1
|
Šakić B. The MRL Model: A Valuable Tool in Studies of Autoimmunity-Brain Interactions. Methods Mol Biol 2025; 2868:221-246. [PMID: 39546233 DOI: 10.1007/978-1-0716-4200-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The link between systemic autoimmunity, brain pathology, and aberrant behavior is still largely unexplored field of biomedical science. Accumulating evidence points to causal relationships between immune factors, neurodegeneration, and neuropsychiatric manifestations. By documenting autoimmunity-associated neuronal degeneration and cytotoxicity of the cerebrospinal fluid from disease-affected subjects, the murine MRL model has shown high validity in revealing principal pathogenic circuits. In addition, unlike any other autoimmune strain, MRL mice produce antibodies commonly found in patients suffering from lupus and other autoimmune disorders. This review highlights the importance of the MRL model as a useful preparation for understanding the links between the immune system and brain function.
Collapse
Affiliation(s)
- Boris Šakić
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
2
|
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: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/26/2023] [Indexed: 03/12/2023]
Abstract
INTRODUCTION Inflammatory mediators are detected in the cerebrospinal fluid of systemic lupus erythematosus patients with central nervous system involvement (NPSLE), yet the underlying cellular and molecular mechanisms leading to neuropsychiatric disease remain elusive. METHODS We performed a comprehensive phenotyping of NZB/W-F1 lupus-prone mice including tests for depression, anxiety and cognition. Immunofluorescence, flow cytometry, RNA-sequencing, qPCR, cytokine quantification and blood-brain barrier (BBB) permeability assays were applied in hippocampal tissue obtained in both prenephritic (3-month-old) and nephritic (6-month-old) lupus mice and matched control strains. Healthy adult hippocampal neural stem cells (hiNSCs) were exposed ex vivo to exogenous inflammatory cytokines to assess their effects on proliferation and apoptosis. RESULTS At the prenephritic stage, BBB is intact yet mice exhibit hippocampus-related behavioural deficits recapitulating the human diffuse neuropsychiatric disease. This phenotype is accounted by disrupted hippocampal neurogenesis with hiNSCs exhibiting increased proliferation combined with decreased differentiation and increased apoptosis in combination with microglia activation and increased secretion of proinflammatory cytokines and chemokines. Among these cytokines, IL-6 and IL-18 directly induce apoptosis of adult hiNSCs ex vivo. During the nephritic stage, BBB becomes disrupted which facilitates immune components of peripheral blood, particularly B-cells, to penetrate into the hippocampus further augmenting inflammation with locally increased levels of IL-6, IL-12, IL-18 and IL-23. Of note, an interferon gene signature was observed only at nephritic-stage. CONCLUSION An intact BBB with microglial activation disrupting the formation of new neurons within the hippocampus represent early events in NPSLE. Disturbances of the BBB and interferon signature are evident later in the course of the disease.
Collapse
Affiliation(s)
- Dionysis Nikolopoulos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece .,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Manolakou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Anastasia Filia
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - George Bertsias
- Laboratory of Autoimmunity-Inflammation, Institute of Molecular Biology and Biotechnology, Heraklion, Greece.,Rheumatology, Clinical Immunology and Allergy Department, Medical School University of Crete, Heraklion, Greece
| | | | - Dimitrios T Boumpas
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece .,School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Medical School, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
3
|
Bendorius M, Po C, Muller S, Jeltsch-David H. From Systemic Inflammation to Neuroinflammation: The Case of Neurolupus. Int J Mol Sci 2018; 19:E3588. [PMID: 30428632 PMCID: PMC6274746 DOI: 10.3390/ijms19113588] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022] Open
Abstract
It took decades to arrive at the general consensus dismissing the notion that the immune system is independent of the central nervous system. In the case of uncontrolled systemic inflammation, the relationship between the two systems is thrown off balance and results in cognitive and emotional impairment. It is specifically true for autoimmune pathologies where the central nervous system is affected as a result of systemic inflammation. Along with boosting circulating cytokine levels, systemic inflammation can lead to aberrant brain-resident immune cell activation, leakage of the blood⁻brain barrier, and the production of circulating antibodies that cross-react with brain antigens. One of the most disabling autoimmune pathologies known to have an effect on the central nervous system secondary to the systemic disease is systemic lupus erythematosus. Its neuropsychiatric expression has been extensively studied in lupus-like disease murine models that develop an autoimmunity-associated behavioral syndrome. These models are very useful for studying how the peripheral immune system and systemic inflammation can influence brain functions. In this review, we summarize the experimental data reported on murine models developing autoimmune diseases and systemic inflammation, and we explore the underlying mechanisms explaining how systemic inflammation can result in behavioral deficits, with a special focus on in vivo neuroimaging techniques.
