1
|
Nagasubramanian K, Gupta K. Interactome analysis implicates class II transactivator (CIITA) in depression and other neuroinflammatory disorders. Int J Neurosci 2024; 134:1153-1171. [PMID: 37933915 DOI: 10.1080/00207454.2023.2279502] [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: 08/02/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Inappropriate inflammatory responses within the nervous system (neuroinflammation) have been implicated in several neurological conditions. Class II transactivator (CIITA), a principal regulator of the major histocompatibility complex II (MHCII), is known to play essential roles in inflammation. Hence, CIITA and its interactors could be potentially involved in multiple neurological disorders. However, the molecular mechanisms underlying CIITA-mediated neuroinflammation (NI) are yet to be understood. MATERIALS AND METHODS In this regard, we analyzed the potential involvement of CIITA and its interactome in the regulation of neuroinflammation. In the present study, using various computational tools, we aimed (1) to identify NI-related proteins, (2) to filter the critical interactors in the CIITA-NI network, and (3) to analyze the protein-disease interactions and the associated molecular pathways through which CIITA could influence neuroinflammation. RESULTS CIITA was found to interact with P T GS2, GSK3B, and NR3C1 and may influence depressive disorders. Further, the IL4/IL13 pathway was found to be potentially underlying the CIITA-interactomemediated effects on neurological disorders. Moreover, CIITA was found to be connected to genes associated with depressive disorder through IL4, wherein CIITA was found to be potentially involved in depressive disorders through IL-4/IL-13 and hippo pathways. However, the present study is based on the existing data on protein interactomes and could be re-evaluated as newer interactions are discovered. Also, the functional mechanisms of CIITA's roles in neuroinflammation must be evaluated further. CONCLUSION Notwithstanding these limitations, the results presented here, could form a basis for further experimental studies to assess CIITA as a potential therapeutic target in managing depression and other neuroinflammatory disorders.
Collapse
Affiliation(s)
- Kishore Nagasubramanian
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Krishnakant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
- NCCS, Pune, India
| |
Collapse
|
2
|
Zahedipour F, Hosseini SA, Reiner Ž, Tedeschi-Reiner E, Jamialahmadi T, Sahebkar A. Therapeutic Effects of Statins: Promising Drug for Topical and Transdermal Administration. Curr Med Chem 2024; 31:3149-3166. [PMID: 37157198 DOI: 10.2174/0929867330666230508141434] [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: 10/07/2022] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 05/10/2023]
Abstract
Statins are HMG-CoA reductase inhibitors and decrease plasma low-density lipoprotein cholesterol (LDL-C) levels. They are well tolerated, and because of their LDL-C-lowering effect, they are utilized to decrease the risk of atherosclerosis and cardiovascular disease. However, statins have pleiotropic effects, including immunomodulatory, anti-inflammatory, antioxidant, and anticancer. Currently, oral administration is the only Food and Drug Administration (FDA)-approved route of administration for statins. However, other administration routes have demonstrated promising results in different pre-clinical and clinical studies. For instance, statins also seem beneficial in dermatitis, psoriasis, vitiligo, hirsutism, uremic pruritus, and graft-versus-host disease. Topically applied statins have been studied to treat seborrhea, acne, rhinophyma, and rosacea. They also have beneficial effects in contact dermatitis and wound healing in animal studies, (HIV) infection, osseointegration, porokeratosis, and some ophthalmologic diseases. Topical and transdermal application of statins is a non-invasive drug administration method that has shown significant results in bypassing the first-pass metabolism in the liver, thereby reducing possible adverse effects. This study reviews the multifaceted molecular and cellular impacts of statins, their topical and transdermal application, novel delivery systems, such as nanosystems for topical and transdermal administration and the challenges concerning this approach.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Atefe Hosseini
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Željko Reiner
- University Hospital Center Zagreb, Department of Internal Medicine, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | | | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Nagasubramanian K, Jha S, Rathore AS, Gupta K. Identification of small molecule modulators of class II transactivator-I using computational approaches. J Biomol Struct Dyn 2023; 41:8349-8361. [PMID: 36224172 DOI: 10.1080/07391102.2022.2133011] [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] [Received: 05/31/2022] [Accepted: 09/30/2022] [Indexed: 10/17/2022]
