1
|
Rinaldi A, Balietti M, Principi E, De Luca M, De Felice E, Narcisi FM, Vilardo L, Rosito M, Piacentini R, D'Alessandro G, D'Agnano I, Maggi L, Conti F, Limatola C, Catalano M. BV2-derived extracellular vesicles modulate microglia inflammatory profile, neuronal plasticity, and behavioural performances in late adult mice. Brain Behav Immun 2024; 122:58-74. [PMID: 39128568 DOI: 10.1016/j.bbi.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/24/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND During aging, both the brain and the immune system undergo a progressive impairment of physiological functions. Microglia, the immunocompetent cells of the central nervous system, shift towards a chronic mild inflammatory state that impacts brain homeostasis. Extracellular vesicles (EVs) released by microglia transport packages of molecular information that mirror the inflammatory status of donor cells and modulate the inflammatory phenotype of recipient microglia and other cell types. RESULTS We demonstrated that intranasal administration of EVs derived from microglial-like BV2 cells to late adult mice (16-20 months of age) shifts microglia toward a "juvenile" morphology affecting their inflammatory profile. Mice treated with BV2-derived EVs have a reduction of anxiety-like behavior and an increased spatial learning, with sex-dependent differences. Further, BV2-derived EVs increased neuronal plasticity both in male and female mice. These findings suggest the involvement of microglial cells in vesicles-mediated anti-aging effect. CONCLUSIONS Our data indicate that BV2-derived EVs could represent a resource to slow down age-dependent inflammation in the mouse brain.
Collapse
Affiliation(s)
- Arianna Rinaldi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Marta Balietti
- IRCCS INRCA, Center for Neurobiology of Aging, Via Birarelli 8, Ancona 60121, Italy
| | - Elisa Principi
- Università Politecnica delle Marche, Department of Experimental and Clinical Medicine, Via Tronto 10/a, Ancona 60126, Italy
| | | | - Eleonora De Felice
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Laura Vilardo
- Institute of Biomedical Technologies, CNR, 20054 Segrate, Italy
| | - Maria Rosito
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; Center for Life Nanoscience & Neuroscience Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli 1, Roma, Italy
| | - Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Igea D'Agnano
- Institute of Biomedical Technologies, CNR, 20054 Segrate, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Fiorenzo Conti
- Università Politecnica delle Marche, Department of Experimental and Clinical Medicine, Via Tronto 10/a, Ancona 60126, Italy; IRCCS INRCA, Center for Neurobiology of Aging, Via Birarelli 8, Ancona 60121, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia, Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.
| |
Collapse
|
2
|
Lee HY, Lee Y, Chung C, Park SI, Shin HJ, Joe EH, Lee SJ, Kim DW, Jo SH, Choi SY. The antipsychotic chlorpromazine reduces neuroinflammation by inhibiting microglial voltage-gated potassium channels. Glia 2024. [PMID: 39435609 DOI: 10.1002/glia.24629] [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: 03/28/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Neuroinflammation, the result of microglial activation, is associated with the pathogenesis of a wide range of psychiatric and neurological disorders. Recently, chlorpromazine (CPZ), a dopaminergic D2 receptor antagonist and schizophrenia therapy, was proposed to exert antiinflammatory effects in the central nervous system. Here, we report that the expression of Kv1.3 channel, which is abundant in T cells, is upregulated in microglia upon infection, and that CPZ specifically inhibits these channels to reduce neuroinflammation. In the mouse medial prefrontal cortex, we show that CPZ lessens Kv1.3 channel activity and reduces proinflammatory cytokine production. In mice treated with LPS, we found that CPZ was capable of alleviating both neuroinflammation and depression-like behavior. Our findings suggest that CPZ acts as a microglial Kv1.3 channel inhibitor and neuroinflammation modulator, thereby exerting therapeutic effects in neuroinflammatory psychiatric/neurological disorders.
Collapse
Affiliation(s)
- Hee-Yoon Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Young Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Chaelin Chung
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Seo-In Park
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Hyo Jung Shin
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sung Joong Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Dong Woon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
- Department of Oral Anatomy & Developmental Biology, Kyung Hee University College of Dentistry, Seoul, Republic of Korea
| | - Su-Hyun Jo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| |
Collapse
|
3
|
Christensen A, McGill CJ, Qian W, Pike CJ. Effects of obesogenic diet and 17β-estradiol in female mice with APOE 3/3, 3/4, and 4/4 genotypes. Front Aging Neurosci 2024; 16:1415072. [PMID: 39347015 PMCID: PMC11427389 DOI: 10.3389/fnagi.2024.1415072] [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: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The main genetic risk factor for Alzheimer's disease (AD) is the apolipoprotein E ε4 allele (APOE4). AD risk associated with APOE4 disproportionately affects women. Furthermore, human and rodent studies indicate that the cognitive deficits associated with APOE4 are greater in females. One modifiable AD risk factor is obesity during middle age. Given that approximately two-thirds of US adults are overweight, it is important to understand how obesity affects AD risk, how it interacts with APOE4, and the extent to which its detrimental effects can be mitigated with therapeutics. One intervention study for women is estrogen-based hormone therapy, which can exert numerous health benefits when administered in early middle age. No experimental studies have examined the interactions among APOE4, obesity, and hormone therapy in aging females. To begin to explore these issues, we considered how obesity outcomes are affected by treatment with estradiol at the onset of middle age in female mice with human APOE3 and APOE4. Furthermore, to explore how gene dosage affects outcomes, we compared mice homozygous for APOE3 (3/3) and homozygous (4/4) or hemizygous (3/4) for APOE4. Mice were examined over a 4-month period that spans the transition into reproductive senescence, a normal age-related change that models many aspects of human perimenopause. Beginning at 5 months of age, mice were maintained on a control diet (10% fat) or high-fat diet (HFD; 60% fat). After 8 weeks, by which time obesity was present in all HFD groups, mice were implanted with an estradiol or vehicle capsule that was maintained for the final 8 weeks. Animals were assessed on a range of metabolic and neural measures. Overall, APOE4 was associated with poorer metabolic function and cognitive performance. However, an obesogenic diet induced relatively greater impairments in metabolic function and cognitive performance in APOE3/3 mice. Estradiol treatment improved metabolic and cognitive outcomes across all HFD groups, with APOE4/4 generally exhibiting the greatest benefit. APOE3/4 mice were intermediate to the homozygous genotypes on many measures but also exhibited unique profiles. Together, these findings highlight the importance of the APOE genotype as a modulator of the risks associated with obesity and the beneficial outcomes of estradiol.
Collapse
Affiliation(s)
| | | | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
4
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. J Neuroinflammation 2024; 21:67. [PMID: 38481312 PMCID: PMC10938757 DOI: 10.1186/s12974-024-03059-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles during development as well as in health and disease. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., Box 8057, St. Louis, MO, 63110, USA.
| | - Haowu Jiang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., CB 8054, St. Louis, MO, 63110, USA.
| |
Collapse
|
5
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. Clin Immunol 2024:109921. [PMID: 38316202 DOI: 10.1016/j.clim.2024.109921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024]
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles in normal brain development, neurodegeneration, and brain cancers. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| | - Haowu Jiang
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine in St Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
6
|
Karperien AL, Jelinek HF. Morphology and Fractal-Based Classifications of Neurons and Microglia in Two and Three Dimensions. ADVANCES IN NEUROBIOLOGY 2024; 36:149-172. [PMID: 38468031 DOI: 10.1007/978-3-031-47606-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Microglia and neurons live physically intertwined, intimately related structurally and functionally in a dynamic relationship in which microglia change continuously over a much shorter timescale than do neurons. Although microglia may unwind and depart from the neurons they attend under certain circumstances, in general, together both contribute to the fractal topology of the brain that defines its computational capabilities. Both neuronal and microglial morphologies are well-described using fractal analysis complementary to more traditional measures. For neurons, the fractal dimension has proved valuable for classifying dendritic branching and other neuronal features relevant to pathology and development. For microglia, fractal geometry has substantially contributed to classifying functional categories, where, in general, the more pathological the biological status, the lower the fractal dimension for individual cells, with some exceptions, including hyper-ramification. This chapter provides a review of the intimate relationships between neurons and microglia, by introducing 2D and 3D fractal analysis methodology and its applications in neuron-microglia function in health and disease.
Collapse
Affiliation(s)
- Audrey L Karperien
- School of Community Health, Charles Sturt University, Albury, NSW, Australia
| | - Herbert F Jelinek
- Department of Medical Sciences and Biotechnology Center, Khalifa University, Abu Dhabi, UAE
| |
Collapse
|
7
|
Kim CS. Roles of Diet-Associated Gut Microbial Metabolites on Brain Health: Cell-to-Cell Interactions between Gut Bacteria and the Central Nervous System. Adv Nutr 2024; 15:100136. [PMID: 38436218 PMCID: PMC10694655 DOI: 10.1016/j.advnut.2023.10.008] [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: 03/16/2023] [Revised: 09/21/2023] [Accepted: 10/26/2023] [Indexed: 03/05/2024] Open
Abstract
Gut microbiota have crucial effects on brain function via the gut-brain axis. Growing evidence suggests that this interaction is mediated by signaling molecules derived from dietary components metabolized by the intestinal microbiota. Although recent studies have provided a substantial understanding of the cell-specific effects of gut microbial molecules in gut microbiome-brain research, further validation is needed. This review presents recent findings on gut microbiota-derived dietary metabolites that enter the systemic circulation and influence the cell-to-cell interactions between gut microbes and cells in the central nervous system (CNS), particularly microglia, astrocytes, and neuronal cells, ultimately affecting cognitive function, mood, and behavior. Specifically, this review highlights the roles of metabolites produced by the gut microbiota via dietary component transformation, including short-chain fatty acids, tryptophan metabolites, and bile acid metabolites, in promoting the function and maturation of brain cells and suppressing inflammatory signals in the CNS. We also discuss future directions for gut microbiome-brain research, focusing on diet-induced microbial metabolite-based therapies as possible novel approaches to mental health treatment.
Collapse
Affiliation(s)
- Chong-Su Kim
- Department of Food and Nutrition, College of Natural Information Sciences, Dongduk Women's University, Seoul 02748, Republic of Korea.
| |
Collapse
|
8
|
Karperien AL, Jelinek HF. Box-Counting Fractal Analysis: A Primer for the Clinician. ADVANCES IN NEUROBIOLOGY 2024; 36:15-55. [PMID: 38468026 DOI: 10.1007/978-3-031-47606-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
This chapter lays out the elementary principles of fractal geometry underpinning much of the rest of this book. It assumes a minimal mathematical background, defines the key principles and terms in context, and outlines the basics of a fractal analysis method known as box counting and how it is used to perform fractal, lacunarity, and multifractal analyses. As a standalone reference, this chapter grounds the reader to be able to understand, evaluate, and apply essential methods to appreciate and heal the exquisitely detailed fractal geometry of the brain.
