1
|
Nikolopoulos D, Manolakou T, Polissidis A, Filia A, Bertsias G, Koutmani Y, Boumpas DT. Microglia activation in the presence of intact blood-brain barrier and disruption of hippocampal neurogenesis via IL-6 and IL-18 mediate early diffuse neuropsychiatric lupus. Ann Rheum Dis 2023; 82:646-657. [PMID: 36898766 PMCID: PMC10176423 DOI: 10.1136/ard-2022-223506] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/26/2023] [Indexed: 03/12/2023]
Abstract
INTRODUCTION Inflammatory mediators are detected in the cerebrospinal fluid of systemic lupus erythematosus patients with central nervous system involvement (NPSLE), yet the underlying cellular and molecular mechanisms leading to neuropsychiatric disease remain elusive. METHODS We performed a comprehensive phenotyping of NZB/W-F1 lupus-prone mice including tests for depression, anxiety and cognition. Immunofluorescence, flow cytometry, RNA-sequencing, qPCR, cytokine quantification and blood-brain barrier (BBB) permeability assays were applied in hippocampal tissue obtained in both prenephritic (3-month-old) and nephritic (6-month-old) lupus mice and matched control strains. Healthy adult hippocampal neural stem cells (hiNSCs) were exposed ex vivo to exogenous inflammatory cytokines to assess their effects on proliferation and apoptosis. RESULTS At the prenephritic stage, BBB is intact yet mice exhibit hippocampus-related behavioural deficits recapitulating the human diffuse neuropsychiatric disease. This phenotype is accounted by disrupted hippocampal neurogenesis with hiNSCs exhibiting increased proliferation combined with decreased differentiation and increased apoptosis in combination with microglia activation and increased secretion of proinflammatory cytokines and chemokines. Among these cytokines, IL-6 and IL-18 directly induce apoptosis of adult hiNSCs ex vivo. During the nephritic stage, BBB becomes disrupted which facilitates immune components of peripheral blood, particularly B-cells, to penetrate into the hippocampus further augmenting inflammation with locally increased levels of IL-6, IL-12, IL-18 and IL-23. Of note, an interferon gene signature was observed only at nephritic-stage. CONCLUSION An intact BBB with microglial activation disrupting the formation of new neurons within the hippocampus represent early events in NPSLE. Disturbances of the BBB and interferon signature are evident later in the course of the disease.
Collapse
Affiliation(s)
- Dionysis Nikolopoulos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece .,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Manolakou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Anastasia Filia
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - George Bertsias
- Laboratory of Autoimmunity-Inflammation, Institute of Molecular Biology and Biotechnology, Heraklion, Greece.,Rheumatology, Clinical Immunology and Allergy Department, Medical School University of Crete, Heraklion, Greece
| | | | - Dimitrios T Boumpas
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece .,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Medical School, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
2
|
Kot M, Neglur PK, Pietraszewska A, Buzanska L. Boosting Neurogenesis in the Adult Hippocampus Using Antidepressants and Mesenchymal Stem Cells. Cells 2022; 11:cells11203234. [PMID: 36291101 PMCID: PMC9600461 DOI: 10.3390/cells11203234] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
The hippocampus is one of the few privileged regions (neural stem cell niche) of the brain, where neural stem cells differentiate into new neurons throughout adulthood. However, dysregulation of hippocampal neurogenesis with aging, injury, depression and neurodegenerative disease leads to debilitating cognitive impacts. These debilitating symptoms deteriorate the quality of life in the afflicted individuals. Impaired hippocampal neurogenesis is especially difficult to rescue with increasing age and neurodegeneration. However, the potential to boost endogenous Wnt signaling by influencing pathway modulators such as receptors, agonists, and antagonists through drug and cell therapy-based interventions offers hope. Restoration and augmentation of hampered Wnt signaling to facilitate increased hippocampal neurogenesis would serve as an endogenous repair mechanism and contribute to hippocampal structural and functional plasticity. This review focuses on the possible interaction between neurogenesis and Wnt signaling under the control of antidepressants and mesenchymal stem cells (MSCs) to overcome debilitating symptoms caused by age, diseases, or environmental factors such as stress. It will also address some current limitations hindering the direct extrapolation of research from animal models to human application, and the technical challenges associated with the MSCs and their cellular products as potential therapeutic solutions.
