Leanne M Lehmann
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Stephanie Davidson
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Seongheon Leo Rho
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Melissa Barker-Haliski
- Center for Epilepsy Drug Discovery (CEDD), Department of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
6
|
Nikolopoulos D, Nakos-Bimpos M, Manolakou T, Polissidis A, Boumpas DT. Impaired serotonin synthesis in hippocampus of murine lupus represents an early neuropsychiatric event. Lupus 2024; 33:166-171. [PMID: 38073556 DOI: 10.1177/09612033231221651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
BACKGROUND Despite significant progress in understanding the mechanisms underlying hippocampal involvement in neuropsychiatric systemic lupus erythematosus (NPSLE), our understanding of how neuroinflammation affects the brain neurotransmitter systems is limited. To date, few studies have investigated the role of neurotransmitters in pathogenesis of NPSLE with contradictory results. METHODS Hippocampal tissue from NZB/W-F1 lupus-prone mice and age-matched control strains were dissected in both pre-nephritic (3-month-old) and nephritic (6-month-old) stages. High-Performance Liquid Chromatography (HPLC) was used to evaluate the level of serotonin (5-HT), dopamine (DA), and their metabolites 5-HIAA and DOPAC, respectively, in mouse hippocampi. RESULTS Lupus mice exhibit decreased levels of serotonin at the early stages of the disease, along with intact levels of its metabolite 5-HIAA. The 5-HT turnover ratio (5-HIAA/5-HT ratio) was increased in the hippocampus of lupus mice at pre-nephritic stage suggesting that low hippocampal serotonin levels in lupus are attributed to decreased serotonin synthesis. Both DA and DOPAC levels remained unaffected in lupus hippocampus at both early and late stages. CONCLUSION Impaired hippocampal serotonin synthesis in the hippocampus of lupus-prone mice represents an early neuropsychiatric event. These findings may have important implications for the use of symptomatic therapy in diffuse NPSLE.
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
| | - Modestos Nakos-Bimpos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy 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
| | - Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, 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
| |
Collapse
|
7
|
Chu CH, Chen JS, Chan YL, Lu WJ, Huang YT, Mao PC, Sze CI, Sun HS. TIAM2S-positive microglia enhance inflammation and neurotoxicity through soluble ICAM-1-mediated immune priming. FASEB J 2023; 37:e23242. [PMID: 37801065 DOI: 10.1096/fj.202300462rr] [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: 03/10/2023] [Revised: 09/05/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
TIAM Rac1-associated GEF 2 short form (TIAM2S) as an oncoprotein alters the immunity of peripheral immune cells to construct an inflammatory tumor microenvironment. However, its role in the activation of microglia, the primary innate immune cells of the brain, and neuroinflammation remains unknown. This study investigated the mechanism underlying TIAM2S shapes immune properties of microglia to facilitate neuron damage. Human microglial clone 3 cell line (HMC3) and human brain samples were applied to determine the presence of TIAM2S in microglia by western blots and double immunostaining. Furthermore, TIAM2S transgenic mice combined with multiple reconstituted primary neuron-glial culture systems and a cytokine array were performed to explore how TIAM2S shaped immune priming of microglia and participated in lipopolysaccharide (LPS)-induced neuron damage. TIAM2S protein was detectable in HMC3 cells and presented in a small portion (~11.1%) of microglia in human brains referred to as TIAM2S-positive microglia. With the property of secreted soluble factor-mediated immune priming, TIAM2S-positive microglia enhanced LPS-induced neuroinflammation and neural damage in vivo and in vitro. The gain- and loss-of-function experiments showed soluble intercellular adhesion molecule-1 (sICAM-1) participated in neurotoxic immune priming of TIAM2S+ microglia. Together, this study demonstrated a novel TIAM2S-positive microglia subpopulation enhances inflammation and neurotoxicity through sICAM-1-mediated immune priming.