Collapse
Affiliation(s)
| | - Herbert F Jelinek
- Department of Medical Sciences and Biotechnology Center, Khalifa University, Abu Dhabi, UAE
| |
Collapse
|
9
|
Boylan BT, Hwang M, Bergmann CC. The Impact of Innate Components on Viral Pathogenesis in the Neurotropic Coronavirus Encephalomyelitis Mouse Model. Viruses 2023; 15:2400. [PMID: 38140641 PMCID: PMC10747027 DOI: 10.3390/v15122400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Recognition of viruses invading the central nervous system (CNS) by pattern recognition receptors (PRRs) is crucial to elicit early innate responses that stem dissemination. These innate responses comprise both type I interferon (IFN-I)-mediated defenses as well as signals recruiting leukocytes to control the infection. Focusing on insights from the neurotropic mouse CoV model, this review discusses how early IFN-I, fibroblast, and myeloid signals can influence protective anti-viral adaptive responses. Emphasis is placed on three main areas: the importance of coordinating the distinct capacities of resident CNS cells to induce and respond to IFN-I, the effects of select IFN-stimulated genes (ISGs) on host immune responses versus viral control, and the contribution of fibroblast activation and myeloid cells in aiding the access of T cells to the parenchyma. By unraveling how the dysregulation of early innate components influences adaptive immunity and viral control, this review illustrates the combined effort of resident CNS cells to achieve viral control.
Collapse
Affiliation(s)
- Brendan T. Boylan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44196, USA; (B.T.B.); (M.H.)
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mihyun Hwang
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44196, USA; (B.T.B.); (M.H.)
- Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Cornelia C. Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44196, USA; (B.T.B.); (M.H.)
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
- Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
- School of Biological Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
10
|
Machado MMF, Ático EM, Banin RM, Hirata BKS, Kempe PRG, Pedroso AP, Thomaz FM, Oyama LM, Ribeiro EB, Bueno AA, Cerutti SM, Telles MM. Ginkgo biloba extract modulates astrocytic and microglial recruitment in the hippocampus and hypothalamus of menopause-induced ovariectomized rats. Brain Res 2023; 1822:148659. [PMID: 39492489 DOI: 10.1016/j.brainres.2023.148659] [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: 08/01/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
BACKGROUND Changes in steroid hormone levels associated with menopause are known to affect body composition, with increased accumulation of visceral fat and impaired actions of appetite-regulating neuropeptides. Anti-obesogenic, antioxidant, anti-inflammatory and neuromodulatory properties have been attributed to Ginkgo biloba extract (GbE) oral supplementation. HYPOTHESIS/PURPOSE We investigated in menopause-induced ovariectomized rats the effects of GbE oral supplementation on microglial reactivity and astrocyte recruitment in hippocampal and hypothalamic subregions involved in the regulation of feeding behavior and energy homeostasis. STUDY DESIGN/METHODS Ovariectomy (Ovx) or false-Ovx (Sham) surgery were performed in 2-month-old female Wistar rats. Sixty days after surgery, Ovx rats were gavaged daily for 14 days with either saline (Ovx + Veh) or GbE 500 mg/Kg (Ovx + GbE). Rats were subsequently sacrificed, brains harvested and subjected to immunohistochemistry and immunofluorescence analyses. RESULTS Ovx increased microglial reactivity in CA1, CA3 and dentate gyrus (DG) in the dorsal hippocampal formation (dHF), as well as in DG in the ventral hippocampal formation (vHF). Additionally, Ovx reduced astrocyte count in dHF CA3. The disturbances found in Ovx + Veh versus Sham were not found in Ovx + GbE versus Sham. Furthermore, higher astrocyte counts in DG of both dHF and vHF were found in Ovx + GbE as compared to Ovx + Veh. In the hypothalamus, Ovx + Veh showed reduced microglial reactivity in the arcuate (ARC) and ventromedial (VMH) nuclei as compared to Ovx + GbE. Ovx + GbE rats presented higher astrocyte counts in ARC compared to Sham rats. CONCLUSION Our results show for the first time in a rodent model of menopause that GbE supplementation modulates astrocyte and microglial recruitment and reactivity in hippocampal and hypothalamic subregions involved in feeding behavior and energy homeostasis. Future research employing other experimental models may further elucidate whether GbE supplementation possesses therapeutic properties upon glial cell reactivity to potentially alleviate changes in energy homeostasis associated with menopause.
Collapse
Affiliation(s)
- Meira M F Machado
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Esther M Ático
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Renata M Banin
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bruna K S Hirata
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Paula R G Kempe
- Laboratory of Nerve Regeneration, Universidade de Campinas, Campinas, Brazil
| | - Amanda P Pedroso
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Thomaz
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Lila M Oyama
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Eliane B Ribeiro
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Worcester WR2 6AJ, United Kingdom.
| | - Suzete M Cerutti
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Mônica M Telles
- Post-graduate Program in Chemical Biology, Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil; Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Hasegawa Y, Kim J, Ursini G, Jouroukhin Y, Zhu X, Miyahara Y, Xiong F, Madireddy S, Obayashi M, Lutz B, Sawa A, Brown SP, Pletnikov MV, Kamiya A. Microglial cannabinoid receptor type 1 mediates social memory deficits in mice produced by adolescent THC exposure and 16p11.2 duplication. Nat Commun 2023; 14:6559. [PMID: 37880248 PMCID: PMC10600150 DOI: 10.1038/s41467-023-42276-5] [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: 03/13/2022] [Accepted: 10/04/2023] [Indexed: 10/27/2023] Open
Abstract
Adolescent cannabis use increases the risk for cognitive impairments and psychiatric disorders. Cannabinoid receptor type 1 (Cnr1) is expressed not only in neurons and astrocytes, but also in microglia, which shape synaptic connections during adolescence. However, the role of microglia in mediating the adverse cognitive effects of delta-9-tetrahydrocannabinol (THC), the principal psychoactive constituent of cannabis, is not fully understood. Here, we report that in mice, adolescent THC exposure produces microglial apoptosis in the medial prefrontal cortex (mPFC), which was exacerbated in a model of 16p11.2 duplication, a representative copy number variation (CNV) risk factor for psychiatric disorders. These effects are mediated by microglial Cnr1, leading to reduction in the excitability of mPFC pyramidal-tract neurons and deficits in social memory in adulthood. Our findings suggest the microglial Cnr1 may contribute to adverse effect of cannabis exposure in genetically vulnerable individuals.
Collapse
Affiliation(s)
- Yuto Hasegawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juhyun Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Gianluca Ursini
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Yan Jouroukhin
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences SUNY, University at Buffalo, Buffalo, NY, USA
| | - Xiaolei Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Miyahara
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Feiyi Xiong
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samskruthi Madireddy
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mizuho Obayashi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR) gGmbH, Mainz, Germany
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Solange P Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences SUNY, University at Buffalo, Buffalo, NY, USA.
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Baek SY, Lee J, Kim T, Lee H, Choi HS, Park H, Koh M, Kim E, Jung ME, Iliopoulos D, Lee JY, Kim J, Lee S. Development of a novel histone deacetylase inhibitor unveils the role of HDAC11 in alleviating depression by inhibition of microglial activation. Biomed Pharmacother 2023; 166:115312. [PMID: 37567072 DOI: 10.1016/j.biopha.2023.115312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023] Open
Abstract
Histone deacetylases (HDACs) are key epigenetic regulators and classified into four subtypes. Despite the various roles of each HDAC isoform, the lack of selective HDAC inhibitors has limited the elucidation of their roles in biological systems. HDAC11, the sole class-IV HDAC, is highly expressed in the brain, however, the role of HDAC11 in microglia is not fully understood. Based on the modification of MC1568, we developed a novel HDAC inhibitor, 5. Interestingly, 5 suppresses lipopolysaccharide-induced microglial activation by the initiation of autophagy and subsequent inhibition of nitric oxide production. Furthermore, we demonstrated that 5 significantly alleviates depression-like behavior by inhibiting microglial activation in mouse brain. Our discovery reveals that specific pharmacological regulation of HDAC11 induces autophagy and reactive nitrogen species balance in microglia for the first time, which makes HDAC11 a new therapeutic target for depressive disorder.
Collapse
Affiliation(s)
- Soo Yeon Baek
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Jeehee Lee
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Department of HY-KIST Bio-convergence, Hanyang University, Seoul 04763, South Korea
| | - Taegwan Kim
- Department of Chemistry and Integrative Institute of Basic Science, Soongsil University, Seoul 06978, South Korea
| | - Hyelim Lee
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Hoon-Seong Choi
- Research Animal Resources Center, Research Resources Division, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Hahnbeom Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Minseob Koh
- Department of Chemistry, Pusan National University, Busan 46241, South Korea
| | - Eunha Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Michael E Jung
- Department of Chemistry & Biochemistry, University of California at Los Angeles (UCLA), Los Angeles, CA 90095-1569, USA
| | - Dimitrios Iliopoulos
- Center for Systems Biomedicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Jeong-Yeon Lee
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, South Korea
| | - Jonghoon Kim
- Department of Chemistry and Integrative Institute of Basic Science, Soongsil University, Seoul 06978, South Korea.
| | - Sanghee Lee
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Department of HY-KIST Bio-convergence, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
13
|
Wang Z, Lipshutz A, Liu ZL, Trzeciak AJ, Miranda IC, Martínez de la Torre C, Schild T, Lazarov T, Rojas WS, Saavedra PHV, Romero-Pichardo JE, Baako A, Geissmann F, Faraco G, Gan L, Etchegaray JI, Lucas CD, Parkhurst CN, Zeng MY, Keshari KR, Perry JSA. Early life high fructose exposure disrupts microglia function and impedes neurodevelopment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553242. [PMID: 37645894 PMCID: PMC10462086 DOI: 10.1101/2023.08.14.553242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Despite the success of fructose as a low-cost food additive, recent epidemiological evidence suggests that high fructose consumption by pregnant mothers or during adolescence is associated with disrupted neurodevelopment 1-7 . An essential step in appropriate mammalian neurodevelopment is the synaptic pruning and elimination of newly-formed neurons by microglia, the central nervous system's (CNS) resident professional phagocyte 8-10 . Whether early life high fructose consumption affects microglia function and if this directly impacts neurodevelopment remains unknown. Here, we show that both offspring born to dams fed a high fructose diet and neonates exposed to high fructose exhibit decreased microglial density, increased uncleared apoptotic cells, and decreased synaptic pruning in vivo . Importantly, deletion of the high affinity fructose transporter SLC2A5 (GLUT5) in neonates completely reversed microglia dysfunction, suggesting that high fructose directly affects neonatal development. Mechanistically, we found that high fructose treatment of both mouse and human microglia suppresses synaptic pruning and phagocytosis capacity which is fully reversed in GLUT5-deficient microglia. Using a combination of in vivo and in vitro nuclear magnetic resonance- and mass spectrometry-based fructose tracing, we found that high fructose drives significant GLUT5-dependent fructose uptake and catabolism, rewiring microglia metabolism towards a hypo-phagocytic state. Importantly, mice exposed to high fructose as neonates exhibited cognitive defects and developed anxiety-like behavior which were rescued in GLUT5-deficient animals. Our findings provide a mechanistic explanation for the epidemiological observation that early life high fructose exposure is associated with increased prevalence of adolescent anxiety disorders.