Abstract
Major histocompatibility complex II (MHCII), a mediator of the innate and adaptive immune system, plays a central role in regulating inflammation and its progression. Class II transactivator (CIITA) is a master regulator of MHCII expression and controls antigen presentation followed by T-cell activation. Regulation of inflammation by modulation of CIITA has been suggested as a promising intervention for several disorders, including neuroinflammation, rheumatoid arthritis and other autoimmune diseases. This study aimed to (i) identify possible pharmacological agents which could bind to and inhibit isoform I of CIITA (CIITA-I) and (ii) determine their strength of interactions. The structure of CIITA-I isoform was predicted using phyre2 and refined via 3D refine. Loops were refined using ModBase, followed by quality assessment based on ERRAT value. The refined 3D structure was subjected to docking via Maestro (from Schrodinger) using glide module against small molecule databases. Molecules having the least glide score and favorable ADME properties were subjected to molecular simulation by GROMACS. We used the 3D refined structure of CIITA-I, with a score of 83.4% in ERRAT for docking studies. The ligand 4-(2-((6-oxo-4-phenyl-1,6-dihydropyrimidin-2-yl) thio) acetamido) benzamide (ZINC5154833), showed maximum glide score (-6.591) followed by N-[4-(3-oxo-3-{4-[3-(trifluoromethyl) phenyl] piperazin-1-yl} propyl)-1,3-thiazol-2-yl] benzamide (F5254-0161, glide score -6.41). Simulation studies using GROMACS showed F5254-0161 to have a more stable interaction with CIITA-I. Based on our analysis, we propose ZINC5154833 and F5254-0161 as potential modulators for CIITA-I.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kishore Nagasubramanian
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Shanker Jha
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Anuranjan Singh Rathore
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Krishnakant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| |
Collapse
|
4
|
Hobson BD, Stanley AT, De Los Santos MB, Culbertson B, Mosharov EV, Sims PA, Sulzer D. Conserved and cell type-specific transcriptional responses to IFN-γ in the ventral midbrain. Brain Behav Immun 2023; 111:277-291. [PMID: 37100211 PMCID: PMC10460506 DOI: 10.1016/j.bbi.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/28/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023] Open
Abstract
Dysregulated inflammation within the central nervous system (CNS) contributes to neuropathology in infectious, autoimmune, and neurodegenerative disease. With the exception of microglia, major histocompatibility complex (MHC) proteins are virtually undetectable in the mature, healthy central nervous system (CNS). Neurons have generally been considered incapable of antigen presentation, and although interferon gamma (IFN-γ) can elicit neuronal MHC class I (MHC-I) expression and antigen presentation in vitro, it has been unclear whether similar responses occur in vivo. Here we directly injected IFN-γ into the ventral midbrain of mature mice and analyzed gene expression profiles of specific CNS cell types. We found that IFN-γ upregulated MHC-I and associated mRNAs in ventral midbrain microglia, astrocytes, oligodendrocytes, and GABAergic, glutamatergic, and dopaminergic neurons. The core set of IFN-γ-induced genes and their response kinetics were similar in neurons and glia, but with a lower amplitude of expression in neurons. A diverse repertoire of genes was upregulated in glia, particularly microglia, which were the only cells to undergo cellular proliferation and express MHC classII (MHC-II) and associated genes. To determine if neurons respond directly via cell-autonomous IFN-γ receptor (IFNGR) signaling, we produced mutant mice with a deletion of the IFN-γ-binding domain of IFNGR1 in dopaminergic neurons, which resulted in a complete loss of dopaminergic neuronal responses to IFN-γ. Our results demonstrate that IFN-γ induces neuronal IFNGR signaling and upregulation of MHC-I and related genes in vivo, although the expression level is low compared to oligodendrocytes, astrocytes, and microglia.
Collapse
Affiliation(s)
- Benjamin D Hobson
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Adrien T Stanley
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Mark B De Los Santos
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Bruce Culbertson
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, United States; Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, United States; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States.
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, United States; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States.