Collapse
Affiliation(s)
- Mykolas Bendorius
- UMR 7242 Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg (ESBS), Laboratoire d'Excellence Médalis, Université de Strasbourg/CNRS, 67412 Illkirch, France.
| | - Chrystelle Po
- ICube UMR 7357, Université de Strasbourg/CNRS, Fédération de Médecine Translationnelle de Strasbourg, 67000 Strasbourg, France.
| | - Sylviane Muller
- UMR 7242 Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg (ESBS), Laboratoire d'Excellence Médalis, Université de Strasbourg/CNRS, 67412 Illkirch, France.
- University of Strasbourg Institute for Advanced Study (USIAS), 67000 Strasbourg, France.
| | - Hélène Jeltsch-David
- UMR 7242 Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg (ESBS), Laboratoire d'Excellence Médalis, Université de Strasbourg/CNRS, 67412 Illkirch, France.
| |
Collapse
|
4
|
Frauenknecht K, Leukel P, Weiss R, von Pein HD, Katzav A, Chapman J, Sommer CJ. Decreased hippocampal cell proliferation in mice with experimental antiphospholipid syndrome. Brain Struct Funct 2018; 223:3463-3471. [PMID: 29936552 DOI: 10.1007/s00429-018-1699-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/14/2018] [Indexed: 12/31/2022]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the presence of antiphospholipid antibodies, which may trigger vascular thrombosis with consecutive infarcts. However, cognitive dysfunctions representing one of the most commonest neuropsychiatric symptoms are frequently present despite the absence of any ischemic brain lesions. Data on the structural and functional basis of the neuropsychiatric symptoms are sparse. To examine the effect of APS on hippocampal neurogenesis and on white matter, we induced experimental APS (eAPS) in adult female Balb/C mice by immunization with β2-glycoprotein 1. To investigate cell proliferation in the dentate gyrus granular cell layer (DG GCL), eAPS and control mice (n = 5, each) were injected with 5-bromo-2'-deoxyuridine (BrdU) once a day for 10 subsequent days. Sixteen weeks after immunization, eAPS resulted in a significant reduction of BrdU-positive cells in the DG GCL compared to control animals. However, double staining with doublecortin and NeuN revealed a largely preserved neurogenesis. Ultrastructural analysis of corpus callosum (CC) axons in eAPS (n = 6) and control mice (n = 7) revealed no significant changes in CC axon diameter or g-ratio. In conclusion, decreased cellular proliferation in the hippocampus of eAPS mice indicates a limited regenerative potential and may represent one neuropathological substrate of cognitive changes in APS while evidence for alterations of white matter integrity is lacking.
Collapse
Affiliation(s)
- Katrin Frauenknecht
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany. .,Institute of Neuropathology, University Hospital of Zurich, University of Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland.
| | - Petra Leukel
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Ronen Weiss
- Department of Neurology, Chaim Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Hashomer, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Harald D von Pein
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Aviva Katzav
- Department of Neurology, Chaim Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Joab Chapman
- Department of Neurology, Chaim Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Clemens J Sommer
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Focus Program Translational Neurosciences (FTN), Rhine Main Neuroscience Network (rmn2), Mainz, Germany
| |
Collapse
|
5
|
Leung JWH, Lau BWM, Chan VSF, Lau CS, So KF. Abnormal increase of neuronal precursor cells and exacerbated neuroinflammation in the corpus callosum in murine model of systemic lupus erythematosus. Restor Neurol Neurosci 2018; 34:443-53. [PMID: 27163251 PMCID: PMC4927870 DOI: 10.3233/rnn-160638] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Purpose: Systemic Lupus Erythematosus (SLE) is an autoimmune disease which is characterised by elevated levels of autoantibodies and cytokines in the body. Via alteration of the regulation of inflammation, damage to different organ systems, including the central nervous system (CNS), was found in SLE patients. Patients diagnosed with SLE were reported to suffer from different kinds of psychiatric signs and symptoms. As neurogenesis has been suggested to be a potential key player of psychiatric symptoms and emotional behavior disturbances, this study aims to investigate whether neurogenesis is altered in an animal model of SLE. Also, neuroinflammation was studied. Methods: Female NZB/W F1 mice were used as an animal model of SLE. Animals were divided into two groups: 1. pre-diseased mice (lupus-prone NZB/W F1 female mice, age 10–15 weeks, negative for proteinuria and with basal levels of serum anti-dsDNA autoantibodies) and 2. diseased mice (NZB/W F1 female mice, > 25 weeks of age, with elevated serum levels of anti-dsDNA autoantibodies and with persistent proteinuria of > 3 mg/ml for more than 2 weeks). Comparisons of the levels of neurogenesis and neuroinflammtion between two groups of mice were studied by the immunohistochemistry. Results: After the onset of SLE symptoms, a reduction of neurogenesis in the hippocampus was found, while there was a dramatic increase of doublecortin (DCX+) neuronal precursor cells in the corpus callosum (CC) and in the subventricular zone (SVZ). Meanwhile, exacerbated inflammation was present in the corpus callosum of the diseased mice, which was suggested by the increased number of GFAP+ cells and IBA-1+ cells. Conclusions: To the best of our knowledge, this is the first study showing an increase of neuronal precursor cells in the corpus callosum of the female NZB/W F1 mice. The present study suggests a coincidence but not a causal relationship between neurogenesis and neuroinflammation. The present results have also provided new insight showing that the altered neurogenesis and neuroinflammation may be a potential neurological mechanism for the cognitive and mood disturbance found in the SLE patients.