Collapse
|
14
|
Li Z, Zhang Z, Zhang Z, Wang Z, Li H. Cognitive impairment after long COVID-19: current evidence and perspectives. Front Neurol 2023; 14:1239182. [PMID: 37583958 PMCID: PMC10423939 DOI: 10.3389/fneur.2023.1239182] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/12/2023] [Indexed: 08/17/2023] Open
Abstract
COVID-19, caused by the SARS-CoV-2 virus, is a respiratory infectious disease. While most patients recover after treatment, there is growing evidence that COVID-19 may result in cognitive impairment. Recent studies reveal that some individuals experience cognitive deficits, such as diminished memory and attention, as well as sleep disturbances, suggesting that COVID-19 could have long-term effects on cognitive function. Research indicates that COVID-19 may contribute to cognitive decline by damaging crucial brain regions, including the hippocampus and anterior cingulate cortex. Additionally, studies have identified active neuroinflammation, mitochondrial dysfunction, and microglial activation in COVID-19 patients, implying that these factors may be potential mechanisms leading to cognitive impairment. Given these findings, the possibility of cognitive impairment following COVID-19 treatment warrants careful consideration. Large-scale follow-up studies are needed to investigate the impact of COVID-19 on cognitive function and offer evidence to support clinical treatment and rehabilitation practices. In-depth neuropathological and biological studies can elucidate precise mechanisms and provide a theoretical basis for prevention, treatment, and intervention research. Considering the risks of the long-term effects of COVID-19 and the possibility of reinfection, it is imperative to integrate basic and clinical research data to optimize the preservation of patients' cognitive function and quality of life. This integration will also offer valuable insights for responding to similar public health events in the future. This perspective article synthesizes clinical and basic evidence of cognitive impairment following COVID-19, discussing potential mechanisms and outlining future research directions.
Collapse
Affiliation(s)
- Zhitao Li
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuoya Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiyong Wang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Li
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Hasegawa Y, Kim J, Ursini G, Jouroukhin Y, Zhu X, Miyahara Y, Xiong F, Madireddy S, Obayashi M, Lutz B, Sawa A, Brown SP, Pletnikov MV, Kamiya A. Microglial cannabinoid receptor type 1 mediates social memory deficits produced by adolescent THC exposure and 16p11.2 duplication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550212. [PMID: 37546830 PMCID: PMC10402026 DOI: 10.1101/2023.07.24.550212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Adolescent cannabis use increases the risk for cognitive impairments and psychiatric disorders. Cannabinoid receptor type 1 (Cnr1) is expressed not only in neurons and astrocytes, but also in microglia, which shape synaptic connections during adolescence. Nonetheless, until now, the role of microglia in mediating the adverse cognitive effects of delta-9-tetrahydrocannabinol (THC), the principal psychoactive constituent of cannabis, has been unexplored. Here, we report that adolescent THC exposure produces microglial apoptosis in the medial prefrontal cortex (mPFC), which was exacerbated in the mouse model of 16p11.2 duplication, a representative copy number variation (CNV) risk factor for psychiatric disorders. These effects are mediated by microglial Cnr1, leading to reduction in the excitability of mPFC pyramidal-tract neurons and deficits in social memory in adulthood. Our findings highlight the importance of microglial Cnr1 to produce the adverse effect of cannabis exposure in genetically vulnerable individuals.
Collapse
|
16
|
Santos RPC, Toscano ECDB, Rachid MA. Anti-inflammatory strategies for hepatic encephalopathy: preclinical studies. ARQUIVOS DE NEURO-PSIQUIATRIA 2023. [PMID: 37487550 PMCID: PMC10371400 DOI: 10.1055/s-0043-1767819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Hepatic encephalopathy (HE) is a potentially reversible neuropsychiatric syndrome. Often, HE causes cognitive and motor dysfunctions due to an acute or chronic insufficiency of the liver or a shunting between the hepatic portal vein and systemic vasculature. Liver damage induces peripheral changes, such as in the metabolism and peripheral inflammatory responses that trigger exacerbated neuroinflammation. In experimental models, anti-inflammatory strategies have demonstrated neuroprotective effects, leading to a reduction in HE-related cognitive and motor impairments. In this scenario, a growing body of evidence has shown that peripheral and central nervous system inflammation are promising preclinical targets. In this review, we performed an overview of FDA-approved drugs and natural compounds which are used in the treatment of other neurological and nonneurological diseases that have played a neuroprotective role in experimental HE, at least in part, through anti-inflammatory mechanisms. Despite the exciting results from animal models, the available data should be critically interpreted, highlighting the importance of translating the findings for clinical essays.
Collapse
Affiliation(s)
- Rafaela Pinto Coelho Santos
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Laboratório de Patologia Celular e Molecular, Belo Horizonte MG, Brazil
| | - Eliana Cristina de Brito Toscano
- Universidade Federal de Juiz de Fora, Faculdade de Medicina, Departamento de Patologia, Laboratório Integrado de Pesquisa em Patologia, Juiz de Fora MG, Brazil
- Universidade Federal de Juiz e Fora, Faculdade de Medicina, Programa de Pós-Graduação em Saúde, Juiz de Fora MG, Brazil
| | - Milene Alvarenga Rachid
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Laboratório de Patologia Celular e Molecular, Belo Horizonte MG, Brazil
| |
Collapse
|
17
|
Dehdar K, Salimi M, Tabasi F, Dehghan S, Sumiyoshi A, Garousi M, Jamaati H, Javan M, Reza Raoufy M. Allergen induces depression-like behavior in association with altered prefrontal-hippocampal circuit in male rats. Neuroscience 2023:S0306-4522(23)00254-3. [PMID: 37286161 DOI: 10.1016/j.neuroscience.2023.05.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 05/27/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
Allergic asthma is a common chronic inflammatory condition associated with psychiatric comorbidities. Notably depression, correlated with adverse outcomes in asthmatic patients. Peripheral inflammation's role in depression has been shown previously. However, evidence regarding the effects of allergic asthma on the medial prefrontal cortex (mPFC)-ventral hippocampus (vHipp) interactions, an important neurocircuitry in affective regulation, is yet to be demonstrated. Herein, we investigated the effects of allergen exposure in sensitized rats on the immunoreactivity of glial cells, depression-like behavior, brain regions volume, as well as activity and connectivity of the mPFC-vHipp circuit. We found that allergen-induced depressive-like behavior was associated with more activated microglia and astrocytes in mPFC and vHipp, as well as reduced hippocampus volume. Intriguingly, depressive-like behavior was negatively correlated with mPFC and hippocampus volumes in the allergen-exposed group. Moreover, mPFC and vHipp activity were altered in asthmatic animals. Allergen disrupted the strength and direction of functional connectivity in the mPFC-vHipp circuit so that, unlike normal conditions, mPFC causes and modulates vHipp activity. Our results provide new insight into the underlying mechanism of allergic inflammation-induced psychiatric disorders, aiming to develop new interventions and therapeutic approaches for improving asthma complications.
Collapse
Affiliation(s)
- Kolsoum Dehdar
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Salimi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farhad Tabasi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran; Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Akira Sumiyoshi
- Institute of Development, Aging and Cancer, Tohoku University, Seiryo-machi, Aoba-ku, Sendai, Japan; National Institutes for Quantum and Radiological Science and Technology, Anagawa, Inage-ku, Chiba, Japan
| | - Mani Garousi
- Department of Electrical and Engineering, Tarbiat Modares University, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
18
|
Schwabenland M, Mossad O, Sievert A, Peres AG, Ringel E, Baasch S, Kolter J, Cascone G, Dokalis N, Vlachos A, Ruzsics Z, Henneke P, Prinz M, Blank T. Neonatal immune challenge poses a sex-specific risk for epigenetic microglial reprogramming and behavioral impairment. Nat Commun 2023; 14:2721. [PMID: 37169749 PMCID: PMC10175500 DOI: 10.1038/s41467-023-38373-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/27/2023] [Indexed: 05/13/2023] Open
Abstract
While the precise processes underlying a sex bias in the development of central nervous system (CNS) disorders are unknown, there is growing evidence that an early life immune activation can contribute to the disease pathogenesis. When we mimicked an early systemic viral infection or applied murine cytomegalovirus (MCMV) systemically in neonatal female and male mice, only male adolescent mice presented behavioral deficits, including reduced social behavior and cognition. This was paralleled by an increased amount of infiltrating T cells in the brain parenchyma, enhanced interferon-γ (IFNγ) signaling, and epigenetic reprogramming of microglial cells. These microglial cells showed increased phagocytic activity, which resulted in abnormal loss of excitatory synapses within the hippocampal brain region. None of these alterations were seen in female adolescent mice. Our findings underscore the early postnatal period's susceptibility to cause sex-dependent long-term CNS deficiencies following infections.
Collapse
Affiliation(s)
- Marius Schwabenland
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Omar Mossad
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Annika Sievert
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adam G Peres
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elena Ringel
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Baasch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Giulia Cascone
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nikolaos Dokalis
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Zsolt Ruzsics
- Institute for Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
19
|
Blank T, Prinz M. Objection non-responsive! How maternal immune activation in pregnancy weakens subsequent microglial immune response. Cell Res 2023; 33:193-194. [PMID: 36446893 PMCID: PMC9977840 DOI: 10.1038/s41422-022-00756-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Affiliation(s)
- Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Center for NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
20
|
Astrocytes regulate neuronal network activity by mediating synapse remodeling. Neurosci Res 2023; 187:3-13. [PMID: 36170922 DOI: 10.1016/j.neures.2022.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/22/2022]
Abstract
Based on experience during our life, neuronal connectivity continuously changes through structural remodeling of synapses. Recent studies have shown that the complex interaction between astrocytes and synapses regulates structural synapse remodeling by inducing the formation and elimination of synapses, as well as their functional maturation. Defects in this astrocyte-mediated synapse remodeling cause problems in not only neuronal network activities but also animal behaviors. Moreover, in various neurological disorders, astrocytes have been shown to play central roles in the initiation and progression of synaptic pathophysiology through impaired interactions with synapses. In this review, we will discuss recent studies identifying the novel roles of astrocytes in neuronal circuit remodeling, focusing on synapse formation and elimination. We will also discuss the potential implication of defective astrocytic function in evoking various brain disorders.
Collapse
|
21
|
Repositioning doxycycline for treating synucleinopathies: Evidence from a pre-clinical mouse model. Parkinsonism Relat Disord 2023; 106:105229. [PMID: 36462409 DOI: 10.1016/j.parkreldis.2022.105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/02/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Parkinson's disease remains orphan of valuable therapies capable to interfere with the disease pathogenesis despite the large number of symptomatic approaches adopted in clinical practice to manage this disease. Treatments simultaneously affecting α-synuclein (α-syn) oligomerization and neuroinflammation may counteract Parkinson's disease and related disorders. Recent data demonstrate that Doxycycline, a tetracycline antibiotic, can inhibit α-syn aggregation as well as neuroinflammation. We herein investigate, for the first time, the potential therapeutic properties of Doxy in a human α-syn A53T transgenic Parkinson's disease mouse model evaluating behavioural, biochemical and histopathological parameters. EXPERIMENTAL APPROACH Human α-syn A53T transgenic mice were treated with Doxycycline (10 mg/kg daily ip) for 30 days. The effect of treatment on motor, cognitive and daily live activity performances were examined. Neuropathological and neurophysiological parameters were assessed through immunocytochemical, electrophysiological and biochemical analysis of cerebral tissue. KEY RESULTS Doxy treatment abolished cognitive and daily life activity deficiencies in A53T mice. The effect on cognitive functions was associated with neuroprotection, inhibition of α-syn oligomerization and gliosis both in the cortex and hippocampus. Doxy treatment restored hippocampal long-term potentiation in association with the inhibition of pro-inflammatory cytokines expression. Moreover, Doxy ameliorated motor impairment and reduced striatal glial activation in A53T mice. CONCLUSIONS AND IMPLICATIONS Our findings promote Doxy as a valuable multi-target therapeutic approach counteracting both symptoms and neuropathology in the complex scenario of α-synucleinopathies.