Collapse
Affiliation(s)
- Marta Kot
- Correspondence: ; Tel.: +48-22-60-86-563
| | | | | | | |
Collapse
|
3
|
CCL4 induces inflammatory signalling and barrier disruption in the neurovascular endothelium. Brain Behav Immun Health 2021; 18:100370. [PMID: 34755124 PMCID: PMC8560974 DOI: 10.1016/j.bbih.2021.100370] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022] Open
Abstract
Background During neuroinflammation many chemokines alter the function of the blood-brain barrier (BBB) that regulates the entry of macromolecules and immune cells into the brain. As the milieu of the brain is altered, biochemical and structural changes contribute to the pathogenesis of neuroinflammation and may impact on neurogenesis. The chemokine CCL4, previously known as MIP-1β, is upregulated in a wide variety of central nervous system disorders, including multiple sclerosis, where it is thought to play a key role in the neuroinflammatory process. However, the effect of CCL4 on BBB endothelial cells (ECs) is unknown. Materials and methods Expression and distribution of CCR5, phosphorylated p38, F-actin, zonula occludens-1 (ZO-1) and vascular endothelial cadherin (VE-cadherin) were analysed in the human BBB EC line hCMEC/D3 by Western blot and/or immunofluorescence in the presence and absence of CCL4. Barrier modulation in response to CCL4 using hCMEC/D3 monolayers was assessed by measuring molecular flux of 70 kDa RITC-dextran and transendothelial lymphocyte migration. Permeability changes in response to CCL4 in vivo were measured by an occlusion technique in pial microvessels of Wistar rats and by fluorescein angiography in mouse retinae. Results CCR5, the receptor for CCL4, was expressed in hCMEC/D3 cells. CCL4 stimulation led to phosphorylation of p38 and the formation of actin stress fibres, both indicative of intracellular chemokine signalling. The distribution of junctional proteins was also altered in response to CCL4: junctional ZO-1 was reduced by circa 60% within 60 min. In addition, surface VE-cadherin was redistributed through internalisation. Consistent with these changes, CCL4 induced hyperpermeability in vitro and in vivo and increased transmigration of lymphocytes across monolayers of hCMEC/D3 cells. Conclusion These results show that CCL4 can modify BBB function and may contribute to disease pathogenesis. The chemokine CCL4 induced phosphorylation of P38 in an in vitro model of the blood-brain barrier (BBB). CCL4 treatment resulted in reduction of plasma membrane VE-cadherin and junctional ZO-1. CCL4 induced neurovascular barrier breakdown in vitro and in vivo.
Collapse
|
4
|
The effects of genotype on inflammatory response in hippocampal progenitor cells: A computational approach. Brain Behav Immun Health 2021; 15:100286. [PMID: 34345870 PMCID: PMC8261829 DOI: 10.1016/j.bbih.2021.100286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023] Open
Abstract
Cell culture models are valuable tools to study biological mechanisms underlying health and disease in a controlled environment. Although their genotype influences their phenotype, subtle genetic variations in cell lines are rarely characterised and taken into account for in vitro studies. To investigate how the genetic makeup of a cell line might affect the cellular response to inflammation, we characterised the single nucleotide variants (SNPs) relevant to inflammation-related genes in an established hippocampal progenitor cell line (HPC0A07/03C) that is frequently used as an in vitro model for hippocampal neurogenesis (HN). SNPs were identified using a genotyping array, and genes associated with chronic inflammatory and neuroinflammatory response gene ontology terms were retrieved using the AmiGO application. SNPs associated with these genes were then extracted from the genotyping dataset, for which a literature search was conducted, yielding relevant research articles for a total of 17 SNPs. Of these variants, 10 were found to potentially affect hippocampal neurogenesis whereby a majority (n=7) is likely to reduce neurogenesis under inflammatory conditions. Taken together, the existing literature seems to suggest that all stages of hippocampal neurogenesis could be negatively affected due to the genetic makeup in HPC0A07/03C cells under inflammation. Additional experiments will be needed to validate these specific findings in a laboratory setting. However, this computational approach already confirms that in vitro studies in general should control for cell lines subtle genetic variations which could mask or exacerbate findings.