Collapse
Affiliation(s)
- Joseph Wai-Hin Leung
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Benson Wui-Man Lau
- Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong
| | - Vera Sau-Fong Chan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Chak-Sing Lau
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Kwok-Fai So
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong.,State Key Laboratory of Brain and Cognitive Science, The University of Hong Kong, Hong Kong.,GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, China.,Ministry of Education CNS Regeneration International Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| |
Collapse
|
6
|
Abstract
The link between systemic autoimmunity, brain pathology, and aberrant behavior is still a largely unexplored field of biomedical science. Accumulating evidence points to causal relationships between immune factors, neurodegeneration, and neuropsychiatric manifestations. By documenting autoimmunity-associated neuronal degeneration and cytotoxicity of the cerebrospinal fluid from disease-affected subjects, the murine MRL model had shown high validity in revealing principal pathogenic circuits. In addition, unlike any other autoimmune strain, MRL mice produce antibodies commonly found in patients suffering from lupus and other autoimmune disorders. This review highlights importance of the MRL model as a useful preparation in understanding the links between immune system and brain function.
Collapse
Affiliation(s)
- Boris Šakić
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
7
|
Zimmermann N, Goulart Corrêa D, Tukamoto G, Netto T, Batista Pereira D, Paz Fonseca R, Gasparetto EL. Brain morphology and cortical thickness variations in systemic lupus erythematosus patients: Differences among neurological, psychiatric, and nonneuropsychiatric manifestations. J Magn Reson Imaging 2016; 46:150-158. [PMID: 27862544 DOI: 10.1002/jmri.25538] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/18/2016] [Indexed: 12/28/2022] Open
Abstract
PURPOSE To determine whether systemic lupus erythematosus (SLE) affecting subcortical white matter volumes, deep gray matter volumes, and cortical thickness differ between groups of SLE patients with psychiatric (P-SLE), neurological (N-SLE), or nonneuropsychiatric (non-NPSLE) presentations. MATERIALS AND METHODS Sixty-seven participants were divided into three groups (P-SLE [n = 19], N-SLE [n = 12], and non-NPSLE [n = 36]) and examined with a 1.5T MRI scanner. The images were segmented in FreeSurfer software into volumetric and cortical thickness measures using T1 3D magnetization prepared rapid gradient echo-weighted imaging. For comparative analyses of volume, multivariate analyses of covariance (MANCOVA) were applied followed by Bonferroni post-hoc tests, with age as a covariate. For cortical thickness analyses, the groups were compared with the Query Design Estimate Contrast tool adjusted for age. RESULTS Globus pallidus volumes in both left (P ≤ 0.01) and right (P ≤ 0.05) hemispheres were larger in the N-SLE group than in the non-NPSLE group, and the left GP volume was greater in the N-SLE group than in the P-SLE group (P ≤ 0.05) (MANCOVA, post-hoc Bonferroni). The P-SLE group presented with thinning of cortical areas relative to the N-SLE (predominantly in the left parietal and right frontal and parietal regions) (P ≤ 0.05) and non-NPSLE (predominantly in parietal and occipital regions) (P ≤ 0.05) groups, whereas the N-SLE group presented with thickening of cortical areas (mostly right frontal and left parietal regions) relative to the non-NPSLE (P ≤ 0.05) and P-SLE groups. CONCLUSION N-SLE patients had greater local volumes and cortical thicknesses than the other two groups, whereas P-SLE patients presented with decreased volumes and cortical thinning. These findings provide evidence of distinct neuroanatomical abnormalities in neurological versus psychiatric manifestations of SLE. LEVEL OF EVIDENCE 2 Technical Efficacy: Stage 3 J. MAGN. RESON. IMAGING 2017;46:150-158.
