1
|
Hart DA. Lithium Ions as Modulators of Complex Biological Processes: The Conundrum of Multiple Targets, Responsiveness and Non-Responsiveness, and the Potential to Prevent or Correct Dysregulation of Systems during Aging and in Disease. Biomolecules 2024; 14:905. [PMID: 39199293 PMCID: PMC11352090 DOI: 10.3390/biom14080905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024] Open
Abstract
Lithium is one of the lightest elements on Earth and it has been in the environment since the formation of the galaxy. While a common element, it has not been found to be an essential element in biological processes, ranging from single cell organisms to Homo sapiens. Instead, at an early stage of evolution, organisms committed to a range of elements such as sodium, potassium, calcium, magnesium, zinc, and iron to serve essential functions. Such ions serve critical functions in ion channels, as co-factors in enzymes, as a cofactor in oxygen transport, in DNA replication, as a storage molecule in bone and liver, and in a variety of other roles in biological processes. While seemingly excluded from a major essential role in such processes, lithium ions appear to be able to modulate a variety of biological processes and "correct" deviation from normal activity, as a deficiency of lithium can have biological consequences. Lithium salts are found in low levels in many foods and water supplies, but the effectiveness of Li salts to affect biological systems came to recent prominence with the work of Cade, who reported that administrating Li salts calmed guinea pigs and was subsequently effective at relatively high doses to "normalize" a subset of patients with bipolar disorders. Because of its ability to modulate many biological pathways and processes (e.g., cyclic AMP, GSK-3beta, inositol metabolism, NaK ATPases, neuro processes and centers, immune-related events, respectively) both in vitro and in vivo and during development and adult life, Li salts have become both a useful tool to better understand the molecular regulation of such processes and to also provide insights into altered biological processes in vivo during aging and in disease states. While the range of targets for lithium action supports its possible role as a modulator of biological dysregulation, it presents a conundrum for researchers attempting to elucidate its specific primary target in different tissues in vivo. This review will discuss aspects of the state of knowledge regarding some of the systems that can be influenced, focusing on those involving neural and autoimmunity as examples, some of the mechanisms involved, examples of how Li salts can be used to study model systems, as well as suggesting areas where the use of Li salts could lead to additional insights into both disease mechanisms and natural processes at the molecular and cell levels. In addition, caveats regarding lithium doses used, the strengths and weaknesses of rodent models, the background genetics of the strain of mice or rats employed, and the sex of the animals or the cells used, are discussed. Low-dose lithium may have excellent potential, alone or in combination with other interventions to prevent or alleviate aging-associated conditions and disease progression.
Collapse
Affiliation(s)
- David A Hart
- Department of Surgery, Faculty of Kinesiology, McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
2
|
Wang K, Hou X, Lu H, Han N, Xie M, Xi A, Xu Z. Ectopic CD4 + T cells in choroid plexus mediate neuropsychiatric lupus symptoms in mice via interferon-γ induced microglia activation. J Autoimmun 2024; 145:103199. [PMID: 38452512 DOI: 10.1016/j.jaut.2024.103199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
Neuropsychiatric systemic lupus erythematosus (NPSLE) is a disabling and potentially life-threatening complication of SLE. This study aims to investigate whether ectopic CD4+ T cells in the choroid plexus mediate NPSLE in mice. Intracerebroventricular (ICV) injection of anti-CD4 antibody effectively depleted CP-resident CD4+ T cells and alleviated NPSLE-like symptoms in MRL/lpr mice. Following ICV injection, the majority of isolated lupus CD4+ T cells from donor MRL/lpr mice predominantly stayed in the CP for at least 28 days in recipient C57BL/6 mice, while nearly all isolated CD4+ T cells from MRL/MpJ mice disappeared within 7 days. ICV injection of lupus CD4+ T cells resulted in NPSLE-like symptoms, including impaired behavioral performances, increased microglial activation, and abnormal microstructure changes. Flow cytometry analysis revealed that the majority of isolated lupus CD4+ T cells were positive for IFN-γ. Neutralizing intracerebral IFN-γ alleviated NPSLE-like symptoms in MRL/lpr mice. Moreover, ICV injection of anti-IFN-γ antibody or microglial depletion by PLX3397 benefited most NPSLE-like symptoms in lupus CD4+ T-treated mice, while ICV injection of IFN-γ mimicked most NPSLE-like symptoms. In conclusion, CP-resident lupus CD4+ T cells contribute to NPSLE-like symptoms in mice via Interferon-γ induced microglia activation. Depleting CP-resident lupus CD4+ T cells, interferon-γ, or activated microglia may be potential therapeutic targets for NPSLE.
Collapse
Affiliation(s)
- Keer Wang
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Xiaoxiao Hou
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Haimei Lu
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Ning Han
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Meijuan Xie
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China; Ji'an Hospital of Traditional Chinese Medicine, Jiangxi, China
| | - Anran Xi
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Zhenghao Xu
- Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science & Wenzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
3
|
Reynolds J, Huang M, Li Y, Meineck M, Moeckel T, Weinmann-Menke J, Mohan C, Schwarting A, Putterman C. Constitutive knockout of interleukin-6 ameliorates memory deficits and entorhinal astrocytosis in the MRL/lpr mouse model of neuropsychiatric lupus. J Neuroinflammation 2024; 21:89. [PMID: 38600510 PMCID: PMC11007930 DOI: 10.1186/s12974-024-03085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 03/31/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Neuropsychiatric lupus (NPSLE) describes the cognitive, memory, and affective emotional burdens faced by many lupus patients. While NPSLE's pathogenesis has not been fully elucidated, clinical imaging studies and cerebrospinal fluid (CSF) findings, namely elevated interleukin-6 (IL-6) levels, point to ongoing neuroinflammation in affected patients. Not only linked to systemic autoimmunity, IL-6 can also activate neurotoxic glial cells the brain. A prior pre-clinical study demonstrated that IL-6 can acutely induce a loss of sucrose preference; the present study sought to assess the necessity of chronic IL-6 exposure in the NPSLE-like disease of MRL/lpr lupus mice. METHODS We quantified 1308 proteins in individual serum or pooled CSF samples from MRL/lpr and control MRL/mpj mice using protein microarrays. Serum IL-6 levels were plotted against characteristic NPSLE neurobehavioral deficits. Next, IL-6 knockout MRL/lpr (IL-6 KO; n = 15) and IL-6 wildtype MRL/lpr mice (IL-6 WT; n = 15) underwent behavioral testing, focusing on murine correlates of learning and memory deficits, depression, and anxiety. Using qPCR, we quantified the expression of inflammatory genes in the cortex and hippocampus of MRL/lpr IL-6 KO and WT mice. Immunofluorescent staining was performed to quantify numbers of microglia (Iba1 +) and astrocytes (GFAP +) in multiple cortical regions, the hippocampus, and the amygdala. RESULTS MRL/lpr CSF analyses revealed increases in IL-17, MCP-1, TNF-α, and IL-6 (a priori p-value < 0.1). Serum levels of IL-6 correlated with learning and memory performance (R2 = 0.58; p = 0.03), but not motivated behavior, in MRL/lpr mice. Compared to MRL/lpr IL-6 WT, IL-6 KO mice exhibited improved novelty preference on object placement (45.4% vs 60.2%, p < 0.0001) and object recognition (48.9% vs 67.9%, p = 0.002) but equivalent performance in tests for anxiety-like disease and depression-like behavior. IL-6 KO mice displayed decreased cortical expression of aif1 (microglia; p = 0.049) and gfap (astrocytes; p = 0.044). Correspondingly, IL-6 KO mice exhibited decreased density of GFAP + cells compared to IL-6 WT in the entorhinal cortex (89 vs 148 cells/mm2, p = 0.037), an area vital to memory. CONCLUSIONS The inflammatory composition of MRL/lpr CSF resembles that of human NPSLE patients. Increased in the CNS, IL-6 is necessary to the development of learning and memory deficits in the MRL/lpr model of NPSLE. Furthermore, the stimulation of entorhinal astrocytosis appears to be a key mechanism by which IL-6 promotes these behavioral deficits.
Collapse
Affiliation(s)
- Joshua Reynolds
- Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY, USA
| | - Michelle Huang
- Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY, USA
| | - Yaxi Li
- University of Houston, Houston, TX, USA
| | - Myriam Meineck
- University Medical Center of the Johannes Gutenberg University, University of Mainz, Mainz, Germany
| | - Tamara Moeckel
- University Medical Center of the Johannes Gutenberg University, University of Mainz, Mainz, Germany
| | - Julia Weinmann-Menke
- University Medical Center of the Johannes Gutenberg University, University of Mainz, Mainz, Germany
| | | | - Andreas Schwarting
- University Medical Center of the Johannes Gutenberg University, University of Mainz, Mainz, Germany
| | - Chaim Putterman
- Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY, USA.
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel.
| |
Collapse
|
4
|
Zhou Y, Chen L, Zheng X, Fang Q, Qian Y, Xu T, Liang J, Zhang H, Han X, Sun L. Microglia orchestrate synaptic and neuronal stripping: Implication in neuropsychiatric lupus. J Cell Mol Med 2024; 28:e18190. [PMID: 38494844 PMCID: PMC10945089 DOI: 10.1111/jcmm.18190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 01/26/2024] [Accepted: 02/09/2024] [Indexed: 03/19/2024] Open
Abstract
Systemic lupus erythematosus (SLE), a multifactorial autoimmune disease, can affect the brain and cause neuropsychiatric dysfunction, also named neuropsychiatric lupus (NPSLE). Microglial activation is observed in NPSLE patients. However, the mechanisms regulating microglia-mediated neurotoxicity in NPSLE remain elusive. Here, we showed that M1-like proinflammatory cytokine levels were increased in the cerebrospinal fluid (CSF) of SLE patients, especially those with neuropsychiatric symptoms. We also demonstrated that MRL/lpr lupus mice developed anxiety-like behaviours and cognitive deficits in the early and active phases of lupus, respectively. An increase in microglial number was associated with upregulation of proinflammatory cytokines in the MRL/lpr mouse brain. RNA sequencing revealed that genes associated with phagocytosis and M1 polarization were upregulated in microglia from lupus mice. Functionally, activated microglia induced synaptic stripping in vivo and promoted neuronal death in vitro. Finally, tofacitinib ameliorated neuropsychiatric disorders in MRL/lpr mice, as evidenced by reductions in microglial number and synaptic/neuronal loss and alleviation of behavioural abnormalities. Thus, our results indicated that classically activated (M1) microglia play a crucial role in NPSLE pathogenesis. Minocycline and tofacitinib were found to alleviate NPSLE by inhibiting micrglial activation, providing a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yishan Zhou
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical SchoolNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjingChina
| | - Liang Chen
- Department of GynecologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiulan Zheng
- School of PharmacyMacau University of Science and TechnologyMacauChina
| | - Qijun Fang
- Department of Traditional Chinese Medicine, Nanjing Drum Tower HospitalNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Yunzhi Qian
- Department of Nutrition, Gillings School of Global Public HealthUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Tianshu Xu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower HospitalNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical SchoolNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjingChina
| | - Huajun Zhang
- Department of Traditional Chinese Medicine, Nanjing Drum Tower HospitalNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Xiaojuan Han
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical SchoolNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjingChina
- Department of Traditional Chinese Medicine, Nanjing Drum Tower HospitalNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical SchoolNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjingChina
- School of PharmacyMacau University of Science and TechnologyMacauChina
| |
Collapse
|
5
|
Liu A, Li Z, Zeng J, Peng Y, Wang S, Bi X, Zhao Z, Zhou S, Zhao AZ, Mu Y, Li F. ω-3 polyunsaturated fatty acid alleviates systemic lupus erythematosus by suppressing autoimmunity in a murine model. Int Immunopharmacol 2024; 126:111299. [PMID: 38043268 DOI: 10.1016/j.intimp.2023.111299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune inflammatory disease that damages multiple organs by the production of autoantibodies. Numerous research studies have demonstrated the anti-inflammatory effects of ω-3 polyunsaturated fatty acids (PUFAs). A diet rich in ω-3 PUFAs reduces chronic inflammatory and autoimmune conditions. Herein, we investigated the protective effect of ω-3 PUFAs against autoimmune injury in SLE. In a TMPD-induced mouse model of SLE, supplementation with eicosapentaenoic acid (EPA)-rich (97%) fish oil was found to alleviate systemic autoimmune phenotypes such as ascites, lipogranulomas and serum dsDNA levels. In addition, EPA also significantly improved renal manifestations, reducing proteinuria, glomerulonephritis, and immune complex deposition. Mechanistically, ω-3 PUFAs were shown to modulate the differentiation of B lymphocyte subsets of primary splenic lymphocytes in the spontaneous murine lupus model MRL/MpJ-Faslpr in vitro, specifically that both EPA and DHA suppressed the number of total B cells, B1B2 cells and plasma cells. Concurrently, they were also found to promote the secretion of the anti-inflammatory cytokine IL10, mainly produced by Breg and Treg cells. Thus, nutritional supplementation with ω-3 PUFAs can regulate B cell's differentiation and anti-inflammatory function and strongly prevent autoimmune responses and lupus nephritis. The diets balance between ω-6 and ω-3 PUFAs intake may represent a promising treatment strategy to prevent or delay the onset of SLE.