Collapse
|
5
|
Hammond JW, Bellizzi MJ, Ware C, Qiu WQ, Saminathan P, Li H, Luo S, Ma SA, Li Y, Gelbard HA. Complement-dependent synapse loss and microgliosis in a mouse model of multiple sclerosis. Brain Behav Immun 2020; 87:739-750. [PMID: 32151684 PMCID: PMC8698220 DOI: 10.1016/j.bbi.2020.03.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, neurodegenerative disease of the CNS characterized by both grey and white matter injury. Microglial activation and a reduction in synaptic density are key features of grey matter pathology that can be modeled with MOG35-55 experimental autoimmune encephalomyelitis (EAE). Complement deposition combined with microglial engulfment has been shown during normal development and in disease as a mechanism for pruning synapses. We tested whether there is excess complement production in the EAE hippocampus and whether complement-dependent synapse loss is a source of degeneration in EAE using C1qa and C3 knockout mice. We found that C1q and C3 protein and mRNA levels were elevated in EAE mice. Genetic loss of C3 protected mice from EAE-induced synapse loss, reduced microglial activation, decreased the severity of the EAE clinical score, and protected memory/freezing behavior after contextual fear conditioning. C1qa KO mice with EAE showed little to no change on these measurements compared to WT EAE mice. Thus, pathologic expression and activation of the early complement pathway, specifically at the level of C3, contributes to hippocampal grey matter pathology in the EAE.
Collapse
Affiliation(s)
- Jennetta W. Hammond
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Correspondence: Jennetta W. Hammond,
University of Rochester, Center for Neurotherapeutics Discovery, 601 Elmwood
Avenue, Box 645, Rochester, NY 14642, USA,
, Phone:
1-585-273-2872
| | - Matthew J. Bellizzi
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Caroline Ware
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Wen Q. Qiu
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Priyanka Saminathan
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Microbiology and Immunology, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Herman Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Shaopeiwen Luo
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Stefanie A. Ma
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Yuanhao Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| |
Collapse
|
6
|
das Neves SP, Santos G, Barros C, Pereira DR, Ferreira R, Mota C, Monteiro S, Fernandes A, Marques F, Cerqueira JJ. Enhanced cognitive performance in experimental autoimmune encephalomyelitis mice treated with dimethyl fumarate after the appearance of disease symptoms. J Neuroimmunol 2020; 340:577163. [PMID: 31982706 DOI: 10.1016/j.jneuroim.2020.577163] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/02/2020] [Accepted: 01/10/2020] [Indexed: 12/23/2022]
Abstract
In multiple sclerosis (MS), cognitive dysfunction is common but difficult to treat. We analyzed the impact of dimethyl fumarate, an MS drug with neuroprotective properties, in spatial memory performance in a mouse model of MS and looked for structural correlates in the hippocampus. Treated mice presented better cognitive performance which was not associated with structural hippocampal damage but with decreased demyelination in the fimbria. Dimethyl fumarate, even if initiated after hindlimb paralysis, ameliorated memory deficits in the MS mouse model due, at least in part, to its positive impact in the demyelination of the main hippocampal output pathway.
Collapse
Affiliation(s)
- Sofia P das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Gisela Santos
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Catarina Barros
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Diana Rodrigues Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Mota
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adelaide Fernandes
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João José Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 2CA, Clinical Academic Center - Braga, Braga, Portugal.