Collapse
Affiliation(s)
- Nicolle Zimmermann
- Federal University of Rio de Janeiro, Department of Radiology, Rio de Janeiro, Brazil
| | - Diogo Goulart Corrêa
- Federal University of Rio de Janeiro, Department of Radiology, Rio de Janeiro, Brazil
| | - Gustavo Tukamoto
- Clínica de Diagnóstico Por Imagem, Multi-imagem, DASA, Rio de Janeiro, Brazil
| | - Tania Netto
- Federal University of Rio de Janeiro, Department of Radiology, Rio de Janeiro, Brazil
| | - Denis Batista Pereira
- Federal University of Rio de Janeiro, Department of Radiology, Rio de Janeiro, Brazil
| | - Rochele Paz Fonseca
- Pontifical Catholic University of Rio Grande do Sul, Department of Psychology, Porto Alegre, Brazil
| | | |
Collapse
|
8
|
Chesnokova V, Pechnick RN, Wawrowsky K. Chronic peripheral inflammation, hippocampal neurogenesis, and behavior. Brain Behav Immun 2016; 58:1-8. [PMID: 26802985 PMCID: PMC4956598 DOI: 10.1016/j.bbi.2016.01.017] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/19/2016] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Adult hippocampal neurogenesis is involved in memory and learning, and disrupted neurogenesis is implicated in cognitive impairment and mood disorders, including anxiety and depression. Some long-term peripheral illnesses and metabolic disorders, as well as normal aging, create a state of chronic peripheral inflammation. These conditions are associated with behavioral disturbances linked to disrupted adult hippocampal neurogenesis, such as cognitive impairment, deficits in learning and memory, and depression and anxiety. Pro-inflammatory cytokines released in the periphery are involved in peripheral immune system-to-brain communication by activating resident microglia in the brain. Activated microglia reduce neurogenesis by suppressing neuronal stem cell proliferation, increasing apoptosis of neuronal progenitor cells, and decreasing survival of newly developing neurons and their integration into existing neuronal circuits. In this review, we summarize evolving evidence that the state of chronic peripheral inflammation reduces adult hippocampal neurogenesis, which, in turn, produces the behavioral disturbances observed in chronic inflammatory disorders. As there are no data available on neurogenesis in humans with chronic peripheral inflammatory disease, we focus on animal models and, in parallel, consider the evidence of cognitive disturbance and mood disorders in human patients.
Collapse
Affiliation(s)
- Vera Chesnokova
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States.
| | - Robert N Pechnick
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific and Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Kolja Wawrowsky
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
9
|
Lupus brain fog: a biologic perspective on cognitive impairment, depression, and fatigue in systemic lupus erythematosus. Immunol Res 2016; 63:26-37. [PMID: 26481913 DOI: 10.1007/s12026-015-8716-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cognitive disturbances, mood disorders and fatigue are common in SLE patients with substantial adverse effects on function and quality of life. Attribution of these clinical findings to immune-mediated disturbances associated with SLE remains difficult and has compromised research efforts in these areas. Improved understanding of the role of the immune system in neurologic processes essential for cognition including synaptic plasticity, long term potentiation and adult neurogenesis suggests multiple potential mechanisms for altered central nervous system function associated with a chronic inflammatory illness such as SLE. This review will focus on the biology of cognition and neuroinflammation in normal circumstances and potential biologic mechanisms for cognitive impairment, depression and fatigue attributable to SLE.
Collapse
|
10
|
TNF-like weak inducer of apoptosis promotes blood brain barrier disruption and increases neuronal cell death in MRL/lpr mice. J Autoimmun 2015; 60:40-50. [PMID: 25911200 DOI: 10.1016/j.jaut.2015.03.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 03/25/2015] [Accepted: 03/29/2015] [Indexed: 01/15/2023]
Abstract
Neuropsychiatric disease is one of the most common manifestations of human systemic lupus erythematosus, but the mechanisms remain poorly understood. In human brain microvascular endothelial cells in vitro, TNF-like weak inducer of apoptosis (TWEAK) decreases tight junction ZO-1 expression and increases the permeability of monolayer cell cultures. Furthermore, knockout (KO) of the TWEAK receptor, Fn14, in the MRL/lpr lupus mouse strain markedly attenuates neuropsychiatric disease, as demonstrated by significant reductions in depressive-like behavior and improved cognitive function. The purpose of the present study was to determine the mechanisms by which TWEAK signaling is instrumental in the pathogenesis of neuropsychiatric lupus (NPSLE). Evaluating brain sections of MRL/lpr Fn14WT and Fn14KO mice, we found that Fn14KO mice displayed significantly decreased cellular infiltrates in the choroid plexus. To evaluate the integrity of the blood brain barrier (BBB) in MRL/lpr mice, Western blot for fibronectin, qPCR for iNOS, and immunohistochemical staining for VCAM-1/ICAM-1 were performed. We found preserved BBB permeability in MRL/lpr Fn14KO mice, attributable to reduced brain expression of VCAM-1/ICAM-1 and iNOS. Additionally, administration of Fc-TWEAK intravenously directly increased the leakage of a tracer (dextran-FITC) into brain tissue. Furthermore, MRL/lpr Fn14KO mice displayed reduced antibody (IgG) and complement (C3, C6, and C4a) deposition in the brain. Finally, we found that MRL/lpr Fn14KO mice manifested reduced neuron degeneration and hippocampal gliosis. Our studies indicate that TWEAK/Fn14 interactions play an important role in the pathogenesis of NPSLE by increasing the accumulation of inflammatory cells in the choroid plexus, disrupting BBB integrity, and increasing neuronal damage, suggesting a novel target for therapy in this disease.