Collapse
Affiliation(s)
- Aolu Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhuang Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Jingwen Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yuerong Peng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shuai Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xinyun Bi
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhenggang Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Sujin Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Allan Zijian Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yunping Mu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Fanghong Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
6
|
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
|
7
|
Tomalla V, Schmeisser MJ, Weinmann-Menke J. Mouse models, antibodies, and neuroimaging: Current knowledge and future perspectives in neuropsychiatric systemic lupus erythematosus (NPSLE). Front Psychiatry 2023; 14:1078607. [PMID: 36970286 PMCID: PMC10031066 DOI: 10.3389/fpsyt.2023.1078607] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
As a chronic autoimmune disease systemic lupus erythematosus (SLE) can also affect the central and the peripheral nervous system causing symptoms which are summed up as neuropsychiatric systemic lupus erythematosus (NPSLE). These symptoms are heterogenous including cognitive impairment, seizures, and fatigue, leading to morbidity or even mortality. At present, little is known about the pathophysiological processes involved in NPSLE. This review focuses on the current knowledge of the pathogenesis of NPSLE gained from the investigation of animal models, autoantibodies, and neuroimaging techniques. The antibodies investigated the most are anti-ribosomal P protein antibodies (Anti-rib P) and anti-N-Methyl-D-Aspartic Acid Receptor 2 antibodies (Anti-NR2), which represent a subpopulation of anti-dsDNA autoantibodies. Experimental data demonstrates that Anti-rib P and Anti-NR2 cause different neurological pathologies when applied intravenously (i.v.), intrathecally or intracerebrally in mice. Moreover, the investigation of lupus-prone mice, such as the MRL/MpJ-Faslpr/lpr strain (MRL/lpr) and the New Zealand black/New Zealand white mice (NZB × NZW F1) showed that circulating systemic antibodies cause different neuropsychiatric symptoms compared to intrathecally produced antibodies. Furthermore, neuroimaging techniques including magnetic resonance imaging (MRI) and positron emission tomography (PET) are commonly used tools to investigate structural and functional abnormalities in NPSLE patients. Current research suggests that the pathogenesis of NPSLE is heterogenous, complex and not yet fully understood. However, it demonstrates that further investigation is needed to develop individual therapy in NPSLE.
Collapse
Affiliation(s)
- Vanessa Tomalla
- Department of Internal Medicine, Division of Nephrology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael J. Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Julia Weinmann-Menke
- Department of Internal Medicine, Division of Nephrology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- *Correspondence: Julia Weinmann-Menke,
| |
Collapse
|
8
|
Karino K, Kono M, Takeyama S, Kudo Y, Kanda M, Abe N, Aso K, Fujieda Y, Kato M, Oku K, Amengual O, Atsumi T. Inhibitor of NF-κB Kinase Subunit ε Contributes to Neuropsychiatric Manifestations in Lupus-Prone Mice Through Microglial Activation. Arthritis Rheumatol 2023; 75:411-423. [PMID: 36098515 DOI: 10.1002/art.42352] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 07/25/2022] [Accepted: 09/08/2022] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by multiorgan dysfunction. Neuropsychiatric SLE (NPSLE) occurs in 30-40% of lupus patients and is the most severe presentation of SLE, frequently resulting in limitation of daily life. Recent studies have shown that microglia, tissue-resident macrophages in the central nervous system, are involved in the pathogenesis of NPSLE. This study was undertaken to explore new therapeutic targets for NPSLE focusing on microglia. METHODS RNA sequencing of microglia in MRL/lpr, lupus-prone mice, as well as that of microglia cultured in vitro with cytokines were performed. A candidate gene, which could be a therapeutic target for NPSLE, was identified, and its role in microglial activation and phagocytosis was investigated using specific inhibitors and small interfering RNA. The effect of intracerebroventricular administration of the inhibitor on the behavioral abnormalities of MRL/lpr was also evaluated. RESULTS Transcriptome analysis revealed the up-regulation of Ikbke, which encodes the inhibitor of NF-κB kinase subunit ɛ (IKBKε) in both microglia from MRL/lpr mice and cytokine-stimulated microglia in vitro. Intracerebroventricular administration of an IKBKε inhibitor ameliorated cognitive function and suppressed microglial activation in MRL/lpr mice. Mechanistically, IKBKε inhibition reduced glycolysis, which dampened microglial activation and phagocytosis. CONCLUSION These findings suggest that IKBKε plays a vital role in the pathogenesis of NPSLE via microglial activation, and it could serve as a therapeutic target for NPSLE.
Collapse
Affiliation(s)
- Kohei Karino
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Michihito Kono
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shuhei Takeyama
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Kudo
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masatoshi Kanda
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University, Sapporo, Japan
| | - Nobuya Abe
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kuniyuki Aso
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuichiro Fujieda
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaru Kato
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kenji Oku
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan, and Department of Rheumatology and Infectious Diseases, School of Medicine, Kitasato University, Sagamihara, Japan
| | - Olga Amengual
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
9
|
Fan Y, Liu X, Wu J, Ni J, Liang J, Hou Y, Dou H. Small molecule compound K-7174 attenuates neuropsychiatric manifestations in lupus-prone mice. Brain Res 2023; 1801:148203. [PMID: 36521514 DOI: 10.1016/j.brainres.2022.148203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
The neuropsychiatric manifestations of systemic lupus erythematosus (NPSLE) present significant morbidity and mortality due to frequent non-response or adverse effects of the current clinical drugs. The disruption of the blood-brain barrier (BBB) contributes to inflammatory NPSLE disease progression. K-7174, a highly piperazine-derived compound, inhibits leukocyte adhesion and inflammatory factor expression. The present study aimed to comprehensively assess the treatment effect of neurobehavioral deficits in MRL/lpr mice, a validated neuropsychiatric lupus model. The intraperitoneal injection of K-7174 alleviated lupus-like symptoms and improved cognitive dysfunction in MRL/lpr mice. Also, it significantly attenuated neuronal degeneration and decreased serum albumin deposition in the hippocampus. Furthermore, K-7174 acted directly on the brain microvascular endothelial bEnd.3 cells and reduced the BBB permeability, manifested by inhibiting the activation of brain microvascular endothelial cells and increasing the expression of tight junctions (TJs). Notably, in vitro experiments showed that K-7174 alleviates the decreased ZO1 and Occludin expression in bEnd.3 cells caused by lactate increase, improving cell permeability via the MCT4/NKAP/CREB signaling pathway. These findings suggested that K-7174 mediates the attenuation of NPSLE in MRL/lpr mice, indicating a promising therapeutic strategy for NPSLE.
Collapse
Affiliation(s)
- Yu Fan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Xuan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jinjin Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jiali Ni
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China.
| |
Collapse
|
10
|
Yun Y, Wang X, Xu J, Jin C, Chen J, Wang X, Wang J, Qin L, Yang P. Pristane induced lupus mice as a model for neuropsychiatric lupus (NPSLE). BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:3. [PMID: 36765366 PMCID: PMC9921421 DOI: 10.1186/s12993-023-00205-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/13/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND The pristane-induced lupus (PIL) model is a useful tool for studying environmental-related systemic lupus erythematosus (SLE). However, neuropsychiatric manifestations in this model have not been investigated in detail. Because neuropsychiatric lupus (NPSLE) is an important complication of SLE, we investigated the neuropsychiatric symptoms in the PIL mouse model to evaluate its suitability for NPSLE studies. RESULTS PIL mice showed olfactory dysfunction accompanied by an anxiety- and depression-like phenotype at month 2 or 4 after pristane injection. The levels of cytokines (IL-1β, IFN-α, IFN-β, IL-10, IFN-γ, IL-6, TNF-α and IL-17A) and chemokines (CCL2 and CXCL10) in the brain and blood-brain barrier (BBB) permeability increased significantly from week 2 or month 1, and persisted throughout the observed course of the disease. Notably, IgG deposition in the choroid plexus and lateral ventricle wall were observed at month 1 and both astrocytes and microglia were activated. Persistent activation of astrocytes was detected throughout the observed course of the disease, while microglial activation diminished dramatically at month 4. Lipofuscin deposition, a sign of neuronal damage, was detected in cortical and hippocampal neurons from month 4 to 8. CONCLUSION PIL mice exhibit a series of characteristic behavioral deficits and pathological changes in the brain, and therefore might be suitable for investigating disease pathogenesis and for evaluating potential therapeutic targets for environmental-related NPSLE.
Collapse
Affiliation(s)
- Yang Yun
- grid.412467.20000 0004 1806 3501Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuejiao Wang
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Jingyi Xu
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chenye Jin
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jingyu Chen
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Xueru Wang
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Jianing Wang
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Ling Qin
- Department of Physiology, China Medical University, Shenyang, China.
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Microstructural Changes in the Corpus Callosum in Systemic Lupus Erythematous. Cells 2023; 12:cells12030355. [PMID: 36766697 PMCID: PMC9913100 DOI: 10.3390/cells12030355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/02/2022] [Accepted: 11/21/2022] [Indexed: 01/20/2023] Open
Abstract
Central nervous system (CNS) involvement in childhood-onset systemic lupus erythematosus (cSLE) occurs in more than 50% of patients. Structural magnetic resonance imaging (MRI) has identified global cerebral atrophy, as well as the involvement of the corpus callosum and hippocampus, which is associated with cognitive impairment. In this cross-sectional study we included 71 cSLE (mean age 24.7 years (SD 4.6) patients and a disease duration of 11.8 years (SD 4.8) and two control groups: (1) 49 adult-onset SLE (aSLE) patients (mean age of 33.2 (SD 3.7) with a similar disease duration and (2) 58 healthy control patients (mean age of 29.9 years (DP 4.1)) of a similar age. All of the individuals were evaluated on the day of the MRI scan (Phillips 3T scanner). We reviewed medical charts to obtain the clinical and immunological features and treatment history of the SLE patients. Segmentation of the corpus callosum was performed through an automated segmentation method. Patients with cSLE had a similar mid-sagittal area of the corpus callosum in comparison to the aSLE patients. When compared to the control groups, cSLE and aSLE had a significant reduction in the mid-sagittal area in the posterior region of the corpus callosum. We observed significantly lower FA values and significantly higher MD, RD, and AD values in the total area of the corpus callosum and in the parcels B, C, D, and E in cSLE patients when compared to the aSLE patients. Low complement, the presence of anticardiolipin antibodies, and cognitive impairment were associated with microstructural changes. In conclusion, we observed greater microstructural changes in the corpus callosum in adults with cSLE when compared to those with aSLE. Longitudinal studies are necessary to follow these changes, however they may explain the worse cognitive function and disability observed in adults with cSLE when compared to aSLE.