Collapse
Affiliation(s)
- Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jia-Shing Chen
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Ya-Ling Chan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Jen Lu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Te Huang
- Department of Geriatrics and Gerontology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pin-Cheng Mao
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-I Sze
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
8
|
Fang S, Wu Z, Guo Y, Zhu W, Wan C, Yuan N, Chen J, Hao W, Mo X, Guo X, Fan L, Li X, Chen J. Roles of microglia in adult hippocampal neurogenesis in depression and their therapeutics. Front Immunol 2023; 14:1193053. [PMID: 37881439 PMCID: PMC10597707 DOI: 10.3389/fimmu.2023.1193053] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Adult hippocampal neurogenesis generates functional neurons from neural progenitor cells in the hippocampal dentate gyrus (DG) to complement and repair neurons and neural circuits, thus benefiting the treatment of depression. Increasing evidence has shown that aberrant microglial activity can disrupt the appropriate formation and development of functional properties of neurogenesis, which will play a crucial role in the occurrence and development of depression. However, the mechanisms of the crosstalk between microglia and adult hippocampal neurogenesis in depression are not yet fully understood. Therefore, in this review, we first introduce recent discoveries regarding the roles of microglia and adult hippocampal neurogenesis in the etiology of depression. Then, we systematically discuss the possible mechanisms of how microglia regulate adult hippocampal neurogenesis in depression according to recent studies, which involve toll-like receptors, microglial polarization, fractalkine-C-X3-C motif chemokine receptor 1, hypothalamic-pituitary-adrenal axis, cytokines, brain-derived neurotrophic factor, and the microbiota-gut-brain axis, etc. In addition, we summarize the promising drugs that could improve the adult hippocampal neurogenesis by regulating the microglia. These findings will help us understand the complicated pathological mechanisms of depression and shed light on the development of new treatment strategies for this disease.
Collapse
Affiliation(s)
- Shaoyi Fang
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhibin Wu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yali Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wenjun Zhu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Chunmiao Wan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Shenzhen People’s Hospital, 2Clinical Medical College, Jinan University, Shenzhen, China
| | - Jianbei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzhi Hao
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaowei Mo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaofang Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lili Fan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaojuan Li
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaxu Chen
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Zhu H, Guan A, Liu J, Peng L, Zhang Z, Wang S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J Neuroinflammation 2023; 20:223. [PMID: 37794488 PMCID: PMC10548593 DOI: 10.1186/s12974-023-02901-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Microglia are so versatile that they not only provide immune surveillance for central nervous system, but participate in neural circuitry development, brain blood vessels formation, blood-brain barrier architecture, and intriguingly, the regulation of emotions and behaviors. Microglia have a profound impact on neuronal survival, brain wiring and synaptic plasticity. As professional phagocytic cells in the brain, they remove dead cell debris and neurotoxic agents via an elaborate mechanism. The functional profile of microglia varies considerately depending on age, gender, disease context and other internal or external environmental factors. Numerous studies have demonstrated a pivotal involvement of microglia in neuropsychiatric disorders, including negative affection, social deficit, compulsive behavior, fear memory, pain and other symptoms associated with major depression disorder, anxiety disorder, autism spectrum disorder and schizophrenia. In this review, we summarized the latest discoveries regarding microglial ontogeny, cell subtypes or state spectrum, biological functions and mechanistic underpinnings of emotional and behavioral disorders. Furthermore, we highlight the potential of microglia-targeted therapies of neuropsychiatric disorders, and propose outstanding questions to be addressed in future research of human microglia.
Collapse
Affiliation(s)
- Hongrui Zhu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Ao Guan
- School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jiayuan Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Li Peng
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zhi Zhang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
10
|
Del Pozo A, Knox KM, Lehmann L, Davidson S, Rho S, Jayadev S, Barker-Haliski M. Chronic evoked seizures in young pre-symptomatic APP/PS1 mice induce serotonin changes and accelerate onset on Alzheimer's disease-related neurpathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522897. [PMID: 36711965 PMCID: PMC9881977 DOI: 10.1101/2023.01.05.522897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE People with early-onset Alzheimer's disease (AD) are at elevated seizure risk. Further, chronic seizures in pre-symptomatic stages may disrupt serotonin pathway-related protein expression, precipitating the onset of AD-related pathology and burden of neuropsychiatric comorbidities. METHODS 2-3-month-old APP/PS1, PSEN2-N141I, and transgenic control mice were sham or corneal kindled for 2 weeks to model chronic seizures. Seizure-induced changes in glia, serotonin pathway proteins, and amyloid beta; levels in hippocampus and prefrontal cortex were quantified. RESULTS APP/PS1 mice experienced worsened mortality versus kindled Tg- controls. APP/PS1 females were also more susceptible to chronic kindled seizures. These changes correlated with a marked downregulation of hippocampal tryptophan hydroxylase 2 and monoamine oxidase A protein expression compared to controls; these changes were not detected in PSEN2-N141I mice. Kindled APP/PS1 mice exhibited amyloid beta; overexpression and glial overactivity without plaque deposition. PSEN2 protein expression was AD model-dependent. SIGNIFICANCE Seizures evoked in pre-symptomatic APP/PS1 mice promotes premature mortality in the absence of pathological amyloid deposition. Disruptions in serotonin pathway metabolism are associated with increased glial reactivity and PSEN2 downregulation without amyloid beta; deposition. This study provides the first direct evidence that seizures occurring prior to amyloid beta, plaque accumulation worsen disease burden in an AD genotype-specific manner.