Collapse
|
11
|
Jeltsch-David H, Muller S. Neuropsychiatric systemic lupus erythematosus and cognitive dysfunction: the MRL-lpr mouse strain as a model. Autoimmun Rev 2014; 13:963-73. [PMID: 25183233 DOI: 10.1016/j.autrev.2014.08.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/10/2014] [Indexed: 12/19/2022]
Abstract
Mouse models of autoimmunity, such as (NZB×NZW)F1, MRL/MpJ-Fas(lpr) (MRL-lpr) and BXSB mice, spontaneously develop systemic lupus erythematosus (SLE)-like syndromes with heterogeneity and complexity that characterize human SLE. Despite their inherent limitations, such models have highly contributed to our current understanding of the pathogenesis of SLE as they provide powerful tools to approach the human disease at the genetic, cellular, molecular and environmental levels. They also allow novel treatment strategies to be evaluated in a complex integrated system, a favorable context knowing that very few murine models that adequately mimic human autoimmune diseases exist. As we move forward with more efficient medications to treat lupus patients, certain forms of the disease that requires to be better understood at the mechanistic level emerge. This is the case of neuropsychiatric (NP) events that affect 50-60% at SLE onset or within the first year after SLE diagnosis. Intense research performed at deciphering NP features in lupus mouse models has been undertaken. It is central to develop the first lead molecules aimed at specifically treating NPSLE. Here we discuss how mouse models, and most particularly MRL-lpr female mice, can be used for studying the pathogenesis of NPSLE in an animal setting, what are the NP symptoms that develop, and how they compare with human SLE, and, with a critical view, what are the neurobehavioral tests that are pertinent for evaluating the degree of altered functions and the progresses resulting from potentially active therapeutics.
Collapse
Affiliation(s)
- Hélène Jeltsch-David
- CNRS, Immunopathologie et chimie thérapeutique/Laboratory of excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, 67000 Strasbourg, France.
| | - Sylviane Muller
- CNRS, Immunopathologie et chimie thérapeutique/Laboratory of excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, 67000 Strasbourg, France.
| |
Collapse
|
12
|
Ji R, Tian S, Lu HJ, Lu Q, Zheng Y, Wang X, Ding J, Li Q, Lu Q. TAM receptors affect adult brain neurogenesis by negative regulation of microglial cell activation. THE JOURNAL OF IMMUNOLOGY 2013; 191:6165-77. [PMID: 24244024 DOI: 10.4049/jimmunol.1302229] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
TAM tyrosine kinases play multiple functional roles, including regulation of the target genes important in homeostatic regulation of cytokine receptors or TLR-mediated signal transduction pathways. In this study, we show that TAM receptors affect adult hippocampal neurogenesis and loss of TAM receptors impairs hippocampal neurogenesis, largely attributed to exaggerated inflammatory responses by microglia characterized by increased MAPK and NF-κB activation and elevated production of proinflammatory cytokines that are detrimental to neuron stem cell proliferation and neuronal differentiation. Injection of LPS causes even more severe inhibition of BrdU incorporation in the Tyro3(-/-)Axl(-/-)Mertk(-/-) triple-knockout (TKO) brains, consistent with the LPS-elicited enhanced expression of proinflammatory mediators, for example, IL-1β, IL-6, TNF-α, and inducible NO synthase, and this effect is antagonized by coinjection of the anti-inflammatory drug indomethacin in wild-type but not TKO brains. Conditioned medium from TKO microglia cultures inhibits neuron stem cell proliferation and neuronal differentiation. IL-6 knockout in Axl(-/-)Mertk(-/-) double-knockout mice overcomes the inflammatory inhibition of neurogenesis, suggesting that IL-6 is a major downstream neurotoxic mediator under homeostatic regulation by TAM receptors in microglia. Additionally, autonomous trophic function of the TAM receptors on the proliferating neuronal progenitors may also promote progenitor differentiation into immature neurons.