Collapse
|
22
|
The Role of Gut Dysbiosis in the Pathophysiology of Neuropsychiatric Disorders. Cells 2022; 12:cells12010054. [PMID: 36611848 PMCID: PMC9818777 DOI: 10.3390/cells12010054] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Mounting evidence shows that the complex gut microbial ecosystem in the human gastrointestinal (GI) tract regulates the physiology of the central nervous system (CNS) via microbiota and the gut-brain (MGB) axis. The GI microbial ecosystem communicates with the brain through the neuroendocrine, immune, and autonomic nervous systems. Recent studies have bolstered the involvement of dysfunctional MGB axis signaling in the pathophysiology of several neurodegenerative, neurodevelopmental, and neuropsychiatric disorders (NPDs). Several investigations on the dynamic microbial system and genetic-environmental interactions with the gut microbiota (GM) have shown that changes in the composition, diversity and/or functions of gut microbes (termed "gut dysbiosis" (GD)) affect neuropsychiatric health by inducing alterations in the signaling pathways of the MGB axis. Interestingly, both preclinical and clinical evidence shows a positive correlation between GD and the pathogenesis and progression of NPDs. Long-term GD leads to overstimulation of hypothalamic-pituitary-adrenal (HPA) axis and the neuroimmune system, along with altered neurotransmitter levels, resulting in dysfunctional signal transduction, inflammation, increased oxidative stress (OS), mitochondrial dysfunction, and neuronal death. Further studies on the MGB axis have highlighted the significance of GM in the development of brain regions specific to stress-related behaviors, including depression and anxiety, and the immune system in the early life. GD-mediated deregulation of the MGB axis imbalances host homeostasis significantly by disrupting the integrity of the intestinal and blood-brain barrier (BBB), mucus secretion, and gut immune and brain immune functions. This review collates evidence on the potential interaction between GD and NPDs from preclinical and clinical data. Additionally, we summarize the use of non-therapeutic modulators such as pro-, pre-, syn- and post-biotics, and specific diets or fecal microbiota transplantation (FMT), which are promising targets for the management of NPDs.
Collapse
|
23
|
Brisch R, Wojtylak S, Saniotis A, Steiner J, Gos T, Kumaratilake J, Henneberg M, Wolf R. The role of microglia in neuropsychiatric disorders and suicide. Eur Arch Psychiatry Clin Neurosci 2022; 272:929-945. [PMID: 34595576 PMCID: PMC9388452 DOI: 10.1007/s00406-021-01334-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
This narrative review examines the possible role of microglial cells, first, in neuroinflammation and, second, in schizophrenia, depression, and suicide. Recent research on the interactions between microglia, astrocytes and neurons and their involvement in pathophysiological processes of neuropsychiatric disorders is presented. This review focuses on results from postmortem, positron emission tomography (PET) imaging studies, and animal models of schizophrenia and depression. Third, the effects of antipsychotic and antidepressant drug therapy, and of electroconvulsive therapy on microglial cells are explored and the upcoming development of therapeutic drugs targeting microglia is described. Finally, there is a discussion on the role of microglia in the evolutionary progression of human lineage. This view may contribute to a new understanding of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ralf Brisch
- Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Szymon Wojtylak
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Arthur Saniotis
- Department of Anthropology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- Department of Pharmacy, Knowledge University, Erbil, Kurdistan Region, Iraq
| | - Johann Steiner
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke-University, Magdeburg, Germany
| | - Tomasz Gos
- Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Jaliya Kumaratilake
- Biological Anthropology and Comparative Anatomy Research Unit, Medical School, The University of Adelaide, Adelaide, Australia
| | - Maciej Henneberg
- Biological Anthropology and Comparative Anatomy Research Unit, Medical School, The University of Adelaide, Adelaide, Australia
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Rainer Wolf
- Department of Nursing and Health, Hochschule Fulda, University of Applied Sciences, Fulda, Germany.
| |
Collapse
|
24
|
Palmas MF, Etzi M, Pisanu A, Camoglio C, Sagheddu C, Santoni M, Manchinu MF, Pala M, Fusco G, De Simone A, Picci L, Mulas G, Spiga S, Scherma M, Fadda P, Pistis M, Simola N, Carboni E, Carta AR. The Intranigral Infusion of Human-Alpha Synuclein Oligomers Induces a Cognitive Impairment in Rats Associated with Changes in Neuronal Firing and Neuroinflammation in the Anterior Cingulate Cortex. Cells 2022; 11:cells11172628. [PMID: 36078036 PMCID: PMC9454687 DOI: 10.3390/cells11172628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/03/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a complex pathology causing a plethora of non-motor symptoms besides classical motor impairments, including cognitive disturbances. Recent studies in the PD human brain have reported microgliosis in limbic and neocortical structures, suggesting a role for neuroinflammation in the development of cognitive decline. Yet, the mechanism underlying the cognitive pathology is under investigated, mainly for the lack of a valid preclinical neuropathological model reproducing the disease’s motor and non-motor aspects. Here, we show that the bilateral intracerebral infusion of pre-formed human alpha synuclein oligomers (H-αSynOs) within the substantia nigra pars compacta (SNpc) offers a valid model for studying the cognitive symptoms of PD, which adds to the classical motor aspects previously described in the same model. Indeed, H-αSynOs-infused rats displayed memory deficits in the two-trial recognition task in a Y maze and the novel object recognition (NOR) test performed three months after the oligomer infusion. In the anterior cingulate cortex (ACC) of H-αSynOs-infused rats the in vivo electrophysiological activity was altered and the expression of the neuron-specific immediate early gene (IEG) Npas4 (Neuronal PAS domain protein 4) and the AMPA receptor subunit GluR1 were decreased. The histological analysis of the brain of cognitively impaired rats showed a neuroinflammatory response in cognition-related regions such as the ACC and discrete subareas of the hippocampus, in the absence of any evident neuronal loss, supporting a role of neuroinflammation in cognitive decline. We found an increased GFAP reactivity and the acquisition of a proinflammatory phenotype by microglia, as indicated by the increased levels of microglial Tumor Necrosis Factor alpha (TNF-α) as compared to vehicle-infused rats. Moreover, diffused deposits of phospho-alpha synuclein (p-αSyn) and Lewy neurite-like aggregates were found in the SNpc and striatum, suggesting the spreading of toxic protein within anatomically interconnected areas. Altogether, we present a neuropathological rat model of PD that is relevant for the study of cognitive dysfunction featuring the disease. The intranigral infusion of toxic oligomeric species of alpha-synuclein (α-Syn) induced spreading and neuroinflammation in distant cognition-relevant regions, which may drive the altered neuronal activity underlying cognitive deficits.
Collapse
Affiliation(s)
| | - Michela Etzi
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Augusta Pisanu
- National Research Council, Institute of Neuroscience, 09040 Cagliari, Italy
| | - Chiara Camoglio
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Michele Santoni
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Maria Francesca Manchinu
- Istituto Di Ricerca Genetica e Biomedica Del Consiglio Nazionale Delle Ricerche, 09040 Monserrato, Italy
| | - Mauro Pala
- Istituto Di Ricerca Genetica e Biomedica Del Consiglio Nazionale Delle Ricerche, 09040 Monserrato, Italy
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Alfonso De Simone
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Luca Picci
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Maria Scherma
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Paola Fadda
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
- Correspondence:
| |
Collapse
|
25
|
Yan L, Yang J, Yu M, Sun W, Han Y, Lu X, Jin C, Wu S, Cai Y. Lanthanum Impairs Learning and Memory by Activating Microglia in the Hippocampus of Mice. Biol Trace Elem Res 2022; 200:1640-1649. [PMID: 35178682 DOI: 10.1007/s12011-021-02637-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
Lanthanum can induce neurotoxicity and impair cognitive function; therefore, research on the mechanism by which the ability to learning and memory is decreased by lanthanum is vitally important for protecting health. Microglia are a type of neuroglia located throughout the brain and spinal cord that play an important role in the central nervous system. When overactive, these cells can cause the excessive production of inflammatory cytokines that can damage neighboring neurons. The purpose of this study was to explore the effect of lanthanum in the form of lanthanum chloride (LaCl3) on learning and the memory of mice and determine whether there is a relationship between hippocampal neurons or learning and memory damage and excessive production of inflammatory cytokines. Four groups of pregnant Chinese Kun Ming mice were exposed to 0, 18, 36, or 72 mM LaCl3 in their drinking water during lactation. The offspring were then exposed to LaCl3 in the breast milk at birth until weaning and then exposed to these concentrations in their drinking water for 2 months after weaning. The results showed that LaCl3 impaired learning and memory in mice and injured their neurons, activated the microglia, and significantly overregulated the mRNA and protein expression of tumor necrosis factor alpha, interleukin (IL)-1β, IL-6, monocyte chemoattractant protein-1, and nitric oxide in the hippocampus. The results of this study suggest that lanthanum can impair learning and memory in mice, possibly by over-activating the microglia.
Collapse
Affiliation(s)
- Licheng Yan
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
- Department of Toxicology, School of Public Health, North China University of Science and Technology, No.21 Bohai road, Caofeidian New Area, Tangshan, 063210, Hebei province, People's Republic of China
| | - Jinghua Yang
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Miao Yu
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Wenchang Sun
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Yarao Han
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Shengwen Wu
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China
| | - Yuan Cai
- Department of Toxicology, School of Public Health, China Medical University, No.77 Puhe road, Shenyang North New Area, Shenyang, 110122, Liaoning province, People's Republic of China.
| |
Collapse
|
26
|
Tan Y, Chu Z, Shan H, Zhangsun D, Zhu X, Luo S. Inflammation Regulation via an Agonist and Antagonists of α7 Nicotinic Acetylcholine Receptors in RAW264.7 Macrophages. Mar Drugs 2022; 20:md20030200. [PMID: 35323499 PMCID: PMC8955479 DOI: 10.3390/md20030200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 02/01/2023] Open
Abstract
The α7 nicotinic acetylcholine receptor (nAChR) is widely distributed in the central and peripheral nervous systems and is closely related to a variety of nervous system diseases and inflammatory responses. The α7 nAChR subtype plays a vital role in the cholinergic anti-inflammatory pathway. In vivo, ACh released from nerve endings stimulates α7 nAChR on macrophages to regulate the NF-κB and JAK2/STAT3 signaling pathways, thereby inhibiting the production and release of downstream proinflammatory cytokines and chemokines. Despite a considerable level of recent research on α7 nAChR-mediated immune responses, much is still unknown. In this study, we used an agonist (PNU282987) and antagonists (MLA and α-conotoxin [A10L]PnIA) of α7 nAChR as pharmacological tools to identify the molecular mechanism of the α7 nAChR-mediated cholinergic anti-inflammatory pathway in RAW264.7 mouse macrophages. The results of quantitative PCR, ELISAs, and transcriptome analysis were combined to clarify the function of α7 nAChR regulation in the inflammatory response. Our findings indicate that the agonist PNU282987 significantly reduced the expression of the IL-6 gene and protein in inflammatory macrophages to attenuate the inflammatory response, but the antagonists MLA and α-conotoxin [A10L]PnIA had the opposite effects. Neither the agonist nor antagonists of α7 nAChR changed the expression level of the α7 nAChR subunit gene; they only regulated receptor function. This study provides a reference and scientific basis for the discovery of novel α7 nAChR agonists and their anti-inflammatory applications in the future.