Collapse
|
12
|
Wang Y, Ren Y, Hong T, Lu D, Zhang F, Cao Y, Wang X. Lipidomics Changes in a Murine Model of Neuropsychiatric Lupus. J Inflamm Res 2022; 15:6569-6580. [PMID: 36506783 PMCID: PMC9733566 DOI: 10.2147/jir.s391595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/18/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose Neuropsychiatric lupus (NPSLE) is one of the important manifestations of systemic lupus erythematosus. Previous studies mainly focused on the disruption of the blood-brain barrier and the production of brain-reactive autoantibodies, However, there is no comprehensive lipidomic analysis in NPSLE. Therefore, this research evaluated the lipidomic analysis in the hippocampus and liver of NPSLE mice with mood disorders, to explore the influence of the liver-brain axis on this disease. Methods MRL/lpr mice and MRL/mpj mice were respectively used as NPSLE and control groups. Behavioral tests and systemic disease characteristics of mice were assessed at the age of 18 weeks. Ultra-Performance Liquid Chromatography-Tandem Mass Spectrometry (UPLC-MS/MS) was used for lipid metabolite determination. Multivariate statistical analysis was used to identify lipid metabolites that were differentially expressed in two groups. Results Our results showed that 355 and 405 lipid metabolites were differentially expressed between the NPSLE and control groups in the hippocampus and liver. According to the pathway enrichment analysis, several pathways were affected, and the glycerophospholipid metabolism pathway was most relevant to the mouse's depressive behavior. Conclusion Based on UPLC-MS/MS, the results provide evidence for how the liver-brain axis affects NPSLE and improve the understanding of NPSLE pathogenesis.
Collapse
Affiliation(s)
- Yihan Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Yating Ren
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Tao Hong
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Dingqi Lu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Fan Zhang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Xinchang Wang
- Department of Rheumatology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China,Correspondence: Xinchang Wang, Department of Rheumatology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China, Tel +86 0571-85288249, Email
| |
Collapse
|
13
|
Moore E, Huang MW, Reynolds CA, Macian F, Putterman C. Choroid Plexus-Infiltrating T Cells as Drivers of Murine Neuropsychiatric Lupus. Arthritis Rheumatol 2022; 74:1796-1807. [PMID: 35637551 PMCID: PMC9825865 DOI: 10.1002/art.42252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVE T cells are critical in the pathogenesis of systemic lupus erythematosus (SLE) in that they secrete inflammatory cytokines, help autoantibody production, and form autoreactive memory T cells. Although the contribution of T cells to several forms of organ-mediated damage in SLE has been previously demonstrated, the role of T cells in neuropsychiatric SLE (NPSLE), which involves diffuse central nervous system manifestations and is observed in 20-40% of SLE patients, is not known. Therefore, we conducted this study to evaluate how behavioral deficits are altered after depletion or transfer of T cells, to directly assess the role of T cells in NPSLE. METHODS MRL/lpr mice, an NPSLE mouse model, were either systemically depleted of CD4+ T cells or intracerebroventricularly injected with choroid plexus (CP)-infiltrating T cells and subsequently evaluated for alterations in neuropsychiatric manifestations. Our study end points included evaluation of systemic disease and assessment of central nervous system changes. RESULTS Systemic depletion of CD4+ T cells ameliorated systemic disease and cognitive deficits. Intracerebroventricular injection of CP-infiltrating T cells exacerbated depressive-like behavior and worsened cognition in recipient mice compared with mice who received injection of splenic lupus T cells or phosphate buffered saline. Moreover, we observed enhanced activation in CP-infiltrating T cells when cocultured with brain lysate-pulsed dendritic cells in comparison to the activation levels observed in cocultures with splenic T cells. CONCLUSION T cells, and more specifically CP-infiltrating antigen-specific T cells, contributed to the pathogenesis of NPSLE in mice, indicating that, in the development of more targeted treatments for NPSLE, modulation of T cells may represent a potential therapeutic strategy.
Collapse
Affiliation(s)
- Erica Moore
- Department of Microbiology and Immunology, Division of RheumatologyAlbert Einstein College of MedicineNew York
| | - Michelle W. Huang
- Department of Microbiology and Immunology, Division of RheumatologyAlbert Einstein College of MedicineNew York
| | - Cara A. Reynolds
- Department of PathologyAlbert Einstein College of MedicineNew York
| | - Fernando Macian
- Department of PathologyAlbert Einstein College of MedicineNew York
| | - Chaim Putterman
- Azrieli Faculty of Medicine of Bar‐Ilan University, Safed, Israel, Galilee Research Institute, Nahariya, Israel, and Department of Microbiology and Immunology, Division of RheumatologyAlbert Einstein College of MedicineNew York
| |
Collapse
|
14
|
Liu Y, Tao X, Tao J. Strategies of Targeting Inflammasome in the Treatment of Systemic Lupus Erythematosus. Front Immunol 2022; 13:894847. [PMID: 35664004 PMCID: PMC9157639 DOI: 10.3389/fimmu.2022.894847] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple organ dysfunction resulting from the production of multiple autoantibodies and adaptive immune system abnormalities involving T and B lymphocytes. In recent years, inflammasomes have been recognized as an important component of innate immunity and have attracted increasing attention because of their pathogenic role in SLE. In short, inflammasomes regulate the abnormal differentiation of immune cells, modulate pathogenic autoantibodies, and participate in organ damage. However, due to the clinical heterogeneity of SLE, the pathogenic roles of inflammasomes are variable, and thus, the efficacy of inflammasome-targeting therapies is uncertain. To provide a foundation for the development of such therapeutic strategies, in this paper, we review the role of different inflammasomes in the pathogenesis of SLE and their correlation with clinical phenotypes and propose some corresponding treatment strategies.
Collapse
Affiliation(s)
- Yaling Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinyu Tao
- Department of Clinical Medicine "5 + 3" Integration, The First Clinical College, Anhui Medical University, Hefei, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
15
|
Qiao X, Lu L, Zhou K, Tan L, Liu X, Ni J, Hou Y, Liang J, Dou H. The correlation between proteoglycan 2 and neuropsychiatric systemic lupus erythematosus. Clin Immunol 2022; 239:109042. [PMID: 35568106 DOI: 10.1016/j.clim.2022.109042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023]
Abstract
The proposed pathogenesis of neuropsychiatric systemic lupus erythematosus (NPSLE) mainly includes ischemia and neuroinflammation mechanisms. Protein encoded by Proteoglycan 2 (PRG2) mRNA is involved in the immune process related to eosinophils, also being found in the placenta and peripheral blood of pregnant women. We evaluated the correlation between PRG2 and NPSLE for the first time and found that PRG2 protein is overexpressed in the serum of patients with NPSLE and correlated with the SLE disease activity index (SLEDAI) subset scores of psychosis. Moreover, we investigated the correlation between hippocampal PRG2 level and hippocampally dependent learning and memory ability in MRL/lpr mice, and discovered that the number of PRG2+GFAP+ astrocytes in the cortex and hypothalamus and the number of PRG2+IBA-1+ microglia in the hippocampus and cortex significantly increased in the MRL/lpr mice. These data provided a reference for the follow-up exploration of the role of PRG2 in SLE or other diseases.
Collapse
Affiliation(s)
- Xiaoyue Qiao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Li Lu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Kangxing Zhou
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Liping Tan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Xuan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Jiali Ni
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
16
|
Xin Y, Zhang B, Zhao J, Liu Q, Yin H, Lu Q. Animal models of systemic lupus erythematosus and their applications in drug discovery. Expert Opin Drug Discov 2022; 17:489-500. [PMID: 35287523 DOI: 10.1080/17460441.2022.2050691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease with substantial phenotypic heterogeneity. Currently, our understanding of the pathogenesis is still limited, and as a result, specific and efficacious therapies are lacking. Various mouse models have been established to serve as powerful tools that will promote a better understanding of the disease and the ability to test novel drugs before clinical application. AREAS COVERED The authors review the existing mouse models of SLE in terms of pathogenesis and manifestations, as well as their applications in drug discovery and development. The areas of focus include promising novel therapeutics that could benefit patients in the future and the contribution of mouse models used in preclinical studies. EXPERT OPINION Given the diversity of SLE mouse models with different characteristics, researchers must select a suitable model based on the mechanism involved. The use of multiple models is needed for drug testing studies to evaluate drug efficacy on different genetic backgrounds and other mechanisms to provide a reference for clinical trials.
Collapse
Affiliation(s)
- Yue Xin
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Bo Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Junpeng Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Qianmei Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Haoyuan Yin
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| |
Collapse
|
17
|
Zhu J, Inomata T, Shih KC, Okumura Y, Fujio K, Huang T, Nagino K, Akasaki Y, Fujimoto K, Yanagawa A, Miura M, Midorikawa-Inomata A, Hirosawa K, Kuwahara M, Shokirova H, Eguchi A, Morooka Y, Chen F, Murakami A. Application of Animal Models in Interpreting Dry Eye Disease. Front Med (Lausanne) 2022; 9:830592. [PMID: 35178415 PMCID: PMC8844459 DOI: 10.3389/fmed.2022.830592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/11/2022] [Indexed: 11/23/2022] Open
Abstract
Different pathophysiologic mechanisms are involved in the initiation, development, and outcome of dry eye disease (DED). Animal models have proven valuable and efficient in establishing ocular surface microenvironments that mimic humans, thus enabling better understanding of the pathogenesis. Several dry eye animal models, including lacrimal secretion insufficiency, evaporation, neuronal dysfunction, and environmental stress models, are related to different etiological factors. Other models may be categorized as having a multifactorial DED. In addition, there are variations in the methodological classification, including surgical lacrimal gland removal, drug-induced models, irradiation impairment, autoimmune antibody-induced models, and transgenic animals. The aforementioned models may manifest varying degrees of severity or specific pathophysiological mechanisms that contribute to the complexity of DED. This review aimed to summarize various dry eye animal models and evaluate their respective characteristics to improve our understanding of the underlying mechanism and identify therapeutic prospects for clinical purposes.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kendrick Co Shih
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuichi Okumura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kenta Fujio
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tianxiang Huang
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Nagino
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasutsugu Akasaki
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keiichi Fujimoto
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ai Yanagawa
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Maria Miura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akie Midorikawa-Inomata
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kunihiko Hirosawa
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mizu Kuwahara
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hurramhon Shokirova
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsuko Eguchi
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuki Morooka
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fang Chen
- Department of Ophthalmology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Neuronal NR4A1 deficiency drives complement-coordinated synaptic stripping by microglia in a mouse model of lupus. Signal Transduct Target Ther 2022; 7:50. [PMID: 35177587 PMCID: PMC8854434 DOI: 10.1038/s41392-021-00867-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/18/2021] [Accepted: 12/19/2021] [Indexed: 12/15/2022] Open
Abstract
Neuropsychiatric lupus (NPSLE) is a frequent manifestation of systemic lupus erythematosus (SLE) that occurs in 40–90% of SLE patients; however, the underlying mechanisms remain elusive, causing a severe lack of therapeutic targets for this condition. Here, we show that complement-coordinated elimination of synapses participated in NPSLE in MRL/lpr mice, a lupus-prone murine model. We demonstrated that lupus mice developed increased anxiety-like behaviors and persistent phagocytic microglial reactivation before overt peripheral lupus pathology. In the lupus brain, C1q was increased and localized at synaptic terminals, causing the apposition of phagocytic microglia and ensuing synaptic engulfment. We further determined that neuronal Nr4a1 signaling was essential for attracting C1q synaptic deposition and subsequent microglia-mediated synaptic elimination. Minocycline-mediated deactivation of microglia, antibody blockade of C1q, or neuronal restoration of Nr4a1 protected lupus mice from synapse loss and NP manifestations. Our findings revealed an active role of neurons in coordinating microglia-mediated synaptic loss and highlighted neuronal Nr4a1 and C1q as critical components amenable to therapeutic intervention in NPSLE.