Collapse
Affiliation(s)
- Rui Ji
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Cagnoli P, Harris RE, Frechtling D, Berkis G, Gracley RH, Graft CC, Lowe SE, Chenevert TL, McCune WJ, Gebarski S, Sundgren PC. Reduced Insular Glutamine and N-acetylaspartate in systemic lupus erythematosus: a single-voxel (1)H-MR spectroscopy study. Acad Radiol 2013; 20:1286-96. [PMID: 24029061 DOI: 10.1016/j.acra.2013.07.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/23/2013] [Accepted: 07/24/2013] [Indexed: 01/01/2023]
Abstract
RATIONALE AND OBJECTIVES To investigate for differences in metabolic concentrations and ratios between patients with systemic lupus erythematosus (SLE) without (group SLE) and those with neurological symptoms (group NPSLE) compared to a healthy control (group HC) in three normal-appearing brain regions: the frontal white matter, right insula (RI), and occipital gray matter and whether changes in any of the metabolites or metabolic ratios are correlated to disease activity and other clinical parameters. MATERIALS AND METHODS Twenty patients with SLE (18 women and 2 men, age range 23.4-64.6 years, mean age 43.9 years), 23 NPSLE patients (23 women, age range 23.7-69.8 years, mean age 42.4 years), and 21 HC (19 women and 2 men, age range 21.0-65.7 years, mean age 43.4 years) were included. All subjects had conventional brain magnetic resonance imaging and (1)H single-voxel spectroscopy, clinical assessment, and laboratory testing. RESULTS NPSLE patients had significantly reduced N-acetylaspartate (NAA)/creatine compared to HC (P = .02) and SLE patients (P = .01) in the RI. Lower glutamine/creatine levels were also detected in RI in both patient groups and in frontal white matter in NPSLE patients compared to HC (P = .01, P = .02). NAA/Cr ratio in the RI was significantly negatively correlated with the Systemic Lupus Erythematosus Disease Activity Index (r = -0.41; P = .008), and patients with active SLE symptoms also had a trend toward lower NAA/creatine ratios (1.02 vs 1.12; P = .07). CONCLUSIONS The present data support previous findings of abnormal metabolic changes in normal-appearing regions in the brain of both SLE and NPSLE patients and raise the possibility that especially NAA, glutamine, and glutamate may be additional biomarkers for cerebral disease activity in SLE patients as these early metabolic changes occur in the brain of SLE patients before neurologic and imaging manifestations become apparent.
Collapse
|
14
|
Loheswaran G, Kapadia M, Gladman M, Pulapaka S, Xu L, Stanojcic M, Sakic B. Altered neuroendocrine status at the onset of CNS lupus-like disease. Brain Behav Immun 2013; 32:86-93. [PMID: 23466502 DOI: 10.1016/j.bbi.2013.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/18/2013] [Accepted: 02/20/2013] [Indexed: 01/07/2023] Open
Abstract
Neuropsychiatric (NP) manifestations and brain atrophy are common, etiologically unexplained complications of the systemic autoimmune disease lupus erythematosus (SLE). Similar to patients with NP SLE, behavioral deficits and neurodegeneration occur in aged, lupus-prone MRL/lpr mice. In order to gain a better understanding of the time course and nature of CNS involvement, we compare the neuro-immuno-endocrine profiles of two lupus-prone MRL/lpr stocks, which differ in disease onset and severity. Mice from stock 485 (characterized by early lupus-like manifestations) display blunted responsiveness to palatable solutions and impaired nocturnal activity as early as 7 weeks of age. They also have increased IgG in cerebrospinal fluid (CSF) before high serum autoantibody levels and splenomegaly are detected. Moreover, when compared to age-matched 6825 controls, 485 mice exhibit elevated serum corticosterone, enlarged left adrenal gland, and enhanced haematoxylin/eosin staining in the hypothalamic paraventricular nucleus. Swimming speed and novel object exploration become impaired only when more severe peripheral manifestations are documented in 17 week-old 485 mice. The obtained results suggest that performance deficits during the prodromal phase of NP SLE-like disease are associated with autoantibodies in CSF and asymmetric activation of the hypothalamus-pituitary-adrenal axis. Subsequent deterioration in behavioral performance evolves alongside systemic autoimmunity and inflammation. Although a leaky blood-CSF barrier is a possible explanation, one may hypothesize that, similar to neonatal lupus, maternal antibodies to brain antigens cross blood-placental barrier during embryogenesis and induce early endocrine and behavioral deficits in offspring.