Collapse
Affiliation(s)
- Yao Tan
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
| | - Zhaoli Chu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (Z.C.); (D.Z.)
| | - Hongyu Shan
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (Z.C.); (D.Z.)
| | - Xiaopeng Zhu
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
- Correspondence: (X.Z.); (S.L.)
| | - Sulan Luo
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (Z.C.); (D.Z.)
- Correspondence: (X.Z.); (S.L.)
| |
Collapse
|
27
|
Stress vulnerability shapes disruption of motor cortical neuroplasticity. Transl Psychiatry 2022; 12:91. [PMID: 35246507 PMCID: PMC8897461 DOI: 10.1038/s41398-022-01855-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic stress is a major cause of neuropsychiatric conditions such as depression. Stress vulnerability varies individually in mice and humans, measured by behavioral changes. In contrast to affective symptoms, motor retardation as a consequence of stress is not well understood. We repeatedly imaged dendritic spines of the motor cortex in Thy1-GFP M mice before and after chronic social defeat stress. Susceptible and resilient phenotypes were discriminated by symptom load and their motor learning abilities were assessed by a gross and fine motor task. Stress phenotypes presented individual short- and long-term changes in the hypothalamic-pituitary-adrenal axis as well as distinct patterns of altered motor learning. Importantly, stress was generally accompanied by a marked reduction of spine density in the motor cortex and spine dynamics depended on the stress phenotype. We found astrogliosis and altered microglia morphology along with increased microglia-neuron interaction in the motor cortex of susceptible mice. In cerebrospinal fluid, proteomic fingerprints link the behavioral changes and structural alterations in the brain to neurodegenerative disorders and dysregulated synaptic homeostasis. Our work emphasizes the importance of synaptic integrity and the risk of neurodegeneration within depression as a threat to brain health.
Collapse
|
28
|
Stress induced microglial activation contributes to depression. Pharmacol Res 2022; 179:106145. [DOI: 10.1016/j.phrs.2022.106145] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
|
29
|
Yin Y, Li H, Wang J, Kong Y, Chang J, Chu G. Implication of microglia in ketamine-induced long-term cognitive impairment in murine pups. Hum Exp Toxicol 2022; 41:9603271221128739. [PMID: 36172893 DOI: 10.1177/09603271221128739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Ketamine, a non-competitive N-methyl-D-aspartate receptor (NMDAR) antagonist, is widely applicable to anesthesia, analgesia, and sedation. However, the function and mechanisms of ketamine in the long-term learning and memory function of neonatal mice are unclear. OBJECTIVE The present study aims to investigate whether long-term learning and memory function will be affected by multiple ketamine exposures in the early development period. METHODS The mRNA and protein levels were measured by RT-qPCR and western blot, respectively. The Morris Water Maze test was performed to assess spatial learning and memory. RESULTS We identified that neonatal exposure to ketamine downsized the positive neurons for microtubule-associated protein doublecortin (DCX) and Ki67 in hippocampal dentate gyrus at the juvenile and late adolescence stages. Double-labeling tests demonstrated that the counts of Iba1+ cells and Ki67+ cells were pronouncedly diminished with exposure to ketamine. Further, qPCR assays to screen the key factors predisposing the populations and maturation of microglia exhibited remarkable decline of CX3CR1 mRNA levels in ketamine group versus the control group. The close relation of microglia to synaptic plasticity was depicted by the significantly downregulated synaptic plasticity-related proteins NR2B and PSD-95 subsequent to multiple exposures to ketamine. Finally, we found that both the protein and mRNA levels of BDNF were markedly decreased in ketamine group versus the control group. CONCLUSION We found that multiple exposures to ketamine in neonatal mice lead to spatial learning and memory dysfunction. The alterations of microglial development and function are the possible mechanisms of long-term learning and memory impairment.
Collapse
Affiliation(s)
- Y Yin
- Department of Anesthesiology, 117851Changzhou Maternity and Child Health Care Hospital, Changzhou, China
| | - H Li
- Department of Anesthesiology, 66322Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - J Wang
- Department of Anesthesiology, 56695Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Kong
- Department of Anesthesiology, 117851Changzhou Maternity and Child Health Care Hospital, Changzhou, China
| | - J Chang
- Department of Anesthesiology, 117851Changzhou Maternity and Child Health Care Hospital, Changzhou, China
| | - G Chu
- Department of Anesthesiology, 117851Changzhou Maternity and Child Health Care Hospital, Changzhou, China
| |
Collapse
|
30
|
Canedo T, Portugal CC, Socodato R, Almeida TO, Terceiro AF, Bravo J, Silva AI, Magalhães JD, Guerra-Gomes S, Oliveira JF, Sousa N, Magalhães A, Relvas JB, Summavielle T. Astrocyte-derived TNF and glutamate critically modulate microglia activation by methamphetamine. Neuropsychopharmacology 2021; 46:2358-2370. [PMID: 34400780 PMCID: PMC8581027 DOI: 10.1038/s41386-021-01139-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/12/2021] [Accepted: 07/24/2021] [Indexed: 02/06/2023]
Abstract
Methamphetamine (Meth) is a powerful illicit psychostimulant, widely used for recreational purposes. Besides disrupting the monoaminergic system and promoting oxidative brain damage, Meth also causes neuroinflammation, contributing to synaptic dysfunction and behavioral deficits. Aberrant activation of microglia, the largest myeloid cell population in the brain, is a common feature in neurological disorders triggered by neuroinflammation. In this study, we investigated the mechanisms underlying the aberrant activation of microglia elicited by Meth in the adult mouse brain. We found that binge Meth exposure caused microgliosis and disrupted risk assessment behavior (a feature that usually occurs in individuals who abuse Meth), both of which required astrocyte-to-microglia crosstalk. Mechanistically, Meth triggered a detrimental increase of glutamate exocytosis from astrocytes (in a process dependent on TNF production and calcium mobilization), promoting microglial expansion and reactivity. Ablating TNF production, or suppressing astrocytic calcium mobilization, prevented Meth-elicited microglia reactivity and re-established risk assessment behavior as tested by elevated plus maze (EPM). Overall, our data indicate that glial crosstalk is critical to relay alterations caused by acute Meth exposure.
Collapse
Affiliation(s)
- Teresa Canedo
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Camila Cabral Portugal
- Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| | - Renato Socodato
- grid.5808.50000 0001 1503 7226Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Tiago Oliveira Almeida
- grid.5808.50000 0001 1503 7226Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Filipa Terceiro
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Joana Bravo
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Isabel Silva
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - João Duarte Magalhães
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sónia Guerra-Gomes
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - João Filipe Oliveira
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal ,grid.410922.c0000 0001 0180 6901IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence, Barcelos, Portugal
| | - Nuno Sousa
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Magalhães
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - João Bettencourt Relvas
- grid.5808.50000 0001 1503 7226Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal ,grid.5808.50000 0001 1503 7226Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Teresa Summavielle
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal. .,ESS.PP, Escola Superior de Saúde do Politécnico do Porto, Porto, Portugal.
| |
Collapse
|
31
|
Mao Y, Evans EE, Mishra V, Balch L, Eberhardt A, Zauderer M, Gold WA. Anti-Semaphorin 4D Rescues Motor, Cognitive, and Respiratory Phenotypes in a Rett Syndrome Mouse Model. Int J Mol Sci 2021; 22:ijms22179465. [PMID: 34502373 PMCID: PMC8431088 DOI: 10.3390/ijms22179465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 01/09/2023] Open
Abstract
Rett syndrome is a neurodevelopmental disorder caused by mutations of the methyl-CpG binding protein 2 gene. Abnormal physiological functions of glial cells contribute to pathogenesis of Rett syndrome. Semaphorin 4D (SEMA4D) regulates processes central to neuroinflammation and neurodegeneration including cytoskeletal structures required for process extension, communication, and migration of glial cells. Blocking SEMA4D-induced gliosis may preserve normal glial and neuronal function and rescue neurological dysfunction in Rett syndrome. We evaluated the pre-clinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody in the Rett syndrome Mecp2T158A transgenic mouse model and investigated the contribution of glial cells as a proposed mechanism of action in treated mice and in primary glial cultures isolated from Mecp2T158A/y mutant mice. SEMA4D is upregulated in neurons while glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1-positive cells are upregulated in Mecp2T158A/y mice. Anti-SEMA4D treatment ameliorates Rett syndrome-specific symptoms and improves behavioural functions in both pre-symptomatic and symptomatic cohorts of hemizygous Mecp2T158A/y male mice. Anti-SEMA4D also reduces astrocyte and microglia activation in vivo. In vitro experiments demonstrate an abnormal cytoskeletal structure in mutant astrocytes in the presence of SEMA4D, while anti-SEMA4D antibody treatment blocks SEMA4D–Plexin B1 signaling and mitigates these abnormalities. These results suggest that anti-SEMA4D immunotherapy may be an effective treatment option to alleviate symptoms and improve cognitive and motor function in Rett syndrome.
Collapse
Affiliation(s)
- Yilin Mao
- Molecular Neurobiology Research Laboratory, Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia;
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Elizabeth E. Evans
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Vikas Mishra
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Leslie Balch
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Allison Eberhardt
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Maurice Zauderer
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Wendy A. Gold
- Molecular Neurobiology Research Laboratory, Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia;
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Molecular Neurobiology Research Laboratory, The Children’s Medical Research Institute, Westmead, NSW 2145, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
32
|
Siemsen BM, Landin JD, McFaddin JA, Hooker KN, Chandler LJ, Scofield MD. Chronic intermittent ethanol and lipopolysaccharide exposure differentially alter Iba1-derived microglia morphology in the prelimbic cortex and nucleus accumbens core of male Long-Evans rats. J Neurosci Res 2021; 99:1922-1939. [PMID: 32621337 PMCID: PMC7779701 DOI: 10.1002/jnr.24683] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/22/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022]
Abstract
Accumulating evidence has linked pathological changes associated with chronic alcohol exposure to neuroimmune signaling mediated by microglia. Prior characterization of the microglial structure-function relationship demonstrates that alterations in activity states occur concomitantly with reorganization of cellular architecture. Accordingly, gaining a better understanding of microglial morphological changes associated with ethanol exposure will provide valuable insight into how neuroimmune signaling may contribute to ethanol-induced reshaping of neuronal function. Here we have used Iba1-staining combined with high-resolution confocal imaging and 3D reconstruction to examine microglial structure in the prelimbic (PL) cortex and nucleus accumbens (NAc) in male Long-Evans rats. Rats were either sacrificed at peak withdrawal following 15 days of exposure to chronic intermittent ethanol (CIE) or 24 hr after two consecutive injections of the immune activator lipopolysaccharide (LPS), each separated by 24 hr. LPS exposure resulted in dramatic structural reorganization of microglia in the PL cortex, including increased soma volume, overall cellular volume, and branching complexity. In comparison, CIE exposure was associated with a subtle increase in somatic volume and differential effects on microglia processes, which were largely absent in the NAc. These data reveal that microglial activation following a neuroimmune challenge with LPS or exposure to chronic alcohol exhibits distinct morphometric profiles and brain region-dependent specificity.