Collapse
|
19
|
Vanarsa K, Sasidharan P, Duran V, Gokaraju S, Nidhi M, Louis Sam Titus ASC, Soomro S, Stock AD, Der E, Putterman C, Greenberg B, Mok CC, Hanly JG, Mohan C. Aptamer-based screen of Neuropsychiatric Lupus cerebrospinal fluid reveals potential biomarkers that overlap with the choroid plexus transcriptome. Arthritis Rheumatol 2022; 74:1223-1234. [PMID: 35099126 DOI: 10.1002/art.42080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 12/28/2021] [Accepted: 01/27/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES As no gold-standard diagnostic test exists for neuropsychiatric systemic lupus erythematosus (NPSLE), we executed a broad screen of NPSLE cerebrospinal fluid (CSF) using an aptamer-based platform. METHODS CSF were obtained from NPSLE patients and subjected to proteomic assay using the aptamer-based screen. Potential biomarkers were identified and validated in independent NPSLE cohorts in comparison with other neurological diseases. RESULTS 40 proteins out of 1129 screened were elevated in NPSLE CSF. By ELISA validation, CSF Angiostatin, α2-Macroglobulin, DAN, Fibronectin, HCC-1, IgM, Lipocalin 2, M-CSF and SERPING1 were significantly elevated in a predominantly Caucasian NPSLE cohort (n=24), compared to patients with other neurological diseases (n=54), with CSF IgM (AUC=0.95) and M-CSF (AUC=0.91) being the most discriminatory. In a second, Hong Kong NPSLE cohort, CSF IgM (AUC=0.78) and Lipocalin-2 (AUC=0.85) were the most discriminatory. Several CSF proteins exhibited high diagnostic specificity for NPSLE in both cohorts. Elevated CSF C3 was associated with acute confusional state. Eleven molecules elevated in NPSLE CSF exhibited concordant elevation in the choroid plexus, suggesting shared origins. CONCLUSIONS CSF Lipocalin-2, M-CSF, IgM and complement C3 emerge as promising CSF biomarkers of NPSLE with diagnostic potential.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Valeria Duran
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Sirisha Gokaraju
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Malavika Nidhi
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Sanam Soomro
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Evan Der
- Albert Einstein College of Medicine, Bronx, NY
| | | | | | | | - John G Hanly
- Division of Rheumatology, Queen Elizabeth II Health Sciences Center and Dalhousie University Halifax, Nova Scotia, Canada
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX
| |
Collapse
|
20
|
Killian M, Colaone F, Haumont P, Nicco C, Cerles O, Chouzenoux S, Cathébras P, Rochereau N, Chanut B, Thomas M, Laroche N, Forest F, Grouard-Vogel G, Batteux F, Paul S. Therapeutic Potential of Anti-Interferon α Vaccination on SjS-Related Features in the MRL/lpr Autoimmune Mouse Model. Front Immunol 2021; 12:666134. [PMID: 34867938 PMCID: PMC8635808 DOI: 10.3389/fimmu.2021.666134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 10/21/2021] [Indexed: 11/25/2022] Open
Abstract
Sjögren’s syndrome (SjS) is a frequent systemic autoimmune disease responsible for a major decrease in patients’ quality of life, potentially leading to life-threatening conditions while facing an unmet therapeutic need. Hence, we assessed the immunogenicity, efficacy, and tolerance of IFN-Kinoid (IFN-K), an anti-IFNα vaccination strategy, in a well-known mouse model of systemic autoimmunity with SjS-like features: MRL/MpJ-Faslpr/lpr (MRL/lpr) mice. Two cohorts (with ISA51 or SWE01 as adjuvants) of 26 female MRL/lpr were divided in parallel groups, “controls” (not treated, PBS and Keyhole Limpet Hemocyanin [KLH] groups) or “IFN-K” and followed up for 122 days. Eight-week-old mice received intra-muscular injections (days 0, 7, 28, 56 and 84) of PBS, KLH or IFN-K, emulsified in the appropriate adjuvant, and blood samples were serially collected. At sacrifice, surviving mice were euthanized and their organs were harvested for histopathological analysis (focus score in salivary/lacrimal glands) and IFN signature evaluation. SjS-like features were monitored. IFN-K induced a disease-modifying polyclonal anti-IFNα antibody response in all treated mice with high IFNα neutralization capacities, type 1 IFN signature’s reduction and disease features’ (ocular and oral sicca syndrome, neuropathy, focus score, glandular production of BAFF) improvement, as reflected by the decrease in Murine Sjögren’s Syndrome Disease Activity Index (MuSSDAI) modelled on EULAR Sjögren’s Syndrome Disease Activity Index (ESSDAI). No adverse effects were observed. We herein report on the strong efficacy of an innovative anti-IFNα vaccination strategy in a mouse model of SjS, paving the way for further clinical development (a phase IIb trial has just been completed in systemic lupus erythematosus with promising results).
Collapse
Affiliation(s)
- Martin Killian
- Centre International de Recherche en Infectiologie (CIRI), Team Groupe Immunité des Muqueuses et Agents Pathogènes (GIMAP), Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, Centre National de la Recherche Scientifique (CNRS), UMR530, Saint-Etienne, France
- Internal Medicine Department, Saint-Etienne University Hospital, Saint-Etienne, France
| | | | | | - Carole Nicco
- Team Stress Oxydant, Prolifération Cellulaire et Inflammation, Institut National de la Santé Et de la Recherche Médicale (INSERM) U1016 Institut Cochin, Paris, France
| | - Olivier Cerles
- Team Stress Oxydant, Prolifération Cellulaire et Inflammation, Institut National de la Santé Et de la Recherche Médicale (INSERM) U1016 Institut Cochin, Paris, France
| | - Sandrine Chouzenoux
- Team Stress Oxydant, Prolifération Cellulaire et Inflammation, Institut National de la Santé Et de la Recherche Médicale (INSERM) U1016 Institut Cochin, Paris, France
| | - Pascal Cathébras
- Centre International de Recherche en Infectiologie (CIRI), Team Groupe Immunité des Muqueuses et Agents Pathogènes (GIMAP), Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, Centre National de la Recherche Scientifique (CNRS), UMR530, Saint-Etienne, France
- Internal Medicine Department, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Nicolas Rochereau
- Centre International de Recherche en Infectiologie (CIRI), Team Groupe Immunité des Muqueuses et Agents Pathogènes (GIMAP), Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, Centre National de la Recherche Scientifique (CNRS), UMR530, Saint-Etienne, France
| | - Blandine Chanut
- Centre International de Recherche en Infectiologie (CIRI), Team Groupe Immunité des Muqueuses et Agents Pathogènes (GIMAP), Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, Centre National de la Recherche Scientifique (CNRS), UMR530, Saint-Etienne, France
| | - Mireille Thomas
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1059-Sainbiose, Université de Lyon, Saint Priest en Jarez, France
| | - Norbert Laroche
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1059-Sainbiose, Université de Lyon, Saint Priest en Jarez, France
| | - Fabien Forest
- Department of Pathology, Saint-Etienne University Hospital, Saint-Etienne, France
| | | | - Frédéric Batteux
- Team Stress Oxydant, Prolifération Cellulaire et Inflammation, Institut National de la Santé Et de la Recherche Médicale (INSERM) U1016 Institut Cochin, Paris, France
| | - Stéphane Paul
- Centre International de Recherche en Infectiologie (CIRI), Team Groupe Immunité des Muqueuses et Agents Pathogènes (GIMAP), Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, Centre National de la Recherche Scientifique (CNRS), UMR530, Saint-Etienne, France
- *Correspondence: Stéphane Paul,
| |
Collapse
|
21
|
Lu L, Wang H, Liu X, Tan L, Qiao X, Ni J, Sun Y, Liang J, Hou Y, Dou H. Pyruvate kinase isoform M2 impairs cognition in systemic lupus erythematosus by promoting microglial synaptic pruning via the β-catenin signaling pathway. J Neuroinflammation 2021; 18:229. [PMID: 34645459 PMCID: PMC8513209 DOI: 10.1186/s12974-021-02279-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 08/31/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Neuropsychiatric systemic lupus erythematosus (NPSLE) is a severe complication, which involves pathological damage to the brain and cognitive function. However, its exact mechanism of action still remains unclear. In this study, we explored the role of microglia in the cognitive dysfunction of NPSLE mice. We also analyzed and compared the metabolites in the hippocampal tissues of the lupus model and control mice. METHODS MRL/MpJ-Faslpr (MRL/lpr) female mice were used as the NPSLE mouse model. Metabolomics was used to assess hippocampal glycolysis levels. Glucose, lactic acid, IL-6, and IL-1β of the hippocampus were detected by ELISA. Based on the glycolysis pathway, we found that pyruvate kinase isoform M2 (PKM2) in the hippocampus was significantly increased. Thus, the expression of PKM2 was detected by qRT-PCR and Western blotting, and the localization of PKM2 in microglia (IBA-1+) or neurons (NeuN+) was assessed by immunofluorescence staining. Flow cytometry was used to detect the number and phenotype of microglia; the changes in microglial phagocytosis and the β-catenin signaling pathway were detected in BV2 cells overexpressing PKM2. For in vivo experiments, MRL/lpr mice were treated with AAV9-shPKM2. After 2 months, Morris water maze and conditional fear tests were applied to investigate the cognitive ability of mice; H&E and immunofluorescence staining were used to evaluate brain damage; flow cytometry was used to detect the phenotype and function of microglia; neuronal synapse damage was monitored by qRT-PCR, Western blotting, and immunofluorescence staining. RESULTS Glycolysis was elevated in the hippocampus of MRL/lpr lupus mice, accompanied by increased glucose consumption and lactate production. Furthermore, the activation of PKM2 in hippocampal microglia was observed in lupus mice. Cell experiments showed that PKM2 facilitated microglial activation and over-activated microglial phagocytosis via the β-catenin signaling pathway. In vivo, AAV9-shPKM2-treated mice showed decreased microglial activation and reduced neuronal synapses loss by blocking the β-catenin signaling pathway. Furthermore, the cognitive impairment and brain damage of MRL/lpr mice were significantly relieved after microglial PKM2 inhibition. CONCLUSION These data indicate that microglial PKM2 have potential to become a novel therapeutic target for treating lupus encephalopathy.
Collapse
Affiliation(s)
- Li Lu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China
| | - Hailin Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China
| | - Xuan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China
| | - Liping Tan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China
| | - Xiaoyue Qiao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China
| | - Jiali Ni
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China
| | - Yang Sun
- The State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences, School of Life Sciences, Nanjing University, Nanjing, 210023, China.
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China.