| |
Collapse
|
7
|
Lin K, Shao R, Wang R, Lu W, Zou W, Chen K, Gao Y, Brietzke E, McIntyre RS, Mansur RB, Zhang L, Yau SY, Su H, Xu G, So KF. Inflammation, brain structure and cognition interrelations among individuals with differential risks for bipolar disorder. Brain Behav Immun 2020; 83:192-199. [PMID: 31614176 DOI: 10.1016/j.bbi.2019.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/27/2019] [Accepted: 10/11/2019] [Indexed: 12/11/2022] Open
Abstract
Neuro-inflammation might impact on clinical manifestations and cognition function via changing the volumes of key brain structures such as the anterior cingulate cortex (ACC) in bipolar disorder (BD). In this study, we investigated the interrelations among interleukin (IL)-6 cytokine level, grey matter (GM) volume of the anterior cingulated cortex (ACC), and attention function among offspring of parents diagnosed with BD. The offspring were categorized as being either asymptomatic or symptomatic based on whether they manifested pre-defined sub-threshold mood symptoms. We found that the symptomatic offspring showed significantly higher serum levels of IL-6 than the asymptomatic offspring (F(1, 59) = 67.65, p < 0.001). On the brain level, we obtained significant interactive effect of group and IL6 level on the ACC GM (PFWE = 0.017). Specifically, the GM volume of the rostral ACC was negatively associated with the levels of IL-6 in the asymptomatic offspring (PFWE = 0.021), but not the symptomatic offspring (PFWE > 0.05). Mediation analyses revealed that the GM volume of the rostral ACC significantly mediated the negative association between the IL-6 levels and attention performance in the asymptomatic offspring (bootstrapping Confidence Interval (CI) = -6.0432 to -0.0731) but not the symptomatic offspring (bootstrapping CI = -0.3197 to 1.3423). Our data suggest that the asymptomatic and symptomatic bipolar offspring may exhibit different neurocognitive-inflammatory profiles, which could be further validated as viable biosignatures for BD risk and resilience.
Collapse
Affiliation(s)
- Kangguang Lin
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Academician Workstation of Mood and Brain Sciences, Guangzhou Medical University, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China; Laboratory of Neuropsychology and Laboratory of Social Cognitive Affective Neuroscience, Department of Psychology, University of Hong Kong, Hong Kong.
| | - Robin Shao
- Laboratory of Neuropsychology and Laboratory of Social Cognitive Affective Neuroscience, Department of Psychology, University of Hong Kong, Hong Kong.
| | - Runhua Wang
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Academician Workstation of Mood and Brain Sciences, Guangzhou Medical University, China
| | - Weicong Lu
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Wenjin Zou
- Department of Radiology, The Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kun Chen
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Academician Workstation of Mood and Brain Sciences, Guangzhou Medical University, China
| | - Yanling Gao
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Academician Workstation of Mood and Brain Sciences, Guangzhou Medical University, China
| | - Elisa Brietzke
- Department of Psychiatry, Queen's University, School of Medicine, Kingston, ON, Canada
| | - Roger S McIntyre
- Academician Workstation of Mood and Brain Sciences, Guangzhou Medical University, China; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Canada
| | - Li Zhang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Huanxing Su
- Academician Workstation of Mood and Brain Sciences, Guangzhou Medical University, China
| | - Guiyun Xu
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Kwok-Fai So
- Academician Workstation of Mood and Brain Sciences, Guangzhou Medical University, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China; The State Key Laboratory of Brain and Cognitive Sciences and Department of Ophthalmology, University of Hong Kong, Hong Kong.
| |
Collapse
|
8
|
Gold Nanoparticles Treatment Reverses Brain Damage in Alzheimer’s Disease Model. Mol Neurobiol 2019; 57:926-936. [DOI: 10.1007/s12035-019-01780-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/09/2019] [Indexed: 12/20/2022]
|
9
|
Tobore TO. On elucidation of the role of mitochondria dysfunction and oxidative stress in multiple sclerosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/ncn3.12335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
Zong X, Dong Y, Li Y, Yang L, Li Y, Yang B, Tucker L, Zhao N, Brann DW, Yan X, Hu S, Zhang Q. Beneficial Effects of Theta-Burst Transcranial Magnetic Stimulation on Stroke Injury via Improving Neuronal Microenvironment and Mitochondrial Integrity. Transl Stroke Res 2019; 11:450-467. [PMID: 31515743 DOI: 10.1007/s12975-019-00731-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/19/2022]
Abstract
Recent work suggests that repetitive transcranial magnetic stimulation (rTMS) may beneficially alter the pathological status of several neurological disorders, although the mechanism remains unclear. The current study was designed to investigate the effects of rTMS on behavioral deficits and potential underlying mechanisms in a rat photothrombotic (PT) stroke model. From day 0 (3 h) to day 5 after the establishment of PT stroke, 5-min daily continuous theta-burst rTMS (3 pulses of 50 Hz repeated every 200 ms, intensity at 200 G) was applied on the infarct hemisphere. We report that rTMS significantly attenuated behavioral deficits and infarct volume after PT stroke. Further investigation demonstrated that rTMS remarkably reduced synaptic loss and neuronal degeneration in the peri-infarct cortical region. Mechanistic studies displayed that beneficial effects of rTMS were associated with robust suppression of reactive micro/astrogliosis and the overproduction of pro-inflammatory cytokines, as well as oxidative stress and oxidative neuronal damage especially at the late stage following PT stroke. Intriguingly, rTMS could effectively induce a shift in microglial M1/M2 phenotype activation and an A1 to A2 switch in astrocytic phenotypes. In addition, the release of anti-inflammatory cytokines and mitochondrial MnSOD in peri-infarct regions were elevated following rTMS treatment. Finally, rTMS treatment efficaciously preserved mitochondrial membrane integrity and suppressed the intrinsic mitochondrial caspase-9/3 apoptotic pathway within the peri-infarct cortex. Our novel findings indicate that rTMS treatment exerted robust neuroprotection when applied at least 3 h after ischemic stroke. The underlying mechanisms are partially associated with improvement of the local neuronal microenvironment by altering inflammatory and oxidative status and preserving mitochondrial integrity in the peri-infarct zone. These findings provide strong support for the promising therapeutic effect of rTMS against ischemic neuronal injury and functional deficits following stroke.