Collapse
Affiliation(s)
- Genane Loheswaran
- Department of Psychiatry & Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada L8N 3Z5
| | | | | | | | | | | | | |
Collapse
|
15
|
Kapadia M, Stanojcic M, Earls AM, Pulapaka S, Lee J, Sakic B. Altered olfactory function in the MRL model of CNS lupus. Behav Brain Res 2012; 234:303-11. [PMID: 22796602 DOI: 10.1016/j.bbr.2012.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/02/2012] [Accepted: 07/06/2012] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder that damages several bodily systems, including the CNS. Brain atrophy and diverse neuropsychiatric manifestations are common and serious complications of SLE. Recently, it has been reported that many patients with CNS involvement also present with olfactory deficits of unknown etiology. Similar to CNS SLE, spontaneous development of lupus-like disease in MRL/lpr mice is accompanied by neurodegeneration in periventricular regions and a constellation of behavioral deficits dependent on olfaction. To test the possibility that olfactory dysfunction also occurs in autoimmune mice, we presently examine odor-guided behaviors using a battery of paradigms. Indeed, lupus-prone males spent less time exploring unfamiliar conspecifics and demonstrated age-dependant performance deficits when exposed to low concentrations of attractant and repellant odors. The emergence of olfactory changes was associated with a skewed distribution of DCX(+) cells in the proximal portion of the rostral migratory stream (RMS). The present results are consistent with the hypothesis that the onset of a SLE-like condition affects periventricular regions, including the RMS, as evidenced by disrupted migration of neuronal precursor cells toward the olfactory bulb. If so, ensuing hyposmia and/or olfactory memory deficit may contribute to altered performance in other behavioral tasks and reflect a prodrome of brain damage induced by chronic autoimmune disease.
Collapse
Affiliation(s)
- Minesh Kapadia
- The Brain-Body Institute, St. Joseph's Healthcare, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
16
|
Cagnoli PC, Sundgren PC, Kairys A, Graft CC, Clauw DJ, Gebarski S, McCune WJ, Schmidt-Wilcke T. Changes in regional brain morphology in neuropsychiatric systemic lupus erythematosus. J Rheumatol 2012; 39:959-67. [PMID: 22467931 DOI: 10.3899/jrheum.110833] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Neuropsychiatric lupus (NPSLE) is a severe and potentially life-threatening condition, reported to occur in 25%-70% of patients with systemic lupus erythematosus (SLE). Brain imaging, especially magnetic resonance imaging, is frequently used to diagnose or exclude overt cerebral pathologies such as edema, hemorrhage, and central thrombosis. More advanced imaging techniques have been applied to demonstrate subtle changes in regional cerebral blood flow and brain structure. We investigated changes in regional gray-matter (GM) volume in SLE patients without neurological manifestations and NPSLE patients at an acute stage of the disease. METHODS Using high-resolution structural images and voxel-based morphometry (VBM), we investigated regional GM volume in 20 NPSLE patients (within 2 weeks of the acute manifestation), 18 SLE patients without neurologic and/or psychiatric manifestations, and 18 healthy controls. RESULTS VBM analyses revealed several regions of GM atrophy in various parts of the brain in NPSLE and SLE patients. GM atrophy was seen in both groups in the temporal and parietal lobes and was most pronounced in the posterior thalamus bilaterally. Both groups showed an increase in regional GM volume in the posterior parahippocampal gyrus. CONCLUSION Our data suggest that changes in regional brain morphology are present in acute NPSLE, but also in SLE (as compared to controls), which might be indicative of a subclinical neurodegenerative process. Further research is needed to investigate whether specific neuropsychiatric symptoms are related to these changes.
Collapse
Affiliation(s)
- Patricia C Cagnoli
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, Michigan 48105, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Sakić B. The MRL model: an invaluable tool in studies of autoimmunity-brain interactions. Methods Mol Biol 2012; 934:277-99. [PMID: 22933151 DOI: 10.1007/978-1-62703-071-7_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The link between systemic autoimmunity, brain pathology, and aberrant behavior is still largely unexplored field of biomedical science. Accumulating evidence points to causal relationships between immune factors, neurodegeneration, and neuropsychiatric manifestations. By documenting autoimmunity-associated neuronal degeneration and cytotoxicity of the cerebrospinal fluid from disease-affected subjects, the murine MRL model had shown high validity in revealing principal pathogenic circuits. In addition, unlike any other autoimmune strain, MRL mice produce antibodies commonly found in patients suffering from lupus and other autoimmune disorders. This review highlights importance of the MRL model as an indispensible preparation in understanding the links between immune system and brain function.