Collapse
Affiliation(s)
- Benjamin M. Siemsen
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Justine D. Landin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jon A. McFaddin
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Kaylee N. Hooker
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Lawrence J. Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Michael D. Scofield
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
33
|
Huang Y, Sun X, Juan Z, Zhang R, Wang R, Meng S, Zhou J, Li Y, Xu K, Xie K. Dexmedetomidine attenuates myocardial ischemia-reperfusion injury in vitro by inhibiting NLRP3 Inflammasome activation. BMC Anesthesiol 2021; 21:104. [PMID: 33823789 PMCID: PMC8022424 DOI: 10.1186/s12871-021-01334-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
Background Myocardial ischemia-reperfusion injury (MIRI) is the most common cause of death worldwide. The NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome plays an important role in the inflammatory response to MIRI. Dexmedetomidine (DEX), a specific agonist of α2-adrenergic receptor, is commonly used for sedation and analgesia in anesthesia and critically ill patients. Several studies have shown that dexmedetomidine has a strong anti-inflammatory effect in many diseases. Here, we investigated whether dexmedetomidine protects against MIRI by inhibiting the activation of the NLRP3 inflammasome in vitro. Methods We established an MIRI model in cardiomyocytes (CMs) alone and in coculture with cardiac fibroblasts (CFs) by hypoxia/reoxygenation (H/R) in vitro. The cells were treated with dexmedetomidine with or without MCC950 (a potent selective NLRP3 inhibitor). The beating rate and cell viability of cardiomyocytes, NLRP3 localization, the expression of inflammatory cytokines and NLRP3 inflammasome-related proteins, and the expression of apoptosis-related proteins, including Bcl2 and BAX, were determined. Results Dexmedetomidine treatment increased the beating rates and viability of cardiomyocytes cocultured with cardiac fibroblasts. The expression of the NLRP3 protein was significantly upregulated in cardiac fibroblasts but not in cardiomyocytes after H/R and was significantly attenuated by dexmedetomidine treatment. Expression of the inflammatory cytokines IL-1β, IL-18 and TNF-α was significantly increased in cardiac fibroblasts after H/R and was attenuated by dexmedetomidine treatment. NLRP3 inflammasome activation induced the increased expression of cleaved caspase1, mature IL-1β and IL-18, while dexmedetomidine suppressed H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts. In addition, dexmedetomidine reduced the expression of Bcl2 and BAX in cocultured cardiomyocytes by suppressing H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts. Conclusion Dexmedetomidine treatment can suppress H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts, thereby alleviating MIRI by inhibiting the inflammatory response. Supplementary Information The online version contains supplementary material available at 10.1186/s12871-021-01334-5.
Collapse
Affiliation(s)
- Yaru Huang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Xiaotong Sun
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Zhaodong Juan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China.
| | - Rui Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Ruoguo Wang
- Department of Pain, Affiliated Hospital of Weifang Medical University, Weifang, 261000, China
| | - Shuqi Meng
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Jiajia Zhou
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Yan Li
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Keyou Xu
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China
| | - Keliang Xie
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, 261021, China.
| |
Collapse
|
34
|
DA SILVA DG, de CARVALHO ILQ, TOSCANO ECDB, SANTOS BÁDSS, OLIVEIRA BDS, CAMPOS MA, da FONSECA FG, CAMARGOS QM, de SOUSA GF, CALIARI MV, TEIXEIRA AL, de MIRANDA AS, RACHID MA. Brain-derived neurotrophic factor is down regulated after bovine alpha-herpesvirus 5 infection in both wild-type and TLR3/7/9 deficient mice. J Vet Med Sci 2021; 83:180-186. [PMID: 33281142 PMCID: PMC7972877 DOI: 10.1292/jvms.20-0204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 11/23/2020] [Indexed: 11/25/2022] Open
Abstract
Neurotrophic factors have been implicated in the control of neuronal survival and plasticity in different brain diseases. Meningoencephalitis caused by bovine alpha-herpesvirus 5 (BoHV-5) infection is a frequent neurological disease of young cattle, being the involvement of apoptosis in the development of neuropathological changes frequently discussed in the literature. It's well known that Toll-like receptors (TLRs) can activate neuroinflammatory response and consequently lead to neuronal loss. However, there are no studies evaluating the expression of neurotrophic factors and their association with brain pathology and TLRs during the infection by BoHV-5. The current study aimed to analyze brain levels of neurotrophic factors along with neuropathological changes during acute infection by BoHV-5 in wild-type (WT) and TLR3/7/9 (TLR3/7/9-/-) deficiency mice. The infection was induced by intracranial inoculation of 1 × 104 TCID50 of BoHV-5. Infected animals presented similar degrees of clinical signs and neuropathological changes. Both infected groups had meningoencephalitis and neuronal damage in CA regions from hippocampus. BoHV-5 infection promoted the proliferation of Iba-1 positive cells throughout the neuropil, mainly located in the frontal cortex. Moreover, significant lower levels of brain-derived neurotrophic factor (BDNF) were detected in both BoHV-5 infected WT and TLR3/7/9 deficient mice, compared with non-infected animals. Our study showed that BDNF down regulation was associated with brain inflammation, reactive microgliosis and neuronal loss after bovine alpha-herpesvirus 5 infection in mice. Moreover, we demonstrated that combined TLR3/7/9 deficiency does not alter those parameters.
Collapse
Affiliation(s)
- Daniele Gonçalves DA SILVA
- Laboratory of Cellular and Molecular Pathology, Department
of General Pathology, Biological Science Institute, Federal University of Minas Gerais,
Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Iracema Luisa Quintino de CARVALHO
- Department of Microbiology, Biological Science Institute,
Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Eliana Cristina de Brito TOSCANO
- Laboratory of Cellular and Molecular Pathology, Department
of General Pathology, Biological Science Institute, Federal University of Minas Gerais,
Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Beatriz Álvares da Silva Senra SANTOS
- Laboratory of Animal Virology, Department of Preventive
Veterinary Medicine, Veterinary School, Federal University of Minas Gerais, Belo
Horizonte, Minas Gerais, 31270-901, Brazil
| | - Bruna da Silva OLIVEIRA
- Department of Morphology, Biological Science Institute,
Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Marco Antônio CAMPOS
- René Rachou Institute, Fiocruz Minas, Belo Horizonte, Minas
Gerais, 30190-002, Brazil
| | - Flávio Guimarães da FONSECA
- Department of Microbiology, Biological Science Institute,
Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Quezya Mendes CAMARGOS
- Laboratory of Cellular and Molecular Pathology, Department
of General Pathology, Biological Science Institute, Federal University of Minas Gerais,
Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Gabriela Ferreira de SOUSA
- Laboratory of Cellular and Molecular Pathology, Department
of General Pathology, Biological Science Institute, Federal University of Minas Gerais,
Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Marcelo Vidigal CALIARI
- Laboratory of Cellular and Molecular Pathology, Department
of General Pathology, Biological Science Institute, Federal University of Minas Gerais,
Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Antônio Lúcio TEIXEIRA
- Neuropsychiatry Program, Department of Psychiatry and
Behavioral Sciences, School of Medicine, University of Texas Health Science Center at
Houston, TX, 77054, USA
| | - Aline Silva de MIRANDA
- Department of Morphology, Biological Science Institute,
Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Milene Alvarenga RACHID
- Laboratory of Cellular and Molecular Pathology, Department
of General Pathology, Biological Science Institute, Federal University of Minas Gerais,
Belo Horizonte, Minas Gerais, 31270-901, Brazil
| |
Collapse
|
35
|
La Vitola P, Balducci C, Baroni M, Artioli L, Santamaria G, Castiglioni M, Cerovic M, Colombo L, Caldinelli L, Pollegioni L, Forloni G. Peripheral inflammation exacerbates α-synuclein toxicity and neuropathology in Parkinson's models. Neuropathol Appl Neurobiol 2021; 47:43-60. [PMID: 32696999 DOI: 10.1111/nan.12644] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022]
Abstract
AIMS Parkinson's disease and related disorders are devastating neurodegenerative pathologies. Since α-synuclein was identified as a main component of Lewy bodies and neurites, efforts have been made to clarify the pathogenic mechanisms of α-synuclein's detrimental effects. α-synuclein oligomers are the most harmful species and may recruit and activate glial cells. Inflammation is emerging as a bridge between genetic susceptibility and environmental factors co-fostering Parkinson's disease. However, direct evidence linking inflammation to the harmful activities of α-synuclein oligomers or to the Parkinson's disease behavioural phenotype is lacking. METHODS To clarify whether neuroinflammation influences Parkinson's disease pathogenesis, we developed: (i) a 'double-hit' approach in C57BL/6 naive mice where peripherally administered lipopolysaccharides were followed by intracerebroventricular injection of an inactive oligomer dose; (ii) a transgenic 'double-hit' model where lipopolysaccharides were given to A53T α-synuclein transgenic Parkinson's disease mice. RESULTS Lipopolysaccharides induced a long-lasting neuroinflammatory response which facilitated the detrimental cognitive activities of oligomers. LPS-activated microglia and astrocytes responded differently to the oligomers with microglia activating further and acquiring a pro-inflammatory M1 phenotype, while astrocytes atrophied. In the transgenic 'double-hit' A53T mouse model, lipopolysaccharides aggravated cognitive deficits and increased microgliosis. Again, astrocytes responded differently to the double challenge. These findings indicate that peripherally induced neuroinflammation potentiates the α-synuclein oligomer's actions and aggravates cognitive deficits in A53T mice. CONCLUSIONS The fine management of both peripheral and central inflammation may offer a promising therapeutic approach to prevent or slow down some behavioural aspects in α-synucleinopathies.
Collapse
Affiliation(s)
- P La Vitola
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - C Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Baroni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Artioli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - G Santamaria
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Castiglioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Cerovic
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Caldinelli
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - L Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - G Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
36
|
Neuroprotective function of microglia in the developing brain. Neuronal Signal 2021; 5:NS20200024. [PMID: 33532089 PMCID: PMC7823182 DOI: 10.1042/ns20200024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 12/16/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system and are important for immune processes. Besides their classical roles in pathological conditions, these cells also dynamically interact with neurons and influence their structure and function in physiological conditions. Recent evidence revealed their role in healthy brain homeostasis, including the regulation of neurogenesis, cell survival, and synapse maturation and elimination, especially in the developing brain. In this review, we summarize the current state of knowledge on microglia in brain development, with a focus on their neuroprotective function. We will also discuss how microglial dysfunction may lead to the impairment of brain function, thereby contributing to disease development.
Collapse
|
37
|
Li S, Liao Y, Dong Y, Li X, Li J, Cheng Y, Cheng J, Yuan Z. Microglial deletion and inhibition alleviate behavior of post-traumatic stress disorder in mice. J Neuroinflammation 2021; 18:7. [PMID: 33402212 PMCID: PMC7786489 DOI: 10.1186/s12974-020-02069-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Alteration of immune status in the central nervous system (CNS) has been implicated in the development of post-traumatic stress disorder (PTSD). However, the nature of overall changes in brain immunocyte landscape in PTSD condition remains unclear. Methods We constructed a mouse PTSD model by electric foot-shocks followed by contextual reminders and verified the PTSD-related symptoms by behavior test (including contextual freezing test, open-field test, and elevated plus maze test). We examined the immunocyte panorama in the brains of the naïve or PTSD mice by using single-cell mass cytometry. Microglia number and morphological changes in the hippocampus, prefrontal cortex, and amygdala were analyzed by histopathological methods. The gene expression changes of those microglia were detected by quantitative real-time PCR. Genetic/pharmacological depletion of microglia or minocycline treatment before foot-shocks exposure was performed to study the role of microglia in PTSD development and progress. Results We found microglia are the major brain immune cells that respond to PTSD. The number of microglia and ratio of microglia to immunocytes was significantly increased on the fifth day of foot-shock exposure. Furthermore, morphological analysis and gene expression profiling revealed temporal patterns of microglial activation in the hippocampus of the PTSD brains. Importantly, we found that genetic/pharmacological depletion of microglia or minocycline treatment before foot-shock exposure alleviated PTSD-associated anxiety and contextual fear. Conclusion Our results demonstrated a critical role for microglial activation in PTSD development and a potential therapeutic strategy for the clinical treatment of PTSD in the form of microglial inhibition. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02069-9.