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China. .,Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, People's Republic of China. .,Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China.
| |
Collapse
|
22
|
Qiao X, Wang H, Lu L, Chen J, Cheng Q, Guo M, Hou Y, Dou H. Hippocampal microglia CD40 mediates NPSLE cognitive dysfunction in mice. J Neuroimmunol 2021; 357:577620. [PMID: 34062352 DOI: 10.1016/j.jneuroim.2021.577620] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
Neuropsychiatric systemic lupus erythematosus (NPSLE) is the most serious and complicated clinical manifestation of lupus erythematosus. Cognitive dysfunction is the most common symptom of NPSLE. A variety of potential mechanisms or mediators related to the pathogenesis of NPSLE cognitive dysfunction have been proposed. However, the involvement of microglia CD40 has not been reported yet. This study aimed to investigate whether hippocampal microglia CD40 of MRL/MpJ-Faslpr (MRL/lpr) mice was involved in NPSLE cognitive dysfunction. This study found, using quantitative polymerase chain reaction, western blotting and immunohistochemistry, that hippocampal CD40 was aberrantly overexpressed in the MRL/lpr lupus mice. It also determined using flow cytometry and immunofluorescence that the aberrantly overexpressed CD40 was mainly derived from hippocampal microglia. The adeno-associated virus was used to inhibit microglia CD40 expression, and the brain damage and cognitive dysfunction of MRL/lpr mice improved. Also, imiquimod (IMQ)-induced lupus mice had the same NPSLE cognitive dysfunction, brain damage, and overexpressed hippocampal microglia CD40 as MRL/lpr mice. Therefore, IMQ-induced lupus mouse was proposed as one of the mouse models for studying NPSLE cognitive dysfunction for the first time in this study. The findings indicated that hippocampal microglia CD40 was involved in the development of NPSLE cognitive dysfunction, thus providing a novel research direction for the study of the pathogenesis of NPSLE.
Collapse
Affiliation(s)
- Xiaoyue Qiao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Hailin Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Li Lu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Jinglei Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Qinpei Cheng
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Meng Guo
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
23
|
Zarfeshani A, Carroll KR, Volpe BT, Diamond B. Cognitive Impairment in SLE: Mechanisms and Therapeutic Approaches. Curr Rheumatol Rep 2021; 23:25. [PMID: 33782842 PMCID: PMC11207197 DOI: 10.1007/s11926-021-00992-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
A wide range of patients with systemic lupus erythematosus (SLE) suffer from cognitive dysfunction (CD) which severely impacts their quality of life. However, CD remains underdiagnosed and poorly understood. Here, we discuss current findings in patients and in animal models. Strong evidence suggests that CD pathogenesis involves known mechanisms of tissue injury in SLE. These mechanisms recruit brain resident cells, in particular microglia, into the pathological process. While systemic immune activation is critical to central nervous system injury, the current focus of therapy is the microglial cell and not the systemic immune perturbation. Further studies are critical to examine additional potential therapeutic targets and more specific treatments based on the cause and progress of the disease.
Collapse
Affiliation(s)
- Aida Zarfeshani
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kaitlin R Carroll
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Bruce T Volpe
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
24
|
Adam K, Iuga A, Tocheva AS, Mor A. A novel mouse model for checkpoint inhibitor-induced adverse events. PLoS One 2021; 16:e0246168. [PMID: 33571254 PMCID: PMC7877613 DOI: 10.1371/journal.pone.0246168] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint inhibitors have demonstrated significant efficacy in the treatment of a variety of cancers, however their therapeutic potential is limited by abstruse immune related adverse events. Currently, no robust animal model exists of checkpoint inhibitor-induced adverse events. Establishing such a model will improve our mechanistic understanding of this process, which in turn will inform design of improved therapies. We developed a mouse model to determine inflammatory toxicities in response to dual checkpoint blockade in the presence of syngeneic tumors. Mice from susceptible genetic backgrounds received intraperitoneal injections of anti-mouse PD-1 and CTLA-4 antibodies. The mice were monitored for weight loss and histologic evidence of inflammation. Blood was collected for basic metabolic panels and titers of anti-nuclear antibodies. In parallel, mice were also treated with prednisolone, which is commonly used to treat immune related adverse events among cancer patients. Among all the genetic backgrounds, B6/lpr mice treated with anti-CTLA-4 and anti-PD-1 antibodies developed more substantial hepatitis, pancreatitis, colitis, and pneumonitis characterized by organ infiltration of immune cells. Mice that developed tissue infiltration demonstrated high serum levels of glucose and high titers of anti-nuclear antibodies. Finally, while administration of prednisolone prevented the development of the inflammatory adverse events, it also abrogated the protective anti-tumor effect of the checkout inhibitors. Genetic background and treatment modalities jointly modified the inflammatory adverse events in tumor bearing mice, suggesting a complex mechanism for checkpoint inhibitor-related inflammation. Future studies will assess additional genetic susceptibility factors and will examine possible contributions from the administration of other anti-inflammatory drugs.
Collapse
Affiliation(s)
- Kieran Adam
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Alina Iuga
- Department of Pathology, Columbia University Medical Center, New York, New York, United States of America
| | - Anna S. Tocheva
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|
25
|
Karnopp TE, Chapacais GF, Freitas EC, Monticielo OA. Lupus animal models and neuropsychiatric implications. Clin Rheumatol 2020; 40:2535-2545. [PMID: 33155159 DOI: 10.1007/s10067-020-05493-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) that involves neurological complications is known as neuropsychiatric systemic lupus erythematosus (NPSLE). Research in humans is difficult due to the disease's great heterogeneity. Animal models are a resource for new discoveries. In this review, we examine experimental models of lupus that present neuropsychiatric manifestations. Spontaneous animal models such as NZB/W F1 and MRL/lpr are commonly used in NPSLE research; these models present few SLE symptoms compared to induced animal models, such as pristane-induced lupus (PIL). The PIL model is known to present eight of the main clinical and laboratory manifestations of SLE described by the American College of Rheumatology. Many cytokines associated with NPSLE are expressed in the PIL model, such as IL-6, TNF-α, and IFN. However, to date, NPSLE manifestations have been poorly studied in the PIL model. In this review article, we discuss whether the PIL model can mimic neuropsychiatric manifestations of SLE. Key Points • PIL model have a strong interferon signature. • Animals with PIL express learning and memory deficit.
Collapse
Affiliation(s)
- Thaís Evelyn Karnopp
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil. .,Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Gustavo Flores Chapacais
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil.,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Curso de Graduação em Biomedicina, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduarda Correa Freitas
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil.,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Odirlei André Monticielo
- Laboratório de Doenças Autoimunes, Divisão de Reumatologia, Centro de Pesquisas Experimentais, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, sala 12109, Porto Alegre, 90035-003, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
26
|
Moore E, Huang MW, Jain S, Chalmers SA, Macian F, Putterman C. The T Cell Receptor Repertoire in Neuropsychiatric Systemic Lupus Erythematosus. Front Immunol 2020; 11:1476. [PMID: 32765512 PMCID: PMC7379895 DOI: 10.3389/fimmu.2020.01476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/05/2020] [Indexed: 11/13/2022] Open
Abstract
Objective: In systemic lupus erythematosus (SLE), widespread T cell infiltration into target organs contributes to inflammation and organ damage. Autoreactive T cells become aberrantly activated in this disease due to dysfunctional T cell receptor signaling that lowers the activation threshold. Characterizing the T cell repertoire can provide further insight into the specific homing and proliferation of these T cells into lupus target organs. In the spontaneous lupus model, MRL/lpr, the TCR repertoire has not been fully elucidated, especially for T cells infiltrating the brain. Our aim was to investigate and compare the TCR repertoire between MRL/lpr mice and its congenic controls, MRL/MpJ, and within MRL/lpr tissues. Methods: Spleen, salivary gland, and brain choroid plexus were isolated from female MRL/lpr mice and MRL/MpJ mice. The TCRβ CDR3 region was analyzed by multiplex PCRs and sequencing. Results: Significant differences were seen not only between the MRL/lpr and MRL/MpJ spleens, but also between MRL/lpr tissues. The TCR repertoire in MRL/lpr choroid plexus tissues had significantly increased clonality and sequence homology compared to MRL/lpr spleen and salivary gland. The consensus sequence, CASSQDWGGYEQYFF, was identified in the MRL/lpr choroid plexus repertoire. Conclusions: The TCR repertoire in lupus prone mice is not uniform between target organs, and suggests that T cells are specifically recruited into the choroid plexus of MRL/lpr mice. Further studies are needed to determine the antigen specificities for these infiltrating T cells in target organs of lupus mice, and their possible contribution to the pathogenesis of neuropsychiatric disease and other lupus manifestations.
Collapse
Affiliation(s)
- Erica Moore
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Michelle W Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Shweta Jain
- Early Discovery and Fundamental Research, Hansoh Bio, Rockville, MD, United States
| | - Samantha A Chalmers
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, United States
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, United States.,Bar-Ilan University Azrieli Faculty of Medicine, Ramat Gan, Israel.,Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
27
|
El Khoury L, Zarfeshani A, Diamond B. Using the Mouse to Model Human Diseases: Cognitive Impairment in Systemic Lupus Erythematosus. J Rheumatol 2020; 47:1145-1149. [PMID: 32295852 PMCID: PMC11207199 DOI: 10.3899/jrheum.200410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this 2020 Dunlop-Dottridge Lecture, the authors discuss cognitive impairment (CI), one of the most prevalent neuropsychiatric syndromes in systemic lupus erythematosus (SLE). Patients often report CI as the most bothersome disease-related manifestation, with a great effect on their quality of life. Nevertheless, studies focusing on CI remain scarce and no effective targeted therapy has been identified. We herein present murine models of CI in SLE with insights into the pathogenesis of this condition as well as the role of the renin angiotensin system in microglial activation. We will discuss the role of neuroimaging as a useful objective assessment tool, describing our experience in previous and ongoing clinical trials of CI in patients with SLE.
Collapse
Affiliation(s)
- Lara El Khoury
- From the Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- L. El Khoury, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; A. Zarfeshani, PhD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; B. Diamond, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health
| | - Aida Zarfeshani
- From the Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
- L. El Khoury, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; A. Zarfeshani, PhD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; B. Diamond, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health
| | - Betty Diamond
- From the Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.
- L. El Khoury, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; A. Zarfeshani, PhD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health; B. Diamond, MD, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health.
| |
Collapse
|
28
|
Blossom SJ, Melnyk SB, Simmen FA. Complex epigenetic patterns in cerebellum generated after developmental exposure to trichloroethylene and/or high fat diet in autoimmune-prone mice. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2020; 22:583-594. [PMID: 31894794 PMCID: PMC7350281 DOI: 10.1039/c9em00514e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Trichloroethylene (TCE) is an environmental contaminant associated with immune-mediated inflammatory disorders and neurotoxicity. Based on known negative effects of developmental overnutrition on neurodevelopment, we hypothesized that developmental exposure to high fat diet (HFD) consisting of 40% kcal fat would enhance neurotoxicity of low-level (6 μg per kg per day) TCE exposure in offspring over either stressor alone. Male offspring were evaluated at ∼6 weeks of age after exposure beginning 4 weeks preconception in the dams until weaning. TCE, whether used as a single exposure or together with HFD, appeared to be more robust than HFD alone in altering one-carbon metabolites involved in glutathione redox homeostasis and methylation capacity. In contrast, opposing effects of expression of key enzymes related to DNA methylation related to HFD and TCE exposure were observed. The mice generated unique patterns of anti-brain antibodies detected by western blotting attributable to both TCE and HFD. Taken together, developmental exposure to TCE and/or HFD appear to act in complex ways to alter brain biomarkers in offspring.