| |
Collapse
|
5
|
Dama G, Hu X, Yan Y, Li Y, Li H, Yang F, Liu Y, Lin J. Identification and protective role of CD34 + stromal cells/telocytes in experimental autoimmune encephalomyelitis (EAE) mouse spleen. Histochem Cell Biol 2023:10.1007/s00418-023-02186-5. [PMID: 37014442 DOI: 10.1007/s00418-023-02186-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 04/05/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a classical animal model of human multiple sclerosis (MS) that is most commonly used to study the neuropathology and therapeutic effects of the disease. Telocytes (TCs) are a specialized type of interstitial or mesenchymal cell first identified by Popescu in various tissues and organs. However, the existence, distribution and role of CD34+ stromal cells (SCs)/TCs in the EAE-induced mouse spleen remain to be elucidated. We conducted immunohistochemistry, immunofluorescence (double staining for CD34 and c-kit, vimentin, F4/80, CD163, Nanog, Sca-1, CD31 or tryptase) and transmission electron microscopy experiments to investigate the existence, distribution and role of CD34+ SCs/TCs in the EAE-induced mouse spleen. Interestingly, immunohistochemistry, double-immunofluorescence, and transmission electron microscopy results revealed that CD34+ SCs/TCs were significantly upregulated in the EAE mouse spleen. Immunohistochemical or double-immunofluorescence staining of CD34+ SCs/TCs showed positive expression for CD34, c-kit, vimentin, CD34/vimentin, c-kit/vimentin and CD34/c-kit, and negative expression for CD31 and tryptase. Transmission electron microscopy (TEM) results demonstrated that CD34+ SCs/TCs established close connections with lymphocytes, reticular cells, macrophages, endothelial cells and erythrocytes. Furthermore, we also found that M1 (F4/80) or M2 (CD163) macrophages, and haematopoietic, pluripotent stem cells were markedly increased in EAE mice. Our results suggest that CD34+ SCs/TCs are abundant and may play a contributing role in modulating the immune response, recruiting macrophages and proliferation of haematopoietic and pluripotent stem cells following injury to promote tissue repair and regeneration in EAE mouse spleens. This suggests that their transplantation combined with stem cells might represent a promising therapeutic target for the treatment and prevention of multiple autoimmune and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Ganesh Dama
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China
- Department of Community Health, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200, Kepala Batas, Pulau Pinang, Malaysia
| | - Xiaoxi Hu
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Yushan Yan
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Yonghai Li
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Han Li
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Fen Yang
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yanli Liu
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China.
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China.
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China.
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China.
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
6
|
Sakowska J, Arcimowicz Ł, Jankowiak M, Papak I, Markiewicz A, Dziubek K, Kurkowiak M, Kote S, Kaźmierczak-Siedlecka K, Połom K, Marek-Trzonkowska N, Trzonkowski P. Autoimmunity and Cancer-Two Sides of the Same Coin. Front Immunol 2022; 13:793234. [PMID: 35634292 PMCID: PMC9140757 DOI: 10.3389/fimmu.2022.793234] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Autoimmune disease results from the immune response against self-antigens, while cancer develops when the immune system does not respond to malignant cells. Thus, for years, autoimmunity and cancer have been considered as two separate fields of research that do not have a lot in common. However, the discovery of immune checkpoints and the development of anti-cancer drugs targeting PD-1 (programmed cell death receptor 1) and CTLA-4 (cytotoxic T lymphocyte antigen 4) pathways proved that studying autoimmune diseases can be extremely helpful in the development of novel anti-cancer drugs. Therefore, autoimmunity and cancer seem to be just two sides of the same coin. In the current review, we broadly discuss how various regulatory cell populations, effector molecules, genetic predisposition, and environmental factors contribute to the loss of self-tolerance in autoimmunity or tolerance induction to cancer. With the current paper, we also aim to convince the readers that the pathways involved in cancer and autoimmune disease development consist of similar molecular players working in opposite directions. Therefore, a deep understanding of the two sides of immune tolerance is crucial for the proper designing of novel and selective immunotherapies.
Collapse
Affiliation(s)
- Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Łukasz Arcimowicz
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Martyna Jankowiak
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ines Papak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Dziubek
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Sachin Kote
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | | | - Karol Połom
- Department of Surgical Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
7
|
Labib D, Wang Z, Prakash P, Zimmer M, Smith MD, Frazel PW, Barbar L, Sapar ML, Calabresi PA, Peng J, Liddelow SA, Fossati V. Proteomic Alterations and Novel Markers of Neurotoxic Reactive Astrocytes in Human Induced Pluripotent Stem Cell Models. Front Mol Neurosci 2022; 15:870085. [PMID: 35592112 PMCID: PMC9113221 DOI: 10.3389/fnmol.2022.870085] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/29/2022] [Indexed: 12/20/2022] Open
Abstract
Astrocytes respond to injury, infection, and inflammation in the central nervous system by acquiring reactive states in which they may become dysfunctional and contribute to disease pathology. A sub-state of reactive astrocytes induced by proinflammatory factors TNF, IL-1α, and C1q ("TIC") has been implicated in many neurodegenerative diseases as a source of neurotoxicity. Here, we used an established human induced pluripotent stem cell (hiPSC) model to investigate the surface marker profile and proteome of TIC-induced reactive astrocytes. We propose VCAM1, BST2, ICOSL, HLA-E, PD-L1, and PDPN as putative, novel markers of this reactive sub-state. We found that several of these markers colocalize with GFAP+ cells in post-mortem samples from people with Alzheimer's disease. Moreover, our whole-cells proteomic analysis of TIC-induced reactive astrocytes identified proteins and related pathways primarily linked to potential engagement with peripheral immune cells. Taken together, our findings will serve as new tools to purify reactive astrocyte subtypes and to further explore their involvement in immune responses associated with injury and disease.