Collapse
Affiliation(s)
- Boris Sakić
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
18
|
Kapadia M, Sakic B. Autoimmune and inflammatory mechanisms of CNS damage. Prog Neurobiol 2011; 95:301-33. [DOI: 10.1016/j.pneurobio.2011.08.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/13/2022]
|
19
|
Telerman A, Lapter S, Sharabi A, Zinger H, Mozes E. Induction of hippocampal neurogenesis by a tolerogenic peptide that ameliorates lupus manifestations. J Neuroimmunol 2011; 232:151-7. [DOI: 10.1016/j.jneuroim.2010.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 10/28/2010] [Accepted: 11/01/2010] [Indexed: 11/27/2022]
|
20
|
Gulinello M, Putterman C. The MRL/lpr mouse strain as a model for neuropsychiatric systemic lupus erythematosus. J Biomed Biotechnol 2011; 2011:207504. [PMID: 21331367 PMCID: PMC3038428 DOI: 10.1155/2011/207504] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 12/28/2010] [Indexed: 12/20/2022] Open
Abstract
To date, CNS disease and neuropsychiatric symptoms of systemic lupus erythematosus (NP-SLE) have been understudied compared to end-organ failure and peripheral pathology. In this review, we focus on a specific mouse model of lupus and the ways in which this model reflects some of the most common manifestations and potential mechanisms of human NP-SLE. The mouse MRL lymphoproliferation strain (a.k.a. MRL/lpr) spontaneously develops the hallmark serological markers and peripheral pathologies typifying lupus in addition to displaying the cognitive and affective dysfunction characteristic of NP-SLE, which may be among the earliest symptoms of lupus. We suggest that although NP-SLE may share common mechanisms with peripheral organ pathology in lupus, especially in the latter stages of the disease, the immunologically privileged nature of the CNS indicates that early manifestations of particularly mood disorders maybe derived from some unique mechanisms. These include altered cytokine profiles that can activate astrocytes, microglia, and alter neuronal function before dysregulation of the blood-brain barrier and development of clinical autoantibody titres.
Collapse
Affiliation(s)
- Maria Gulinello
- Behavioral Core Facility, Department of Neuroscience, Albert Einstein College of Medicine, 1410 Pelham Pkwy S Kennedy 925, Bronx, NY 10461, USA.
| | | |
Collapse
|
21
|
Huehnchen P, Prozorovski T, Klaissle P, Lesemann A, Ingwersen J, Wolf SA, Kupsch A, Aktas O, Steiner B. Modulation of adult hippocampal neurogenesis during myelin-directed autoimmune neuroinflammation. Glia 2010; 59:132-42. [DOI: 10.1002/glia.21082] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 08/31/2010] [Indexed: 01/04/2023]
|
22
|
Loheswaran G, Stanojcic M, Xu L, Sakic B. Autoimmunity as a principal pathogenic factor in the refined model of neuropsychiatric lupus. ACTA ACUST UNITED AC 2010. [DOI: 10.1111/j.1759-1961.2010.00014.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Stanojcic M, Loheswaran G, Xu L, Hoffman SA, Sakic B. Intrathecal antibodies and brain damage in autoimmune MRL mice. Brain Behav Immun 2010; 24:289-97. [PMID: 19853033 DOI: 10.1016/j.bbi.2009.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 10/12/2009] [Accepted: 10/18/2009] [Indexed: 12/20/2022] Open
Abstract
Neuropsychiatric (NP) manifestations and brain pathology are poorly understood and potentially fatal concomitants of systemic lupus erythematosus (SLE). For many years, autoantibodies to brain tissue (i.e., brain-reactive antibodies, BRA) were proposed as a key factor in pathogenesis of CNS manifestations. Recent evidence suggests that intrathecal BRA, rather than serum autoantibodies, are a better predictor of disturbed brain morphology and function. We presently test this hypothesis by examining the relationship among BRA in cerebrospinal fluid (CSF), behavioral deficits, and brain pathology in a well-established animal model of CNS lupus. We showed earlier that significant diversity in disease manifestations within genetically homogenous MRL-lpr mice allows for constructive and informative correlational analysis. Therefore, levels of CSF antibodies were presently correlated with behavioral, neuropathological and immune measures in a cohort of diseased MRL-lpr males (N=40). ELISA, Western Blotting, standardized behavioral battery, digital planimetry, HE staining, and immunohistochemistry were employed in overall data collection. The IgG antibodies from CSF were binding to different regions of brain parenchyma, with dentate gyrus, amygdale, and subventricular zones showing enhanced immunoreactivity. High levels of CSF antibodies correlated with increased immobility in the forced-swim test and density of HE(+) cells in the paraventricular nucleus. Peripheral measures of autoimmunity were associated with other deficits in behavior and neuropathology. This correlation pattern suggests that etiology of brain damage in lupus-prone mice is multifactorial. Intrathecal BRA may be important in altering motivated responses and activity of major neuroendocrine axes at the onset of SLE-like disease.
Collapse
Affiliation(s)
- Mile Stanojcic
- Department of Psychiatry and Behavioural Neurosciences, The Brain-Body Institute, McMaster University, Hamilton, Ontario, Canada L8N 4A6
| | | | | | | | | |
Collapse
|