Collapse
Affiliation(s)
- Sarah J Blossom
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Research Institute, Little Rock, AR 72202, USA.
| | | | | |
Collapse
|
29
|
Nomura A, Noto D, Murayama G, Chiba A, Miyake S. Unique primed status of microglia under the systemic autoimmune condition of lupus-prone mice. Arthritis Res Ther 2019; 21:303. [PMID: 31888754 PMCID: PMC6936062 DOI: 10.1186/s13075-019-2067-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/25/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of various autoantibodies. This disease causes disabling neuropsychiatric symptoms even in the absence of apparent inflammation in the central nervous system (CNS), but the mechanisms involved remain unknown. Innate immune-mediated inflammation has attracted attention as a pathogenic mechanism in neuropsychiatric diseases. METHODS We investigated the CNS of lupus-prone mice focusing on innate immunity. Three strains of lupus-prone mice, FcγRIIB-/-Yaa, an F1 hybrid of NZB and NZW (NZB/NZW) mice, and MRL/Faslpr (MRL/lpr) mice were used to analyze CNS immunopathology. RESULTS Flow cytometry analysis demonstrated the numbers of brain CD45+ cells were increased compared with controls in lupus-prone mice. Upregulation of MHC class I and PDCA1 was observed in microglia and CD11b+ myeloid cells of lupus-prone mice, indicating they were activated in response to interferons (IFN). Microglial gene expression analysis of FcγRIIB-/-Yaa mice revealed the upregulation of IFN-responsive genes and inflammation-related genes including Axl, Clec7a, and Itgax, which were previously reported in neurodegenerative conditions and primed conditions. Upregulated chemokine gene expressions including Ccl5 and Cxcl10 were concurrent with increased numbers of T cells and monocytes, especially Ly6Clo monocytes in the CNS. Upregulation of Axl, Clec7a, Itgax, Ccl5, and Cxcl10 was also observed in NZB/NZW mice, indicating common lupus pathology. The primed status of microglia in FcγRIIB-/-Yaa mice was also demonstrated by morphological changes such as enlarged cell bodies with hypertrophic processes, and hyperreactivity to lipopolysaccharide. Immunohistochemistry of FcγRIIB-/-Yaa mice indicated reactive responses of astrocytes and vascular endothelium. Behavioral studies of FcγRIIB-/-Yaa mice revealed depressive-like behavior and heat hyperalgesia in the forced swim test and the tail-flick test, respectively. CONCLUSIONS Our data indicated that microglia in lupus exhibit a unique primed phenotype characterized by the upregulated expressions of neurodegeneration-related genes and IFN-responsive genes. Interaction with peripheral cells and brain resident cells was presumed to orchestrate neuroinflammation. Targeting innate immune cells, such as microglia and monocytes, may be a promising therapeutic approach for neuropsychiatric SLE.
Collapse
Affiliation(s)
- Atsushi Nomura
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Daisuke Noto
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Goh Murayama
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Asako Chiba
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
30
|
Analysis of brain metabolites by gas chromatography-mass spectrometry reveals the risk-benefit concerns of prednisone in MRL/lpr lupus mice. Inflammopharmacology 2019; 28:425-435. [PMID: 31786803 DOI: 10.1007/s10787-019-00668-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/14/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Neuropsychiatric systemic lupus erythematosus (NPSLE) is a common cause of disability in systemic lupus erythematosus (SLE). This study aims to investigate the metabolic changes in the hypothalamus and frontal cortex in lupus-prone MRL/lpr mice. METHODS Metabolic changes were analyzed using gas chromatography-mass spectrometry (GC-MS). RESULTS According to the principal component analysis (PCA), the metabolic profiles were different between the frontal cortex and hypothalamus, but they were comparable between MRL/lpr and MRL/MpJ mice (16 weeks of age). By OPLS-DA, eight cortical and six hypothalamic differential metabolites were identified in MRL/lpr as compared to MRL/MpJ mice. Among these differential metabolites, we found a decrease of N-acetyl-L-aspartate (NAA, a potential marker of neuronal integrity), an increase of pyruvate and a decrease of glutamate in the frontal cortex but not in the hypothalamus. Prednisone treatment (3 mg/kg from 8 weeks of age) relieved the decrease of NAA but further increased the accumulation of pyruvate in the frontal cortex, additionally affected eight enriched pathways in the hypothalamus, and led to significant imbalances between the excitation and inhibition in both the frontal cortex and hypothalamus. CONCLUSION These results suggest that the frontal cortex may be more preferentially affected than the hypothalamus in SLE. Prednisone disrupted rather than relieved metabolic abnormalities in the brain, especially in the hypothalamus, indicating that the risk-benefit balance of prednisone for SLE or NPSLE remains to be further evaluated.
Collapse
|
31
|
Killian M, Batteux F, Paul S. The MRL/lpr Mouse Model: An Important Animal Model for Systemic Sjögren Syndrome and Polyautoimmunity. J Rheumatol 2019; 47:157. [PMID: 31523043 DOI: 10.3899/jrheum.190820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Martin Killian
- EA 3064, Groupe Immunité des Muqueuses et Agents Pathogènes, Université de Lyon, and Internal Medicine Department, Saint-Etienne University Hospital, Saint-Etienne;
| | - Frédéric Batteux
- Team Stress oxydant, prolifération cellulaire et inflammation, INSERM U1016 Institut Cochin, Paris
| | - Stéphane Paul
- EA 3064, Groupe Immunité des Muqueuses et Agents Pathogènes, Université de Lyon, Saint-Etienne, France
| |
Collapse
|
32
|
Cigliano L, Spagnuolo MS, Boscaino F, Ferrandino I, Monaco A, Capriello T, Cocca E, Iannotta L, Treppiccione L, Luongo D, Maurano F, Rossi M, Bergamo P. Dietary Supplementation with Fish Oil or Conjugated Linoleic Acid Relieves Depression Markers in Mice by Modulation of the Nrf2 Pathway. Mol Nutr Food Res 2019; 63:e1900243. [DOI: 10.1002/mnfr.201900243] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/25/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Luisa Cigliano
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Maria Stefania Spagnuolo
- Institute for Animal Production System in Mediterranean EnvironmentNational Research Council (ISPAAM, CNR) Naples Italy
| | - Floriana Boscaino
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Ida Ferrandino
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Antonio Monaco
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Teresa Capriello
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Ennio Cocca
- Institute of Biosciences and Bio‐ResourcesNational Research Council (CNR‐IBBR) Naples Italy
| | - Lucia Iannotta
- Department of BiologyUniversity “Federico II” of Naples Naples Italy
| | - Lucia Treppiccione
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Diomira Luongo
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Francesco Maurano
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Mauro Rossi
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| | - Paolo Bergamo
- Institute of Food SciencesNational Research Council (CNR‐ISA) Avellino Italy
| |
Collapse
|
33
|
Kello N, Anderson E, Diamond B. Cognitive Dysfunction in Systemic Lupus Erythematosus: A Case for Initiating Trials. Arthritis Rheumatol 2019; 71:1413-1425. [PMID: 31102496 PMCID: PMC6716992 DOI: 10.1002/art.40933] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/14/2019] [Indexed: 12/25/2022]
Abstract
Cognitive dysfunction (CD) is an insidious and underdiagnosed manifestation of systemic lupus erythematosus (SLE) that has a considerable impact on quality of life, which can be devastating. Given the inconsistencies in the modes of assessment and the difficulties in attribution to SLE, the reported prevalence of CD ranges from 5% to 80%. Although clinical studies of SLE-related CD have been hampered by heterogeneous subject populations and a lack of sensitive and standardized cognitive tests or other validated objective biomarkers for CD, there are, nonetheless, strong data from mouse models and from the clinical arena that show CD is related to known disease mechanisms. Several cytokines, inflammatory molecules, and antibodies have been associated with CD. Proposed mechanisms for antibody- and cytokine-mediated neuronal injury include the abrogation of blood-brain barrier integrity with direct access of soluble molecules in the circulation to the brain and ensuing neurotoxicity and microglial activation. No treatments for SLE-mediated CD exist, but potential candidates include agents that inhibit microglial activation, such as angiotensin-converting enzyme inhibitors, or that protect blood-brain barrier integrity, such as C5a receptor blockers. Structural and functional neuroimaging data have shown a range of regional abnormalities in metabolism and white matter microstructural integrity in SLE patients that correlate with CD and could in the future become diagnostic tools and outcome measures in clinical trials aimed at preserving cognitive function in SLE.
Collapse
Affiliation(s)
- Nina Kello
- Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, USA
| | - Erik Anderson
- Elmezzi Graduate School, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, USA
| | - Betty Diamond
- Institute of Molecular Medicine, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, USA
| |
Collapse
|
34
|
Huang MW, Stock AD, Mike EV, Herlitz L, Kolbeck R, Putterman C. Anti-IFNAR treatment does not reverse neuropsychiatric disease in MRL/ lpr lupus mice. Lupus 2019; 28:1510-1523. [PMID: 31474191 DOI: 10.1177/0961203319872265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Many systemic lupus erythematosus patients display a type I interferon (IFN) signature, and IFNα levels positively correlate with disease severity. Previous studies blocking the type I IFN pathway systemically in lupus models showed some beneficial effects. However, its effects on neuropsychiatric manifestations have yet to be carefully assessed, even though IFNα has been associated with induction of depression. Our aim was to investigate whether disrupting the type I IFN pathway would attenuate the development of murine neuropsychiatric lupus. METHODS Female MRL/lpr mice were administered an antitype I IFN receptor (IFNAR) antibody or a control antibody intraperitoneally three times weekly for 12 weeks starting at age 4-5 weeks. Behavior was assessed during and at the end of the treatment schedule. RESULTS No significant differences were seen between the anti-IFNAR- and control-treated mice when assessing for depression-like behavior or cognitive dysfunction, although anti-IFNAR antibody-treated mice displayed significant decreases in levels of IFN-stimulated genes. Anti-IFNAR treatment also did not significantly improve brain histology, cellular infiltration, or blood-brain barrier integrity. CONCLUSIONS Surprisingly, our results showed no improvement in neuropsychiatric disease and suggest that the role of IFNAR signaling in the pathogenesis of neuropsychiatric lupus continues to need to be carefully assessed.
Collapse
Affiliation(s)
- M W Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - A D Stock
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - E V Mike
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - L Herlitz
- Department of Pathology, Cleveland Clinic, Cleveland, OH, USA
| | - R Kolbeck
- Research and Early Development, Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - C Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
35
|
Stock AD, Der E, Gelb S, Huang M, Weidenheim K, Ben-Zvi A, Putterman C. Tertiary lymphoid structures in the choroid plexus in neuropsychiatric lupus. JCI Insight 2019; 4:124203. [PMID: 31167973 PMCID: PMC6629135 DOI: 10.1172/jci.insight.124203] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 04/23/2019] [Indexed: 12/20/2022] Open
Abstract
The central nervous system manifestations of systemic lupus erythematosus (SLE) remain poorly understood. Given the well-defined role of autoantibodies in other lupus manifestations, extensive work has gone into the identification of neuropathic autoantibodies. However, attempts to translate these findings to patients with SLE have yielded mixed results. We used the MRL/MpJ-Faslpr/lpr mouse, a well-established, spontaneous model of SLE, to establish the immune effectors responsible for brain disease. Transcriptomic analysis of the MRL/MpJ-Faslpr/lpr choroid plexus revealed an expression signature driving tertiary lymphoid structure formation, including chemokines related to stromal reorganization and lymphocyte compartmentalization. Additionally, transcriptional profiles indicated various stages of lymphocyte activation and germinal center formation. The extensive choroid plexus infiltrate present in MRL/MpJ-Faslpr/lpr mice with overt neurobehavioral deficits included locally proliferating B and T cells, intercellular interactions between lymphocytes and antigen-presenting cells, as well as evidence for in situ somatic hypermutation and class switch recombination. Furthermore, the choroid plexus was a site for trafficking lymphocytes into the brain. Finally, histological evaluation in human lupus patients with neuropsychiatric manifestations revealed increased leukocyte migration through the choroid plexus. These studies identify a potential new pathway underlying neuropsychiatric lupus and support tertiary lymphoid structure formation in the choroid plexus as a novel mechanism of brain-immune interfacing.