Collapse
Affiliation(s)
- Xuemei Zong
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University; the Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu province, China.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yuyu Li
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University; the Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu province, China
| | - Luodan Yang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yong Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Baocheng Yang
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University; the Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu province, China.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Lorelei Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Ningjun Zhao
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University; the Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu province, China
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Xianliang Yan
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University; the Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu province, China
| | - Shuqun Hu
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University; the Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu province, China.
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
11
|
Raeis-Abdollahi E, Nabavizadeh F, Tajik L, Sadeghipour HR. Effects of prenatal exposure to chrysotile asbestos on hippocampal neurogenesis and long-term behavioral changes in adult male rat offspring. Behav Brain Res 2019; 371:111962. [PMID: 31116961 DOI: 10.1016/j.bbr.2019.111962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 02/06/2023]
Abstract
Prenatal development is a critical period of life that many environmental pollutants have been suggested to influence fetal growth. Nevertheless, there are still a few investigations into the prenatal exposure to chrysotile asbestos and its neurodevelopmental and behavioral outcome in offspring. In this study, twenty-eight pregnant Wistar rats were divided into four groups and received three-times repeated intraperitoneal injections of normal saline, chrysotile, ascorbic acid and the combination of chrysotile and ascorbic acid on gestational days 11, 14 and 17. The maternal serum levels of malondialdehyde (MDA) and prooxidant-antioxidant balance (PAB) and hippocampal MDA content in adult male offspring were measured. At postnatal day (PND) 60, elevated plus maze was performed to determine anxiety-like behavior, also depression-like behavior was examined using a forced swim test at PND 61- 62. Thereafter, the quantitative analysis of Ki-67, NeuN and GFAP positive cells in the hippocampal dentate gyrus were studied by immunostaining. Our data showed that prenatal exposure to chrysotile increased the maternal serum level of MDA and PAB as well as hippocampal MDA content in adult male offspring, also increased the depression- and anxiety-like behaviors of adult male offspring and decreased the hippocampal Ki-67+, NeuN+ and GFAP+ cells in dantate gyrus of adult male offspring. However, co-administration of ascorbic acid and chrysotile decreased hippocampal lipid peroxidation and increased the Ki-67+, NeuN+ and GFAP+ cells in adult male offspring. In summary, these results indicated that oxidative stress induced by prenatal exposure to chrysotile, lead to the long-lasting decrease of the hippocampal cell proliferation and neuronal differentiation as well as astrogliosis of adult male offspring that exhibit more depression- and anxiety-like behaviors in adulthood and co-treatment of ascorbic acid with chrysotile asbestos attenuated the changes.