Collapse
Affiliation(s)
- Shuoshuo Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Yajin Liao
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
| | - Yuan Dong
- Department of Biochemistry, Medical College, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xiaoheng Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Jun Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
| | - Jinbo Cheng
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China.
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, 100850, China. .,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
38
|
Bassett B, Subramaniyam S, Fan Y, Varney S, Pan H, Carneiro AMD, Chung CY. Minocycline alleviates depression-like symptoms by rescuing decrease in neurogenesis in dorsal hippocampus via blocking microglia activation/phagocytosis. Brain Behav Immun 2021; 91:519-530. [PMID: 33176182 DOI: 10.1016/j.bbi.2020.11.009] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/12/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Clinical studies examining the potential of anti-inflammatory agents, specifically of minocycline, as a treatment for depression has shown promising results. However, mechanistic insights into the neuroprotective and anti-inflammatory actions of minocycline need to be provided. We evaluated the effect of minocycline on chronic mild stress (CMS) induced depressive-like behavior, and behavioral assays revealed minocycline ameliorate depressive behaviors. Multiple studies suggest a role of microglia in depression, revealing that microglia activation correlates with a decrease in neurogenesis and increased depressive-like behavior. The effect of minocycline on microglia activation in different areas of the dorsal or ventral hippocampus in stressed mice was examined by immunohistochemistry. We observed the increase in the number of activated microglia expressing CD68 after exposure to three weeks of chronic stress, whereas no changes in total microglia number were observed. These changes were observed throughout the DG, CA1 and CA2 regions in dorsal hippocampus but restricted to the DG of the ventral hippocampus. In vitro experiments including western blotting and phagocytosis assay were used to investigate the effect of minocycline on microglia activation. Activation of primary microglia by LPS in vitro causes and ERK1/2 activation, enhancement of iNOS expression and phagocytic activity, and alterations in cellular morphology that are reversed by minocycline exposure, suggesting that minocycline directly acts on microglia to reduce phagocytic potential. Our results suggest the most probable mechanism by which minocycline reverses the pathogenic phagocytic potential of neurotoxic M1 microglia, and reduces the negative phenotypes associated with reduced neurogenesis caused by exposure to chronic stress.
Collapse
Affiliation(s)
- Ben Bassett
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Selvaraj Subramaniyam
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yang Fan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Seth Varney
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Hope Pan
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ana M D Carneiro
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Chang Y Chung
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; Division of Natural Science, Duke Kunshan University, Kunshan 215316, China.
| |
Collapse
|
39
|
Forloni G, La Vitola P, Cerovic M, Balducci C. Inflammation and Parkinson's disease pathogenesis: Mechanisms and therapeutic insight. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:175-202. [PMID: 33453941 DOI: 10.1016/bs.pmbts.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
After Alzheimer's disease, Parkinson's disease is the most frequent neurodegenerative disorder. Although numerous treatments have been developed to control the disease symptomatology, with some successes, an efficacious therapy affecting the causes of PD is still a goal to pursue. The genetic evidence and the identification of α-synuclein as the main component of intracellular Lewy bodies, the neuropathological hallmark of PD and related disorders, have changed the approach to these disorders. More recently, the detrimental role of α-synuclein has been further extended to explain the wide spread of cerebral pathology through its oligomers. To emphasize the central pathogenic role of these soluble aggregates, we have defined synucleinopathies and other neurodegenerative disorders associated with protein misfolding as oligomeropathies. Another common element in the pathogenesis of oligomeropathies is the role played by inflammation, both at the peripheral and cerebral levels. In the brain parenchyma, inflammatory reaction has been considered an obvious consequence of neuronal degeneration, but recent observations indicate a direct contribution of glial alteration in the early phase of the disease. Furthermore, systemic inflammation also influences the development of neuronal dysfunction caused by specific elements, β amyloid, α-synuclein, tau or prion. However, each disorder has its own specific pathological process and within the same pathological condition, it is possible to find inter-individual differences. This heterogeneity might explain the difficulties developing efficacious therapeutic approaches, even though the possibility of intervention is supported by robust biological evidence. We have recently demonstrated that peripheral inflammation can amplify the neuronal dysfunction induced by α-synuclein oligomers and the neuropathological consequences observed in a Parkinson's disease model. In both cases, activation of microglia was incremented by the "double hit" process, compared to the single treatment. In contrast, astrocyte activation was attenuated and these cells appeared damaged when chronic inflammation was combined with α-synuclein exposure. This evidence might indicate a more specific anti-inflammatory strategy rather than the generic anti-inflammatory treatment.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Pietro La Vitola
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Milica Cerovic
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Claudia Balducci
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
40
|
Post-inflammatory behavioural despair in male mice is associated with reduced cortical glutamate-glutamine ratios, and circulating lipid and energy metabolites. Sci Rep 2020; 10:16857. [PMID: 33033375 PMCID: PMC7545201 DOI: 10.1038/s41598-020-74008-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
Post-inflammatory behaviours in rodents are widely used to model human depression and to test the efficacy of novel anti-depressants. Mice injected with lipopolysaccharide (LPS) display a depressive-like phenotype twenty-four hours after endotoxin administration. Despite the widespread use of this model, the mechanisms that underlie the persistent behavioural changes after the transient peripheral inflammatory response remain elusive. The study of the metabolome, the collection of all the small molecule metabolites in a sample, combined with multivariate statistical techniques provides a way of studying biochemical pathways influenced by an LPS challenge. Adult male CD-1 mice received an intraperitoneal injection of either LPS (0.83 mg/kg) or saline, and were assessed for depressive-like behaviour 24 h later. In a separate mouse cohort, pro-inflammatory cytokine gene expression and 1H nuclear magnetic resonance (NMR) metabolomics measurements were made in brain tissue and blood. Statistical analyses included Independent Sample t-tests for gene expression data, and supervised multi-variate analysis using orthogonal partial least squares discriminant analysis for metabolomics. Both plasma and brain metabolites in male mice were altered following a single peripheral LPS challenge that led to depressive-like behaviour in the forced swim test. The plasma metabolites altered by LPS are involved in energy metabolism, including lipoproteins, glucose, creatine, and isoleucine. In the brain, glutamate, serine, and N-acetylaspartate (NAA) were reduced after LPS, whereas glutamine was increased. Serine-modulated glutamatergic signalling and changes in bioenergetics may mediate the behavioural phenotype induced by LPS. In light of other data supporting a central imbalance of glutamate-glutamine cycling in depression, our results suggest that aberrant central glutaminergic signalling may underpin the depressive-like behaviours that result from both inflammation and non-immune pathophysiology. Normalising glutaminergic signalling, rather than seeking to increase serotonergic signalling, might prove to be a more coherent approach to the development of new treatments for mood disorder.
Collapse
|
41
|
Pinto B, Morelli G, Rastogi M, Savardi A, Fumagalli A, Petretto A, Bartolucci M, Varea E, Catelani T, Contestabile A, Perlini LE, Cancedda L. Rescuing Over-activated Microglia Restores Cognitive Performance in Juvenile Animals of the Dp(16) Mouse Model of Down Syndrome. Neuron 2020; 108:887-904.e12. [PMID: 33027640 PMCID: PMC7736620 DOI: 10.1016/j.neuron.2020.09.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 01/01/2023]
Abstract
Microglia are brain-resident immune cells and regulate mechanisms essential for cognitive functions. Down syndrome (DS), the most frequent cause of genetic intellectual disability, is caused by a supernumerary chromosome 21, containing also genes related to the immune system. In the hippocampus of the Dp(16) mouse model of DS and DS individuals, we found activated microglia, as assessed by their morphology; activation markers; and, for DS mice, electrophysiological profile. Accordingly, we found increased pro-inflammatory cytokine levels and altered interferon signaling in Dp(16) hippocampi. DS mice also showed decreased spine density and activity of hippocampal neurons and hippocampus-dependent cognitive behavioral deficits. Depletion of defective microglia or treatment with a commonly used anti-inflammatory drug rescued the neuronal spine and activity impairments and cognitive deficits in juvenile Dp(16) mice. Our results suggest an involvement of microglia in Dp(16)-mouse cognitive deficits and identify a new potential therapeutic approach for cognitive disabilities in DS individuals. DS mice display microglia alterations and cognitive impairment Depletion of microglia rescues cognitive impairment in DS mice Acetaminophen treatment rescues microglia and cognitive impairments in DS mice Brain samples of DS people recapitulate microglia alterations observed in DS mice
Collapse
Affiliation(s)
- Bruno Pinto
- BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Giovanni Morelli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Mohit Rastogi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Amos Fumagalli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Emilio Varea
- Cellular Biology Department, University of Valencia, Valencia, Spain
| | - Tiziano Catelani
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Laura E Perlini
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy; Dulbecco Telethon Institute, Rome, Italy.
| |
Collapse
|
42
|
The Association of Saliva Cytokines and Pediatric Sports-Related Concussion Outcomes. J Head Trauma Rehabil 2020; 35:354-362. [PMID: 32881769 DOI: 10.1097/htr.0000000000000605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES This study aimed to explore cytokine alterations following pediatric sports-related concussion (SRC) and whether a specific cytokine profile could predict symptom burden and time to return to sports (RTS). SETTING Sports Medicine Clinic. PARTICIPANTS Youth ice hockey participants (aged 12-17 years) were recruited prior to the 2013-2016 hockey season. DESIGN Prospective exploratory cohort study. MAIN MEASURE Following SRC, saliva samples were collected and a Sport Concussion Assessment Tool version 3 (SCAT3) was administered within 72 hours of injury and analyzed for cytokines. Additive regression of decision stumps was used to model symptom burden and length to RTS based on cytokine and clinical features. RRelieFF feature selection was used to determine the predictive value of each cytokine and clinical feature, as well as to identify the optimal cytokine profile for the symptom burden and RTS. RESULTS Thirty-six participants provided samples post-SRC (81% male; age 14.4 ± 1.3 years). Of these, 10 features, sex, number of previous concussions, and 8 cytokines, were identified to lead to the best prediction of symptom severity (r = 0.505, P = .002), while 12 cytokines, age, and history of previous concussions predicted the number of symptoms best (r = 0.637, P < .001). The prediction of RTS led to the worst results, requiring 21 cytokines, age, sex, and number of previous concussions as features (r = -0.320, P = .076). CONCLUSIONS In pediatric ice hockey participants following SRC, there is evidence of saliva cytokine profiles that are associated with increased symptom burden. However, further studies are needed.