Collapse
Affiliation(s)
- Ariel D. Stock
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Evan Der
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Sivan Gelb
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, The Hebrew University, Ein-Kerem, Jerusalem, Israel
| | - Michelle Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Ayal Ben-Zvi
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, The Hebrew University, Ein-Kerem, Jerusalem, Israel
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
- Division of Rheumatology, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
36
|
Lu F, Lu H, Xie M, Li S, Zu Y, Zhou J, Yu J, Wang S, Ruan Y, Wen C, Xu Z. Limited preventive effect of prednisone on neuropsychiatric symptoms in murine systemic lupus erythematosus. Inflammopharmacology 2019; 27:511-520. [PMID: 30911862 DOI: 10.1007/s10787-019-00587-4] [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: 12/29/2018] [Accepted: 03/15/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate whether glucocorticoids, the hallmark medication for systemic lupus erythematosus (SLE), could prevent the development of neuropsychiatric SLE (NPSLE). METHODS The protective effects of prednisone on NPSLE were tested using the open field, object recognition/placement, forced swim, tail suspension, and sucrose preference tests in MRL/lpr mice. Auto-antibody titres and the weight of lymph nodes were also measured. RESULTS MRL/lpr mice exhibited mild depression at the age of 8 weeks before progressing with spatial cognitive impairment and severe depression-like behaviour at the age of 16 weeks. Treating MRL/lpr mice with prednisone (5 mg/kg) from the age of 8 weeks decreased anti-cardiolipin and anti-N-methyl-D-aspartate (NMDA) receptor antibody titres in the brain, reduced the weight of lymph nodes, and prolonged the floating latency in the forced swim test. However, prednisone (3 or 5 mg/kg) had no preventive effect on the development of spatial cognitive impairment and other depression-like behaviours in MRL/lpr mice. The dose of prednisone had a positive correlation with the floating latency in the forced swim test, while it offered no effects on all other behavioural tests. CONCLUSION Our results provide evidence that early treatment with prednisone had a limited effect on the development of neuropsychiatric symptoms in MRL/lpr mice. Further work is needed in other models beyond NPSLE in MRL/lpr mice before any definitive conclusions are made on the efficacy of prednisone in human NPSLE.
Collapse
Affiliation(s)
- Feilong Lu
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haimei Lu
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Meijuan Xie
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shan Li
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yunyun Zu
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jia Zhou
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jie Yu
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuang Wang
- Epilepsy Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Yeping Ruan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chengping Wen
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Laboratory of Rheumatology, College of Basic Medical Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - Zhenghao Xu
- Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China. .,Laboratory of Rheumatology, College of Basic Medical Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China.
| |
Collapse
|
37
|
Srikumar BN, Naidu PS, Kalidindi N, Paschapur M, Adepu B, Subramani S, Nagar J, Srivastava R, Sreedhara MV, Prasad DS, Das ML, Louis JV, Kuchibhotla VK, Dudhgaonkar S, Pieschl RL, Li YW, Bristow LJ, Ramarao M, Vikramadithyan RK. Diminished responses to monoaminergic antidepressants but not ketamine in a mouse model for neuropsychiatric lupus. J Psychopharmacol 2019; 33:25-36. [PMID: 30484737 DOI: 10.1177/0269881118812102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND A significant proportion of patients suffering from major depression fail to remit following treatment and develop treatment-resistant depression. Developing novel treatments requires animal models with good predictive validity. MRL/lpr mice, an established model of systemic lupus erythematosus, show depression-like behavior. AIMS We evaluated responses to classical antidepressants, and associated immunological and biochemical changes in MRL/lpr mice. METHODS AND RESULTS MRL/lpr mice showed increased immobility in the forced swim test, decreased wheel running and sucrose preference when compared with the controls, MRL/MpJ mice. In MRL/lpr mice, acute fluoxetine (30 mg/kg, intraperitoneally (i.p.)), imipramine (10 mg/kg, i.p.) or duloxetine (10 mg/kg, i.p.) did not decrease the immobility time in the Forced Swim Test. Interestingly, acute administration of combinations of olanzapine (0.03 mg/kg, subcutaneously)+fluoxetine (30 mg/kg, i.p.) or bupropion (10 mg/kg, i.p.)+fluoxetine (30 mg/kg, i.p.) retained efficacy. A single dose of ketamine but not three weeks of imipramine (10 mg/kg, i.p.) or escitalopram (5 mg/kg, i.p.) treatment in MRL/lpr mice restored sucrose preference. Further, we evaluated inflammatory, immune-mediated and neuronal mechanisms. In MRL/lpr mice, there was an increase in autoantibodies' titers, [3H]PK11195 binding and immune complex deposition. There was a significant infiltration of the brain by macrophages, neutrophils and T-lymphocytes. p11 mRNA expression was decreased in the prefrontal cortex. Further, there was an increase in the 5-HT2aR expression, plasma corticosterone and indoleamine 2,3-dioxygenase activity. CONCLUSION In summary, the MRL/lpr mice could be a useful model for Treatment Resistant Depression associated with immune dysfunction with potential to expedite antidepressant drug discovery.
Collapse
Affiliation(s)
- Bettadapura N Srikumar
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Pattipati S Naidu
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | | | - Mahesh Paschapur
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Bharath Adepu
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Siva Subramani
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Jignesh Nagar
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Ratika Srivastava
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Muppana V Sreedhara
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Durga Shiva Prasad
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Manish Lal Das
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Justin V Louis
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Vijaya K Kuchibhotla
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Shailesh Dudhgaonkar
- 1 Disease Sciences and Technology, Biocon-Bristol-Myers Squibb R&D Center, Bangalore, India
| | - Rick L Pieschl
- 2 Neuroscience Biology, Bristol-Myers Squibb Company, Wallingford, CT, USA
| | - Yu-Wen Li
- 2 Neuroscience Biology, Bristol-Myers Squibb Company, Wallingford, CT, USA
| | - Linda J Bristow
- 2 Neuroscience Biology, Bristol-Myers Squibb Company, Wallingford, CT, USA
| | | | | |
Collapse
|
38
|
Magro-Checa C, Steup-Beekman GM, Huizinga TW, van Buchem MA, Ronen I. Laboratory and Neuroimaging Biomarkers in Neuropsychiatric Systemic Lupus Erythematosus: Where Do We Stand, Where To Go? Front Med (Lausanne) 2018; 5:340. [PMID: 30564579 PMCID: PMC6288259 DOI: 10.3389/fmed.2018.00340] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/19/2018] [Indexed: 01/18/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by multi-systemic involvement. Nervous system involvement in SLE leads to a series of uncommon and heterogeneous neuropsychiatric (NP) manifestations. Current knowledge on the underlying pathogenic processes and their subsequent pathophysiological changes leading to NP-SLE manifestations is incomplete. Several putative laboratory biomarkers have been proposed as contributors to the genesis of SLE-related nervous system damage. Alongside the laboratory biomarkers, several neuroimaging tools have shown to reflect the nature of tissue microstructural damage associated with SLE, and thus were suggested to contribute to the understanding of the pathophysiological changes and subsequently help in clinical decision making. However, the number of useful biomarkers in NP-SLE in clinical practice is disconcertingly modest. In some cases it is not clear whether the biomarker is truly involved in pathogenesis, or the result of non-specific pathophysiological changes in the nervous system (e.g., neuroinflammation) or whether it is the consequence of a concomitant underlying abnormality related to SLE activity. In order to improve the diagnosis of NP-SLE and provide a better targeted care to these patients, there is still a need to develop and validate a range of biomarkers that reliably capture the different aspects of disease heterogeneity. This article critically reviews the current state of knowledge on laboratory and neuroimaging biomarkers in NP-SLE, discusses the factors that need to be addressed to make these biomarkers suitable for clinical application, and suggests potential future research paths to address important unmet needs in the NP-SLE field.
Collapse
Affiliation(s)
- César Magro-Checa
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands.,Department of Rheumatology, Zuyderland Medical Center, Heerlen, Netherlands
| | | | - Tom W Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Itamar Ronen
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
39
|
Meneses G, Cárdenas G, Espinosa A, Rassy D, Pérez-Osorio IN, Bárcena B, Fleury A, Besedovsky H, Fragoso G, Sciutto E. Sepsis: developing new alternatives to reduce neuroinflammation and attenuate brain injury. Ann N Y Acad Sci 2018; 1437:43-56. [DOI: 10.1111/nyas.13985] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Gabriela Meneses
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Graciela Cárdenas
- Instituto Nacional de Neurología y Neurocirugía; SSA; Mexico City Mexico
| | - Alejandro Espinosa
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Dunia Rassy
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Ivan Nicolás Pérez-Osorio
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Brandon Bárcena
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Agnes Fleury
- Instituto Nacional de Neurología y Neurocirugía; SSA; Mexico City Mexico
| | - Hugo Besedovsky
- The Institute of Physiology and Pathophysiology, Medical Faculty; Philipps University; Marburg Germany
| | - Gladis Fragoso
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| | - Edda Sciutto
- Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| |
Collapse
|
40
|
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: 5.8] [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
|
41
|
Mike EV, Makinde HM, Der E, Stock A, Gulinello M, Gadhvi GT, Winter DR, Cuda CM, Putterman C. Neuropsychiatric Systemic Lupus Erythematosus Is Dependent on Sphingosine-1-Phosphate Signaling. Front Immunol 2018; 9:2189. [PMID: 30319641 PMCID: PMC6168636 DOI: 10.3389/fimmu.2018.02189] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022] Open
Abstract
About 40% of patients with systemic lupus erythematosus experience diffuse neuropsychiatric manifestations, including impaired cognition and depression. Although the pathogenesis of diffuse neuropsychiatric SLE (NPSLE) is not fully understood, loss of brain barrier integrity, autoreactive antibodies, and pro-inflammatory cytokines are major contributors to disease development. Fingolimod, a sphingosine-1-phosphate (S1P) receptor modulator, prevents lymphocyte egress from lymphoid organs through functional antagonism of S1P receptors. In addition to reducing the circulation of autoreactive lymphocytes, fingolimod has direct neuroprotective effects such as preserving brain barrier integrity and decreasing pro-inflammatory cytokine secretion by astrocytes and microglia. Given these effects, we hypothesized that fingolimod would attenuate neurobehavioral deficits in MRL-lpr/lpr (MRL/lpr) mice, a validated neuropsychiatric lupus model. Fingolimod treatment was initiated after the onset of disease, and mice were assessed for alterations in cognitive function and emotionality. We found that fingolimod significantly attenuated spatial memory deficits and depression-like behavior in MRL/lpr mice. Immunofluorescent staining demonstrated a dramatic lessening of brain T cell and macrophage infiltration, and a significant reduction in cortical leakage of serum albumin, in fingolimod treated mice. Astrocytes and endothelial cells from treated mice exhibited reduced expression of inflammatory genes, while microglia showed differential regulation of key immune pathways. Notably, cytokine levels within the cortex and hippocampus were not appreciably decreased with fingolimod despite the improved neurobehavioral profile. Furthermore, despite a reduction in splenomegaly, lymphadenopathy, and circulating autoantibody titers, IgG deposition within the brain was unaffected by treatment. These findings suggest that fingolimod mediates attenuation of NPSLE through a mechanism that is not dependent on reduction of autoantibodies or cytokines, and highlight modulation of the S1P signaling pathway as a novel therapeutic target in lupus involving the central nervous system.