Collapse
Affiliation(s)
- Ehsan Raeis-Abdollahi
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Tajik
- Department of Biology, Payame Noor University, Tehran, Iran
| | - Hamid Reza Sadeghipour
- Electrophysiology Research Centre, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Abstract
The fovea centralis, an anatomically concave pit located at the center of the macula, is avascular, hypoxic, and characteristic of stem-cell niches of other tissues. We hypothesized that in the fovea, undifferentiated retinal-stem-cell-like cells may exist, and that neurogenesis may occur. Hence, we performed an immunohistological study using cynomolgus monkey retinas. After preparing frozen tissue sections of the retina including the foveal pit, immunostaining was performed for glial fibrillary acidic protein (GFAP), nestin, vimentin, neuron-specific class III β-tubulin (Tuj-1), arrestin 4, neurofilament, CD117, CD44, Ki67, and cellular retinaldehyde-binding protein (CRALBP), followed by fluorescence and/or confocal microscopy examinations. Immunostaining of the tissue sections enabled clear observation of strongly GFAP-positive cells that corresponded to the inner-half layer of the foveolar Müller cell cone. The surface layer of the foveal slope was partially costained with GFAP and vimentin. Tuj-1-positive cells were observed in the innermost layer of the foveolar retina, which spanned to the surrounding ganglion cell layer. Moreover, colocalization of Tuj-1 and GFAP was observed at the foveal pit. The coexpression of CD117 and CD44 was found in the interphotoreceptor matrix of the fovea. The foveolar cone stained positive for both nestin and arrestin 4, however, the photoreceptor layer outside of the foveola displayed weak staining for nestin. Colocalization of nestin and vimentin was observed in the inner half of the Henle layer, while colocalization of nestin and neurofilament was observed in the outer half, predominantly. Scattered Ki67-positive cells were observed in the cellular processes of the outer plexiform layer and the ganglion cell layer around the foveola. Immunostaining for CRALBP was negative in most parts of the GFAP-positive area. The Müller cell cone was divided into GFAP-strongly positive cells, presumably astrocytes, in the inner layer and nestin-positive/GFAP-weakly positive radial glia-like cells in the outer layer. These findings indicated that groups of such undifferentiated cells in the foveola might be involved in maintaining morphology and regeneration.
Collapse
|
13
|
Rocca MA, Barkhof F, De Luca J, Frisén J, Geurts JJG, Hulst HE, Sastre-Garriga J, Filippi M. The hippocampus in multiple sclerosis. Lancet Neurol 2018; 17:918-926. [PMID: 30264730 DOI: 10.1016/s1474-4422(18)30309-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 07/18/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022]
Abstract
Some of the clinical manifestations of multiple sclerosis, such as memory impairment and depression, are, at least partly, related to involvement of the hippocampus. Pathological studies have shown extensive demyelination, neuronal damage, and synaptic abnormalities in the hippocampus of patients with multiple sclerosis, and improvements in MRI technology have provided novel ways to assess hippocampal involvement in vivo. It is now accepted that clinical manifestations related to the hippocampus are due not only to focal hippocampal damage, but also to disconnection of the hippocampus from several brain networks. Evidence suggests anatomical and functional subspecialisation of the different hippocampal subfields, resulting in variability between regions in the extent to which damage and repair occur. The hippocampus also has important roles in plasticity and neurogenesis, both of which potentially contribute to functional preservation and restoration. These findings underline the importance of evaluation of the hippocampus not only to improve understanding of the clinical manifestations of multiple sclerosis, but also as a potential future target for treatment.
Collapse
Affiliation(s)
- Maria A Rocca
- Neuroimaging Research Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-location VUmc, Amsterdam, Netherlands; Institute of Neurology and Institute of Healthcare Engineering, UCL Institute of Neurology, London, UK
| | - John De Luca
- Kessler Foundation, West Orange, NJ, USA; Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institute, Sweden
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-location VUmc, Amsterdam, Netherlands
| | - Hanneke E Hulst
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-location VUmc, Amsterdam, Netherlands
| | - Jaume Sastre-Garriga
- Department of Neurology/Neuroimmunology, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.