Collapse
|
11
|
Marinho LSR, Chiarantin GMD, Ikebara JM, Cardoso DS, de Lima-Vasconcellos TH, Higa GSV, Ferraz MSA, De Pasquale R, Takada SH, Papes F, Muotri AR, Kihara AH. The impact of antidepressants on human neurodevelopment: Brain organoids as experimental tools. Semin Cell Dev Biol 2023; 144:67-76. [PMID: 36115764 DOI: 10.1016/j.semcdb.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/10/2022] [Accepted: 09/10/2022] [Indexed: 11/23/2022]
Abstract
The use of antidepressants during pregnancy benefits the mother's well-being, but the effects of such substances on neurodevelopment remain poorly understood. Moreover, the consequences of early exposure to antidepressants may not be immediately apparent at birth. In utero exposure to selective serotonin reuptake inhibitors (SSRIs) has been related to developmental abnormalities, including a reduced white matter volume. Several reports have observed an increased incidence of autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) after prenatal exposure to SSRIs such as sertraline, the most widely prescribed SSRI. The advent of human-induced pluripotent stem cell (hiPSC) methods and assays now offers appropriate tools to test the consequences of such compounds for neurodevelopment in vitro. In particular, hiPSCs can be used to generate cerebral organoids - self-organized structures that recapitulate the morphology and complex physiology of the developing human brain, overcoming the limitations found in 2D cell culture and experimental animal models for testing drug efficacy and side effects. For example, single-cell RNA sequencing (scRNA-seq) and electrophysiological measurements on organoids can be used to evaluate the impact of antidepressants on the transcriptome and neuronal activity signatures in developing neurons. While the analysis of large-scale transcriptomic data depends on dimensionality reduction methods, electrophysiological recordings rely on temporal data series to discriminate statistical characteristics of neuronal activity, allowing for the rigorous analysis of the effects of antidepressants and other molecules that affect the developing nervous system, especially when applied in combination with relevant human cellular models such as brain organoids.
Collapse
Affiliation(s)
| | | | - Juliane Midori Ikebara
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | - Débora Sterzeck Cardoso
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | | | - Guilherme Shigueto Vilar Higa
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil; Department of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, São Paulo, SP 05508-000, Brazil
| | | | - Roberto De Pasquale
- Department of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, São Paulo, SP 05508-000, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | - Fabio Papes
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil; Center for Medicinal Chemistry, University of Campinas, Campinas, SP 13083-875, Brazil; Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, Center for Academic Research and Training in Anthropogeny, Kavli Institute for Brain and Mind, Archealization Center (ArchC), La Jolla, CA 92037, USA.
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil.
| |
Collapse
|
12
|
Higuchi Y, Arakawa H. Serotonergic mediation of the brain-wide neurogenesis: Region-dependent and receptor-type specific roles on neurogenic cellular transformation. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100102. [PMID: 37638344 PMCID: PMC10458724 DOI: 10.1016/j.crneur.2023.100102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/18/2023] [Accepted: 07/15/2023] [Indexed: 08/29/2023] Open
Abstract
Brain serotonin (5-hydroxytryptamine, 5-HT) is a key molecule for the mediation of depression-related brain states, but the neural mechanisms underlying 5-HT mediation need further investigation. A possible mechanism of the therapeutic antidepressant effects is neurogenic cell production, as stimulated by 5-HT signaling. Neurogenesis, the proliferation of neural stem cells (NSCs), and cell differentiation and maturation occur across brain regions, particularly the hippocampal dentate gyrus and the subventricular zone, throughout one's lifespan. 5-HT plays a major role in the mediation of neurogenic processes, which in turn leads to the therapeutic effect on depression-related states. In this review article, we aim to identify how the neuronal 5-HT system mediates the process of neurogenesis, including cell proliferation, cell-type differentiation and maturation. First, we will provide an overview of the neurogenic cell transformation that occurs in brain regions containing or lacking NSCs. Second, we will review brain region-specific mechanisms of 5-HT-mediated neurogenesis by comparing regions localized to NSCs, i.e., the hippocampus and subventricular zone, with those not containing NSCs. Highlighting these 5-HT mechanisms that mediate neurogenic cell production processes in a brain-region-specific manner would provide unique insights into the role of 5-HT in neurogenesis and its associated effects on depression.