Collapse
Affiliation(s)
- Elise V Mike
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Hadijat M Makinde
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Evan Der
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ariel Stock
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maria Gulinello
- Dominick P. Purpura Department of Neuroscience Animal Behavioral Core, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Gaurav T Gadhvi
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Deborah R Winter
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Carla M Cuda
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
42
|
Hao F, Tian M, Feng Y, Quan C, Chen Y, Chen S, Wei M. Abrogation of Lupus Nephritis in Somatic Hypermutation-Deficient MRL/lpr Mice. THE JOURNAL OF IMMUNOLOGY 2018; 200:3905-3912. [PMID: 29728506 DOI: 10.4049/jimmunol.1800115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/31/2018] [Indexed: 01/31/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease posing threats to multiple organs in the human body. As a typical manifestation of SLE, lupus nephritis is characterized by a series of pathological changes in glomerulus as well as accumulation of pathogenic autoreactive IgG with complement in the kidney that dramatically disrupts renal functions. Activation-induced deaminase (AID), which governs both somatic hypermutation (SHM) and class-switch recombination (CSR), has been shown to be essential for the regulation of SLE. However, the relative contributions of SHM and CSR to SLE pathology have not been determined. Based on the available AIDG23S mice, we successfully established an AIDG23S MRL/lpr mouse model, in which SHM is specifically abolished, although CSR is largely unaffected. We found that the abrogation of SHM effectively alleviated SLE-associated histopathological alterations, such as expansion of the mesangial matrix and thickening of the basement membrane of Bowman's capsule as well as infiltration of inflammatory cells. Compared with SLE mice, AIDG23S MRL/lpr mice exhibited decreased proteinuria, blood urea nitrogen, and creatinine, indicating that the loss of SHM contributed to the recovery of renal functions. As a consequence, the life span of those SHM-deficient MRL/lpr mice was extended. Together, we provide direct evidence pinpointing a vital role of SHM in the control of SLE development.
Collapse
Affiliation(s)
- Fengqi Hao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, 130024, People's Republic of China.,School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130022, People's Republic of China; and
| | - Miaomiao Tian
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Chao Quan
- Ministry of Education Key Laboratory of Model Animal for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, 210061, People's Republic of China
| | - Yixi Chen
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, 130024, People's Republic of China
| | - Shuai Chen
- Ministry of Education Key Laboratory of Model Animal for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing, 210061, People's Republic of China
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, 130024, People's Republic of China;
| |
Collapse
|
43
|
Jain S, Stock A, Macian F, Putterman C. A Distinct T Follicular Helper Cell Subset Infiltrates the Brain in Murine Neuropsychiatric Lupus. Front Immunol 2018; 9:487. [PMID: 29593732 PMCID: PMC5859360 DOI: 10.3389/fimmu.2018.00487] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/23/2018] [Indexed: 11/30/2022] Open
Abstract
Neuropsychiatric symptoms in systemic lupus erythematosus (SLE) are not uncommon, yet the mechanisms underlying disease initiation and progression in the brain are incompletely understood. Although the role of T cells in other lupus target organs such as the kidney is well defined, which T cells contribute to the pathogenesis of neuropsychiatric SLE is not known. The present study was aimed at characterizing the CD4 T cell populations that are present in the choroid plexus (CP) of MRL/MpJ-faslpr mice, the primary site of brain infiltration in this classic lupus mouse model which exhibits a prominent neurobehavioral phenotype. T cells infiltrating the CP of MRL/MpJ-faslpr mice were characterized and subset identification was done by multiparameter flow cytometry. We found that the infiltrating CD4 T cells are activated and have an effector phenotype. Importantly, CD4 T cells have a T follicular helper cell (TFH) like phenotype, as evidenced by their surface markers and signature cytokine, IL-21. In addition, CD4 TFH cells also secrete significant levels of IFN-γ and express Bcl-6, thereby conforming to a potentially pathogenic T helper population that can drive the disease progression. Interestingly, the regulatory axis comprising CD4 T regulatory cells is diminished. These results suggest that accumulation of CD4 TFH in the brain of MRL/MpJ-faslpr mice may contribute to the neuropsychiatric manifestations of SLE, and point to this T cell subset as a possible novel therapeutic candidate.
Collapse
Affiliation(s)
- Shweta Jain
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ariel Stock
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Chaim Putterman
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
44
|
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
|
45
|
Monaco A, Ferrandino I, Boscaino F, Cocca E, Cigliano L, Maurano F, Luongo D, Spagnuolo MS, Rossi M, Bergamo P. Conjugated linoleic acid prevents age-dependent neurodegeneration in a mouse model of neuropsychiatric lupus via the activation of an adaptive response. J Lipid Res 2018; 59:48-57. [PMID: 29167408 PMCID: PMC5748496 DOI: 10.1194/jlr.m079400] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/14/2017] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress is a key mediator of autoimmune/neurodegenerative disorders. The antioxidant/anti-inflammatory effect of a synthetic conjugated linoleic acid (CLA) mixture in MRL/MpJ-Fas lpr mice (MRL/lpr), an animal model of neuropsychiatric lupus, was previously associated with the improvement of nuclear factor-E2-related factor 2 (Nrf2) defenses in the spleen and liver. However, little is known about the neuroprotective ability of a CLA mixture. This study investigated the age-dependent progression of oxidative stress and the hyperactivation of redox-sensitive compensatory pathways (macroautophagy, Nrf2) in old/diseased MRL/lpr mice brains and examines the effect produced by dietary CLA supplementation. Disrupted redox homeostasis was evidenced in the blood, liver, and brain of 21- to 22-week-old MRL/lpr (Old) mice compared with 8- to 10-week-old MRL/lpr (Young) animals. This alteration was associated with significant hyperactivation of compensatory mechanisms (macroautophagy, Nrf2, and astrocyte activation) in the brains of Old mice. Five-week daily supplementation with CLA (650 mg/kg-1 body weight) of 16-week-old (CLA+Old) mice diminished all the pathological hallmarks at a level comparable to Young mice or healthy controls (BALB/c). Such data demonstrated that MRL/lpr mice can serve as a valuable model for the evaluation of the effectiveness of neuroprotective drugs. Notably, the preventive effect provided by CLA supplementation against age-associated neuronal damage and hyperactivation of compensatory mechanisms suggests that the activation of an adaptive response is at least in part accountable for its neuroprotective ability.
Collapse
Affiliation(s)
- Antonio Monaco
- Department of Biology, University "Federico II" of Naples, Italy
| | - Ida Ferrandino
- Department of Biology, University "Federico II" of Naples, Italy
| | - Floriana Boscaino
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Ennio Cocca
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, Italy
| | - Luisa Cigliano
- Department of Biology, University "Federico II" of Naples, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Diomira Luongo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Maria Stefania Spagnuolo
- Institute for Animal Production System in Mediterranean Environment, National Research Council (ISPAAM, CNR) Naples, Italy
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Paolo Bergamo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| |
Collapse
|
46
|
He X, Zhang Y, Zhu A, Zeng K, Zhang X, Gong L, Peng Y, Lai K, Qu S. Suppression of interleukin 17 contributes to the immunomodulatory effects of adipose-derived stem cells in a murine model of systemic lupus erythematosus. Immunol Res 2017; 64:1157-1167. [PMID: 27617336 DOI: 10.1007/s12026-016-8866-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Due to roles in immunoregulation and low immunogenicity, mesenchymal stem cells have been suggested to be potent regulators of the immune response and may represent promising treatments for autoimmune disease. Adipose-derived stem cells (ADSCs), stromal cells derived from adipose tissue, were investigated with allogeneic ADSCs in B6.MRL/lpr mice, a murine model of systemic lupus erythematosus (SLE). We intravenously injected allogeneic ADSCs into SLE mice after disease onset and report that ADSCs reduced anti-ds DNA antibodies in serum and proteinuria in SLE mice. Also, ADSCs decreased IL-17 and IL-6 expression in serum of SLE mice. ADSCs alleviated renal damage and inflammatory cell infiltration and edema of the renal interstitium. Furthermore, ADSCs significantly downregulated renal IL-17 and CD68 expression, suggesting that ADSCs suppressed renal inflammation. ADSCs also decreased IL-17 mRNA expression and increased Foxp3, ROR-γt and miR-23b mRNA expression in renal tissue in SLE mice. ADSCs reduced renal protein expression of TAB 2 and IKK-α in SLE mice. Thus, ADSCs may be a novel potential therapy for treating SLE.
Collapse
Affiliation(s)
- Xiaoliang He
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China.,Department of Blood Transfusion, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, People's Republic of China
| | - Yunlong Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China.,Department of Blood Transfusion, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, People's Republic of China
| | - Ai Zhu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiuping Zhang
- Teaching Center of Experimental Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Li Gong
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yusheng Peng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Kuan Lai
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Shaogang Qu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China. .,Department of Blood Transfusion, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, People's Republic of China.
| |
Collapse
|
47
|
Lei HW, Wang JY, Dang QJ, Yang F, Liu X, Zhang JH, Li Y. Neuropsychiatric involvement in lupus is associated with the Nogo-a/NgR1 pathway. J Neuroimmunol 2017; 311:22-28. [DOI: 10.1016/j.jneuroim.2017.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/24/2017] [Accepted: 06/18/2017] [Indexed: 12/11/2022]
|
48
|
Pikman R, Kivity S, Levy Y, Arango MT, Chapman J, Yonath H, Shoenfeld Y, Gofrit SG. Neuropsychiatric SLE: from animal model to human. Lupus 2017; 26:470-477. [PMID: 28394237 DOI: 10.1177/0961203317694261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Animal models are a key element in disease research and treatment. In the field of neuropsychiatric lupus research, inbred, transgenic and disease-induced mice provide an opportunity to study the pathogenic routes of this multifactorial illness. In addition to achieving a better understanding of the immune mechanisms underlying the disease onset, supplementary metabolic and endocrine influences have been discovered and investigated. The ever-expanding knowledge about the pathologic events that occur at disease inception enables us to explore new drugs and therapeutic approaches further and to test them using the same animal models. Discovery of the molecular targets that constitute the pathogenic basis of the disease along with scientific advancements allow us to target these molecules with monoclonal antibodies and other specific approaches directly. This novel therapy, termed "targeted biological medication" is a promising endeavor towards producing drugs that are more effective and less toxic. Further work to discover additional molecular targets in lupus' pathogenic mechanism and to produce drugs that neutralize their activity is needed to provide patients with safe and efficient methods of controlling and treating the disease.
Collapse
Affiliation(s)
- R Pikman
- 1 Israel Defense Forces Medical Corps, Ramat Gan, Israel
| | - S Kivity
- 2 Department of Medicine A, Sheba Medical Center, Tel-Hashomer, Israel.,3 The Zabludovicz Center for Autoimmune Diseases.,4 The Dr Pinchas Borenstein Talpiot Medical Leadership Program 2013; and Sheba Medical Center, Tel-Hashomer, Israel.,5 Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Israel
| | - Y Levy
- 6 Department of Medicine E, Meir Medical Center, Kfar Saba, Israel; affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M-T Arango
- 3 The Zabludovicz Center for Autoimmune Diseases.,7 Doctoral Program in Biomedical Sciences, Universidad del Rosario, Bogotá-Colombia
| | - J Chapman
- 5 Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Israel.,8 Department of Neurology, Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - H Yonath
- 2 Department of Medicine A, Sheba Medical Center, Tel-Hashomer, Israel.,5 Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Israel.,9 The Danek Gartner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Israel
| | - Y Shoenfeld
- 3 The Zabludovicz Center for Autoimmune Diseases.,5 Sackler Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Israel.,10 Incumbent of the Laura Schwarz-Kip Chair for Research of Autoimmune Diseases, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - S G Gofrit
- 2 Department of Medicine A, Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
49
|
Muller S, Brun S, René F, de Sèze J, Loeffler JP, Jeltsch-David H. Autophagy in neuroinflammatory diseases. Autoimmun Rev 2017; 16:856-874. [DOI: 10.1016/j.autrev.2017.05.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 05/20/2017] [Indexed: 12/12/2022]
|
50
|
Pozsgay J, Szekanecz Z, Sármay G. Antigen-specific immunotherapies in rheumatic diseases. Nat Rev Rheumatol 2017; 13:525-537. [DOI: 10.1038/nrrheum.2017.107] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|