| | | |
Collapse
|
14
|
Cacciaguerra L, Pagani E, Mesaros S, Dackovic J, Dujmovic-Basuroski I, Drulovic J, Valsasina P, Filippi M, Rocca MA. Dynamic volumetric changes of hippocampal subfields in clinically isolated syndrome patients: A 2-year MRI study. Mult Scler 2018; 25:1232-1242. [DOI: 10.1177/1352458518787347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background:Different subregional patterns of hippocampal involvement have been observed in diverse multiple sclerosis (MS) phenotypes.Objective:To evaluate the occurrence of regional hippocampal variations in clinically isolated syndrome (CIS) patients, their relationships with focal white matter (WM) lesions, and their prognostic implications.Methods:Brain dual-echo and three-dimensional (3D) T1-weighted scans were acquired from 14 healthy controls and 36 CIS patients within 2 months from clinical onset and after 3, 12, and 24 months. Radial distance distribution was assessed using 3D parametric surface mesh models. A cognitive screening was also performed.Results:Patients showed clusters of reduced radial distance in the Cornu Ammonis 1 from month 3, progressively extending to the subiculum, negatively correlated with ipsilateral T2 and T1 lesion volume. Increased radial distance appeared in the right dentate gyrus after 3 ( p < 0.05), 12, and 24 ( p < 0.001) months, and in the left one after 3 and 24 months ( p < 0.001), positively correlated with lesional measures. Hippocampal volume variations were more pronounced in patients converting to MS after 24 months and did not correlate with cognitive performance.Conclusion:Regional hippocampal changes occur in CIS, are more pronounced in patients converting to MS, and are modulated by focal WM lesions.
Collapse
Affiliation(s)
- Laura Cacciaguerra
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy/Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Sharlota Mesaros
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Dackovic
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Jelena Drulovic
- Clinic of Neurology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Paola Valsasina
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy/Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Assunta Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy/Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
15
|
Wang Z, He X, Fan X. Postnatal administration of memantine rescues TNF-α-induced decreased hippocampal precursor proliferation. Neurosci Lett 2018; 662:173-180. [DOI: 10.1016/j.neulet.2017.10.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/19/2017] [Accepted: 10/13/2017] [Indexed: 01/16/2023]
|
16
|
Yang L, Tucker D, Dong Y, Wu C, Lu Y, Li Y, Zhang J, Liu TCY, Zhang Q. Photobiomodulation therapy promotes neurogenesis by improving post-stroke local microenvironment and stimulating neuroprogenitor cells. Exp Neurol 2017; 299:86-96. [PMID: 29056360 DOI: 10.1016/j.expneurol.2017.10.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/27/2017] [Accepted: 10/17/2017] [Indexed: 12/24/2022]
Abstract
Recent work has indicated that photobiomodulation (PBM) may beneficially alter the pathological status of several neurological disorders, although the mechanism currently remains unclear. The current study was designed to investigate the beneficial effect of PBM on behavioral deficits and neurogenesis in a photothrombotic (PT) model of ischemic stroke in rats. From day 1 to day 7 after the establishment of PT model, 2-minute daily PBM (CW, 808nm, 350mW/cm2, total 294J at scalp level) was applied on the infarct injury area (1.8mm anterior to the bregma and 2.5mm lateral from the midline). Rats received intraperitoneal injections of 5-bromodeoxyuridine (BrdU) twice daily (50mg/kg) from day 2 to 8 post-stoke, and samples were collected at day 14. We demonstrated that PBM significantly attenuated behavioral deficits and infarct volume induced by PT stroke. Further investigation displayed that PBM remarkably enhanced neurogenesis and synaptogenesis, as evidenced by immunostaining of BrdU, Ki67, DCX, MAP2, spinophilin, and synaptophysin. Mechanistic studies suggested beneficial effects of PBM were accompanied by robust suppression of reactive gliosis and the production of pro-inflammatory cytokines. On the contrary, the release of anti-inflammatory cytokines, cytochrome c oxidase activity and ATP production in peri-infarct regions were elevated following PBM treatment. Intriguingly, PBM could effectively switch an M1 microglial phenotype to an anti-inflammatory M2 phenotype. Our novel findings indicated that PBM is capable of promoting neurogenesis after ischemic stroke. The underlying mechanisms may rely on: 1) promotion of proliferation and differentiation of internal neuroprogenitor cells in the peri-infarct zone; 2) improvement of the neuronal microenvironment by altering inflammatory status and promoting mitochondrial function. These findings provide strong support for the promising therapeutic effect of PBM on neuronal repair following ischemic stroke.
Collapse
Affiliation(s)
- Luodan Yang
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, Guangzhou, China; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Chongyun Wu
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Yujiao Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yong Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Juan Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Quanguang Zhang
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, Guangzhou, China; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
17
|
High dose tetrabromobisphenol A impairs hippocampal neurogenesis and memory retention. Food Chem Toxicol 2017; 106:223-231. [PMID: 28564613 DOI: 10.1016/j.fct.2017.05.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 12/20/2022]
|