Collapse
Affiliation(s)
- Yuki Higuchi
- Department of Systems Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hiroyuki Arakawa
- Department of Systems Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
13
|
Gulyaeva NV. Glucocorticoids Orchestrate Adult Hippocampal Plasticity: Growth Points and Translational Aspects. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:565-589. [PMID: 37331704 DOI: 10.1134/s0006297923050012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 06/20/2023]
Abstract
The review analyzes modern concepts about the control of various mechanisms of the hippocampal neuroplasticity in adult mammals and humans by glucocorticoids. Glucocorticoid hormones ensure the coordinated functioning of key components and mechanisms of hippocampal plasticity: neurogenesis, glutamatergic neurotransmission, microglia and astrocytes, systems of neurotrophic factors, neuroinflammation, proteases, metabolic hormones, neurosteroids. Regulatory mechanisms are diverse; along with the direct action of glucocorticoids through their receptors, there are conciliated glucocorticoid-dependent effects, as well as numerous interactions between various systems and components. Despite the fact that many connections in this complex regulatory scheme have not yet been established, the study of the factors and mechanisms considered in the work forms growth points in the field of glucocorticoid-regulated processes in the brain and primarily in the hippocampus. These studies are fundamentally important for the translation into the clinic and the potential treatment/prevention of common diseases of the emotional and cognitive spheres and respective comorbid conditions.
Collapse
Affiliation(s)
- Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, 115419, Russia
| |
Collapse
|
14
|
Johnston JN, Kadriu B, Allen J, Gilbert JR, Henter ID, Zarate CA. Ketamine and serotonergic psychedelics: An update on the mechanisms and biosignatures underlying rapid-acting antidepressant treatment. Neuropharmacology 2023; 226:109422. [PMID: 36646310 PMCID: PMC9983360 DOI: 10.1016/j.neuropharm.2023.109422] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
The discovery of ketamine as a rapid-acting antidepressant spurred significant research to understand its underlying mechanisms of action and to identify other novel compounds that may act similarly. Serotonergic psychedelics (SPs) have shown initial promise in treating depression, though the challenge of conducting randomized controlled trials with SPs and the necessity of long-term clinical observation are important limitations. This review summarizes the similarities and differences between the psychoactive effects associated with both ketamine and SPs and the mechanisms of action of these compounds, with a focus on the monoaminergic, glutamatergic, gamma-aminobutyric acid (GABA)-ergic, opioid, and inflammatory systems. Both molecular and neuroimaging aspects are considered. While their main mechanisms of action differ-SPs increase serotonergic signaling while ketamine is a glutamatergic modulator-evidence suggests that the downstream mechanisms of action of both ketamine and SPs include mechanistic target of rapamycin complex 1 (mTORC1) signaling and downstream GABAA receptor activity. The similarities in downstream mechanisms may explain why ketamine, and potentially SPs, exert rapid-acting antidepressant effects. However, research on SPs is still in its infancy compared to the ongoing research that has been conducted with ketamine. For both therapeutics, issues with regulation and proper controls should be addressed before more widespread implementation. This article is part of the Special Issue on "Ketamine and its Metabolites".