Collapse
|
43
|
Nakamura Y, Kimura S, Takada N, Takemura M, Iwamoto M, Hisaoka-Nakashima K, Nakata Y, Morioka N. Stimulation of toll-like receptor 4 downregulates the expression of α7 nicotinic acetylcholine receptors via histone deacetylase in rodent microglia. Neurochem Int 2020; 138:104751. [DOI: 10.1016/j.neuint.2020.104751] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/23/2020] [Accepted: 05/03/2020] [Indexed: 12/17/2022]
|
44
|
Chang X, Li J, Niu S, Xue Y, Tang M. Neurotoxicity of metal‐containing nanoparticles and implications in glial cells. J Appl Toxicol 2020; 41:65-81. [DOI: 10.1002/jat.4037] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Xiaoru Chang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Jiangyan Li
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| |
Collapse
|
45
|
Kokkosis AG, Tsirka SE. Neuroimmune Mechanisms and Sex/Gender-Dependent Effects in the Pathophysiology of Mental Disorders. J Pharmacol Exp Ther 2020; 375:175-192. [PMID: 32661057 DOI: 10.1124/jpet.120.266163] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022] Open
Abstract
Innate and adaptive immune mechanisms have emerged as critical regulators of CNS homeostasis and mental health. A plethora of immunologic factors have been reported to interact with emotion- and behavior-related neuronal circuits, modulating susceptibility and resilience to mental disorders. However, it remains unclear whether immune dysregulation is a cardinal causal factor or an outcome of the pathologies associated with mental disorders. Emerging variations in immune regulatory pathways based on sex differences provide an additional framework for discussion in these psychiatric disorders. In this review, we present the current literature pertaining to the effects that disrupted immune pathways have in mental disorder pathophysiology, including immune dysregulation in CNS and periphery, microglial activation, and disturbances of the blood-brain barrier. In addition, we present the suggested origins of such immune dysregulation and discuss the gender and sex influence of the neuroimmune substrates that contribute to mental disorders. The findings challenge the conventional view of these disorders and open the window to a diverse spectrum of innovative therapeutic targets that focus on the immune-specific pathophenotypes in neuronal circuits and behavior. SIGNIFICANCE STATEMENT: The involvement of gender-dependent inflammatory mechanisms on the development of mental pathologies is gaining momentum. This review addresses these novel factors and presents the accumulating evidence introducing microglia and proinflammatory elements as critical components and potential targets for the treatment of mental disorders.
Collapse
Affiliation(s)
- Alexandros G Kokkosis
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Stella E Tsirka
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| |
Collapse
|
46
|
Siebold L, Krueger AC, Abdala JA, Figueroa JD, Bartnik-Olson B, Holshouser B, Wilson CG, Ashwal S. Cosyntropin Attenuates Neuroinflammation in a Mouse Model of Traumatic Brain Injury. Front Mol Neurosci 2020; 13:109. [PMID: 32670020 PMCID: PMC7332854 DOI: 10.3389/fnmol.2020.00109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/22/2020] [Indexed: 12/21/2022] Open
Abstract
Aim: Traumatic brain injury (TBI) is a leading cause of mortality/morbidity and is associated with chronic neuroinflammation. Melanocortin receptor agonists including adrenocorticotropic hormone (ACTH) ameliorate inflammation and provide a novel therapeutic approach. We examined the effect of long-acting cosyntropin (CoSyn), a synthetic ACTH analog, on the early inflammatory response and functional outcome following experimental TBI. Methods: The controlled cortical impact model was used to induce TBI in mice. Mice were assigned to injury and treatment protocols resulting in four experimental groups including sham + saline, sham + CoSyn, TBI + saline, and TBI + CoSyn. Treatment was administered subcutaneously 3 h post-injury and daily injections were given for up to 7 days post-injury. The early inflammatory response was evaluated at 3 days post-injury through the evaluation of cytokine expression (IL1β and TNFα) and immune cell response. Quantification of immune cell response included cell counts of microglia/macrophages (Iba1+ cells) and neutrophils (MPO+ cells) in the cortex and hippocampus. Behavioral testing (n = 10–14 animals/group) included open field (OF) and novel object recognition (NOR) during the first week following injury and Morris water maze (MWM) at 10–15 days post-injury. Results: Immune cell quantification showed decreased accumulation of Iba1+ cells in the perilesional cortex and CA1 region of the hippocampus for CoSyn-treated TBI animals compared to saline-treated. Reduced numbers of MPO+ cells were also found in the perilesional cortex and hippocampus in CoSyn treated TBI mice compared to their saline-treated counterparts. Furthermore, CoSyn treatment reduced IL1β expression in the cortex of TBI mice. Behavioral testing showed a treatment effect of CoSyn for NOR with CoSyn increasing the discrimination ratio in both TBI and Sham groups, indicating increased memory performance. CoSyn also decreased latency to find platform during the early training period of the MWM when comparing CoSyn to saline-treated TBI mice suggesting moderate improvements in spatial memory following CoSyn treatment. Conclusion: Reduced microglia/macrophage accumulation and neutrophil infiltration in conjunction with moderate improvements in spatial learning in our CoSyn treated TBI mice suggests a beneficial anti-inflammatory effect of CoSyn following TBI.
Collapse
Affiliation(s)
- Lorraine Siebold
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Amy C Krueger
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jonathan A Abdala
- The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Johnny D Figueroa
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Center for Health Disparities and Molecular Medicine, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Brenda Bartnik-Olson
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Barbara Holshouser
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Christopher G Wilson
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,The Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States.,Department of Pediatrics, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Stephen Ashwal
- Department of Pediatrics, Loma Linda University Medical Center, Loma Linda, CA, United States
| |
Collapse
|
47
|
Kuter KZ, Cenci MA, Carta AR. The role of glia in Parkinson's disease: Emerging concepts and therapeutic applications. PROGRESS IN BRAIN RESEARCH 2020; 252:131-168. [PMID: 32247363 DOI: 10.1016/bs.pbr.2020.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Originally believed to primarily affect neurons, Parkinson's disease (PD) has recently been recognized to also affect the functions and integrity of microglia and astroglia, two cell categories of fundamental importance to brain tissue homeostasis, defense, and repair. Both a loss of glial supportive-defensive functions and a toxic gain of glial functions are implicated in the neurodegenerative process. Moreover, the chronic treatment with L-DOPA may cause maladaptive glial plasticity favoring a development of therapy complications. This chapter focuses on the pathophysiology of PD from a glial point of view, presenting this rapidly growing field from the first discoveries made to the most recent developments. We report and compare histopathological and molecular findings from experimental models of PD and human studies. We moreover discuss the important role played by astrocytes in compensatory adaptations taking place during presymptomatic disease stages. We finally describe examples of potential therapeutic applications stemming from an increased understanding of the important roles of glia in PD.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy.
| |
Collapse
|
48
|
Zhang L, Zheng H, Wu R, Kosten TR, Zhang XY, Zhao J. The effect of minocycline on amelioration of cognitive deficits and pro-inflammatory cytokines levels in patients with schizophrenia. Schizophr Res 2019; 212:92-98. [PMID: 31416745 DOI: 10.1016/j.schres.2019.08.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cognitive deficits of schizophrenia are predictors of poor function, but antipsychotic medication has limited efficacy for cognitive deficits. These deficits in learning and memory may result from activity of pro-inflammatory cytokines, which microglia produce. The microglia inhibitor minocycline might arrest this cytokine damage to the hippocampus and reverse the cognitive deficits of schizophrenia. METHODS A double-blind, placebo-controlled study involved 75 patients with schizophrenia who randomly received low dose (100 mg/day) or high dose minocycline (200 mg/day) or placebo added to risperidone. MATRICS Consensus Cognitive Battery (MCCB) was used to assess the cognitive functioning, and serum levels of Interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) were assessed. RESULTS Minocyclinehigh dose group was significantly superior to minocyclinelow dose or placebo group not only for the improvements in cognitive tests' scores as well (P < 0.05), but for IL-1β and IL-6 serum levels reduction (P < 0.01). The amelioration of cognitive deficits with minocycline correlated not only with the remission of negative symptoms, but also with the reduction in serum levels of IL-1β and IL-6. CONCLUSIONS Minocycline adjunctive treatment was effective in improving cognitive deficits of patients with schizophrenia. The beneficial effect of minocycline may be related to reducing pro-inflammatory cytokines through microglia inhibition.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, Central South University; Chinese National Clinical Research Center on Mental Disorders, Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China; Department of Psychiatry, Guangzhou First People's Hospital, the Second Affiliated Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Hongbo Zheng
- GuangzhouBaiyun Psychiatric Hospital, Guangzhou, Guangdong, China
| | - Rengrong Wu
- Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, Central South University; Chinese National Clinical Research Center on Mental Disorders, Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China
| | - Thomas R Kosten
- Department of Psychiatry, Baylor College of Medicine, Houston, TX, USA
| | - Xiang-Yang Zhang
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, TX, USA
| | - Jingping Zhao
- Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, Central South University; Chinese National Clinical Research Center on Mental Disorders, Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China; The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, Guangdong, China.
| |
Collapse
|
49
|
Morris G, Puri BK, Walker AJ, Maes M, Carvalho AF, Bortolasci CC, Walder K, Berk M. Shared pathways for neuroprogression and somatoprogression in neuropsychiatric disorders. Neurosci Biobehav Rev 2019; 107:862-882. [PMID: 31545987 DOI: 10.1016/j.neubiorev.2019.09.025] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/13/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
Activated immune-inflammatory, oxidative and nitrosative stress (IO&NS) pathways and consequent mitochondrial aberrations are involved in the pathophysiology of psychiatric disorders including major depression, bipolar disorder and schizophrenia. They offer independent and shared contributions to pathways underpinning medical comorbidities including insulin resistance, metabolic syndrome, obesity and cardiovascular disease - herein conceptualized as somatoprogression. This narrative review of human studies aims to summarize relationships between IO&NS pathways, neuroprogression and somatoprogression. Activated IO&NS pathways, implicated in the neuroprogression of psychiatric disorders, affect the pathogenesis of comorbidities including insulin resistance, dyslipidaemia, obesity and hypertension, and by inference, metabolic syndrome. These conditions activate IO&NS pathways, exacerbating neuroprogression in psychiatric disorders. The processes whereby proinflammatory cytokines, nitrosative and endoplasmic reticulum stress, NADPH oxidase isoforms, PPARγ inactivation, SIRT1 deficiency and intracellular signalling pathways impact lipid metabolism and storage are considered. Through associations between body mass index, chronic neuroinflammation and FTO expression, activation of IO&NS pathways arising from somatoprogression may contribute to neuroprogression. Early evidence highlights the potential of adjuvants targeting IO&NS pathways for treating somatoprogression and neuroprogression.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Adam J Walker
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Andre F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Chiara C Bortolasci
- Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Ken Walder
- Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
50
|
Abstract
Srivastava PK, van Eyll J, Godard P, Mazzuferi M, Delahaye-Duriez A, Steenwinckel JV, et al. A systems-level framework for drug discovery identifies Csf1R as an anti-epileptic drug target. Nat Commun. 2018;9(1):3561. doi:10.1038/s41467-018-06008-4. The identification of drug targets is highly challenging, particularly for diseases of the brain. To address this problem, we developed and experimentally validated a general computational framework for drug target discovery that combines gene regulatory information with causal reasoning (“Causal Reasoning Analytical Framework for Target discovery”-CRAFT). Using a systems genetics approach and starting from gene expression data from the target tissue, CRAFT provides a predictive framework for identifying cell membrane receptors with a direction-specified influence over disease-related gene expression profiles. As proof of concept, we applied CRAFT to epilepsy and predicted the tyrosine kinase receptor Csf1R as a potential therapeutic target. The predicted effect of Csf1R blockade in attenuating epilepsy seizures was validated in 3 preclinical models of epilepsy. These results highlight CRAFT as a systems-level framework for target discovery and suggest Csf1R blockade as a novel therapeutic strategy in epilepsy. The CRAFT is applicable to disease settings other than epilepsy.
Collapse
|