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Josh Allen
- The Alfred Centre, Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Ioline D Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
15
|
Antidepressants as a potential candidate to reduce microglia activation in neurodegenerative diseases. A systematic review and meta-analysis of preclinical studies. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2023. [DOI: 10.1016/j.jadr.2023.100465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
16
|
Ceasrine AM, Devlin BA, Bolton JL, Green LA, Jo YC, Huynh C, Patrick B, Washington K, Sanchez CL, Joo F, Campos-Salazar AB, Lockshin ER, Kuhn C, Murphy SK, Simmons LA, Bilbo SD. Maternal diet disrupts the placenta-brain axis in a sex-specific manner. Nat Metab 2022; 4:1732-1745. [PMID: 36443520 PMCID: PMC10507630 DOI: 10.1038/s42255-022-00693-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
High maternal weight is associated with detrimental outcomes in offspring, including increased susceptibility to neurological disorders such as anxiety, depression and communicative disorders. Despite widespread acknowledgement of sex biases in the development of these disorders, few studies have investigated potential sex-biased mechanisms underlying disorder susceptibility. Here, we show that a maternal high-fat diet causes endotoxin accumulation in fetal tissue, and subsequent perinatal inflammation contributes to sex-specific behavioural outcomes in offspring. In male offspring exposed to a maternal high-fat diet, increased macrophage Toll-like receptor 4 signalling results in excess microglial phagocytosis of serotonin (5-HT) neurons in the developing dorsal raphe nucleus, decreasing 5-HT bioavailability in the fetal and adult brains. Bulk sequencing from a large cohort of matched first-trimester human samples reveals sex-specific transcriptome-wide changes in placental and brain tissue in response to maternal triglyceride accumulation (a proxy for dietary fat content). Further, fetal brain 5-HT levels decrease as placental triglycerides increase in male mice and male human samples. These findings uncover a microglia-dependent mechanism through which maternal diet can impact offspring susceptibility for neuropsychiatric disorder development in a sex-specific manner.
Collapse
Affiliation(s)
- Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Jessica L Bolton
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Lauren A Green
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Young Chan Jo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Carolyn Huynh
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Bailey Patrick
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Kamryn Washington
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Cristina L Sanchez
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Faith Joo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | | | - Elana R Lockshin
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Cynthia Kuhn
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Leigh Ann Simmons
- Department of Human Ecology, Perinatal Origins of Disparities Center, University of California, Davis, Davis, CA, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
- Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
17
|
Extracellular Vesicles in Chronic Demyelinating Diseases: Prospects in Treatment and Diagnosis of Autoimmune Neurological Disorders. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111943. [PMID: 36431078 PMCID: PMC9693249 DOI: 10.3390/life12111943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Extracellular vesicles (EVs) represent membrane-enclosed structures that are likely to be secreted by all living cell types in the animal organism, including cells of peripheral (PNS) and central nervous systems (CNS). The ability to cross the blood-brain barrier (BBB) provides the possibility not only for various EV-loaded molecules to be delivered to the brain tissues but also for the CNS-to-periphery transmission of these molecules. Since neural EVs transfer proteins and RNAs are both responsible for functional intercellular communication and involved in the pathogenesis of neurodegenerative diseases, they represent attractive diagnostic and therapeutic targets. Here, we discuss EVs' role in maintaining the living organisms' function and describe deviations in EVs' structure and malfunctioning during various neurodegenerative diseases.
Collapse
|
18
|
Gonçalves de Andrade E, González Ibáñez F, Tremblay MÈ. Microglia as a Hub for Suicide Neuropathology: Future Investigation and Prevention Targets. Front Cell Neurosci 2022; 16:839396. [PMID: 35663424 PMCID: PMC9158339 DOI: 10.3389/fncel.2022.839396] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/22/2022] [Indexed: 12/27/2022] Open
Abstract
Suicide is a complex public health challenge associated worldwide with one death every 40 s. Research advances in the neuropathology of suicidal behaviors (SB) have defined discrete brain changes which may hold the key to suicide prevention. Physiological differences in microglia, the resident immune cells of the brain, are present in post-mortem tissue samples of individuals who died by suicide. Furthermore, microglia are mechanistically implicated in the outcomes of important risk factors for SB, including early-life adversity, stressful life events, and psychiatric disorders. SB risk factors result in inflammatory and oxidative stress activities which could converge to microglial synaptic remodeling affecting susceptibility or resistance to SB. To push further this perspective, in this Review we summarize current areas of opportunity that could untangle the functional participation of microglia in the context of suicide. Our discussion centers around microglial state diversity in respect to morphology, gene and protein expression, as well as function, depending on various factors, namely brain region, age, and sex.
Collapse
Affiliation(s)
- Elisa Gonçalves de Andrade
- Neuroscience Graduate Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Fernando González Ibáñez
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
19
|
Yang B, Ryu JS, Rim C, Shin JU, Kwon MS. Possible role of arginase 1 positive microglia on depressive/anxiety-like behaviors in atopic dermatitis mouse model. Arch Pharm Res 2022; 45:11-28. [DOI: 10.1007/s12272-022-01369-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
|
20
|
Haidar MA, Ibeh S, Shakkour Z, Reslan MA, Nwaiwu J, Moqidem YA, Sader G, Nickles RG, Babale I, Jaffa AA, Salama M, Shaito A, Kobeissy F. Crosstalk between Microglia and Neurons in Neurotrauma: An Overview of the Underlying Mechanisms. Curr Neuropharmacol 2022; 20:2050-2065. [PMID: 34856905 PMCID: PMC9886840 DOI: 10.2174/1570159x19666211202123322] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022] Open
Abstract
Microglia are the resident immune cells of the brain and play a crucial role in housekeeping and maintaining homeostasis of the brain microenvironment. Upon injury or disease, microglial cells become activated, at least partly, via signals initiated by injured neurons. Activated microglia, thereby, contribute to both neuroprotection and neuroinflammation. However, sustained microglial activation initiates a chronic neuroinflammatory response which can disturb neuronal health and disrupt communications between neurons and microglia. Thus, microglia-neuron crosstalk is critical in a healthy brain as well as during states of injury or disease. As most studies focus on how neurons and microglia act in isolation during neurotrauma, there is a need to understand the interplay between these cells in brain pathophysiology. This review highlights how neurons and microglia reciprocally communicate under physiological conditions and during brain injury and disease. Furthermore, the modes of microglia-neuron communication are exposed, focusing on cell-contact dependent signaling and communication by the secretion of soluble factors like cytokines and growth factors. In addition, it has been discussed that how microglia-neuron interactions could exert either beneficial neurotrophic effects or pathologic proinflammatory responses. We further explore how aberrations in microglia-neuron crosstalk may be involved in central nervous system (CNS) anomalies, namely traumatic brain injury (TBI), neurodegeneration, and ischemic stroke. A clear understanding of how the microglia-neuron crosstalk contributes to the pathogenesis of brain pathologies may offer novel therapeutic avenues of brain trauma treatment.
Collapse
Affiliation(s)
- Muhammad Ali Haidar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Judith Nwaiwu
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yomna Adel Moqidem
- Biotechnology Program, School of Science and Engineering, The American University in Cairo, Cairo, Egypt
| | - Georgio Sader
- Faculty of Medicine, University of Balamand, Balamand, Lebanon
| | - Rachel G. Nickles
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Ismail Babale
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Aneese A. Jaffa
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Mohamed Salama
- Institute of Global Health and Human Ecology (I-GHHE), The American University in Cairo, New Cairo 11835, Egypt
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Abdullah Shaito
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
21
|
Park SH, Lee YS, Yang HJ, Song GJ. Fluoxetine Potentiates Phagocytosis and Autophagy in Microglia. Front Pharmacol 2021; 12:770610. [PMID: 34899324 PMCID: PMC8662994 DOI: 10.3389/fphar.2021.770610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/07/2021] [Indexed: 11/18/2022] Open
Abstract
Fluoxetine is a classic antidepressant drug, and its immunomodulatory effects have recently been reported in many disease models. In addition, it has strong antineuroinflammatory effects in stroke and neurodegenerative animal models. However, the effect of fluoxetine on microglia phagocytosis and its molecular mechanisms have not yet been studied. In this study, we investigated whether fluoxetine has a regulatory effect on microglial function. Microglia cell lines and primary mouse microglia were treated with fluoxetine, and the production of inflammatory cytokines and neurotrophic factors and the phagocytosis of amyloid β were measured. Fluoxetine significantly attenuated the production of lipopolysaccharide-induced proinflammatory cytokines and oxidative stress in microglia. Fluoxetine also significantly potentiated microglia phagocytosis and autophagy. In addition, autophagy flux inhibitors attenuated fluoxetine-induced phagocytosis. In conclusion, fluoxetine induces autophagy and potentiates phagocytosis in microglia, which can be a novel molecular mechanism of the neuroinflammatory and neuroprotective effects of fluoxetine.
Collapse
Affiliation(s)
- Sung Hee Park
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Korea
| | - Young-Sun Lee
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Korea.,The Convergence Institute of Healthcare and Medical Science, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea
| | - Hyun-Jeong Yang
- Department of Integrative Biosciences, University of Brain Education, Cheonan, Korea
| | - Gyun Jee Song
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Korea.,The Convergence Institute of Healthcare and Medical Science, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea
| |
Collapse
|