Collapse
Affiliation(s)
- David Labib
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Zhen Wang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Priya Prakash
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, United States
| | - Matthew Zimmer
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | - Paul W. Frazel
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, United States
| | - Lilianne Barbar
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Maria L. Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Shane A. Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, United States
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, United States
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| |
Collapse
|
8
|
Manouchehri N, Salinas VH, Rabi Yeganeh N, Pitt D, Hussain RZ, Stuve O. Efficacy of Disease Modifying Therapies in Progressive MS and How Immune Senescence May Explain Their Failure. Front Neurol 2022; 13:854390. [PMID: 35432156 PMCID: PMC9009145 DOI: 10.3389/fneur.2022.854390] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
The advent of disease modifying therapies (DMT) in the past two decades has been the cornerstone of successful clinical management of multiple sclerosis (MS). Despite the great strides made in reducing the relapse frequency and occurrence of new signal changes on neuroimaging in patients with relapsing remitting MS (RRMS) by approved DMT, it has been challenging to demonstrate their effectiveness in non-active secondary progressive MS (SPMS) and primary progressive MS (PPMS) disease phenotypes. The dichotomy of DMT effectiveness between RRMS and progressive MS informs on distinct pathogeneses of the different MS phenotypes. Conversely, factors that render patients with progressive MS resistant to therapy are not understood. Thus far, age has emerged as the main correlate of the transition from RRMS to SPMS. Whether it is aging and age-related factors or the underlying immune senescence that qualitatively alter immune responses as the disease transitions to SPMS, that diminish DMT effectiveness, or both, is currently not known. Here, we will discuss the role of immune senescence on different arms of the immune system, and how it may explain relative DMT resistance.
Collapse
Affiliation(s)
- Navid Manouchehri
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Victor H. Salinas
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Negar Rabi Yeganeh
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - David Pitt
- Department of Neurology, Yale University, New Haven, CT, United States
| | - Rehana Z. Hussain
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Medical Service Dallas, Veterans Affairs Medical Center, Dallas, TX, United States
- *Correspondence: Olaf Stuve
| |
Collapse
|
9
|
Zahedipour F, Guest PC, Majeed M, Al-Rasadi K, Jamialahmadi T, Sahebkar A. Multiplex Testing of the Effect of Statins on Disease Severity Risk in COVID-19 Cases. Methods Mol Biol 2022; 2511:273-284. [PMID: 35838967 DOI: 10.1007/978-1-0716-2395-4_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Statins have pleiotropic effects on inflammatory responses in addition to their lipid-lowering action, which contributes to their favorable effect on cardiovascular disorders. Statins affect adhesion, migration, antigen presentation, and cytokine generation of immune cells. Pre-clinical and clinical studies suggest that statin intervention targeted early in the infection might help COVID-19 patients to reduce the effects of acute respiratory distress syndrome (ARDS), the cytokine storm, and vascular collapse by modulating harmful pathogenic mechanisms. This chapter presents a protocol for measuring blood-based biomarkers predictive of these responses in COVID-19 patients using two specific multiplex immunoassays that target proteins that differ widely in concentration.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | | | | | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Medicine, The University of Western Australia, Perth, Australia.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Sanmarco LM, Polonio CM, Wheeler MA, Quintana FJ. Functional immune cell-astrocyte interactions. J Exp Med 2021; 218:212503. [PMID: 34292315 PMCID: PMC8302447 DOI: 10.1084/jem.20202715] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Astrocytes are abundant glial cells in the central nervous system (CNS) that control multiple aspects of health and disease. Through their interactions with components of the blood–brain barrier (BBB), astrocytes not only regulate BBB function, they also sense molecules produced by peripheral immune cells, including cytokines. Here, we review the interactions between immune cells and astrocytes and their roles in health and neurological diseases, with a special focus on multiple sclerosis (MS). We highlight known pathways that participate in astrocyte crosstalk with microglia, NK cells, T cells, and other cell types; their contribution to the pathogenesis of neurological diseases; and their potential value as therapeutic targets.
Collapse
Affiliation(s)
- Liliana M Sanmarco
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Carolina M Polonio
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Neuroimmune Interactions Laboratory, Immunology Department, Instituto de Ciências Biomédicas IV, University of São Paulo, São Paulo, Brazil
| | - Michael A Wheeler
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Francisco J Quintana
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
11
|
Liddelow SA, Marsh SE, Stevens B. Microglia and Astrocytes in Disease: Dynamic Duo or Partners in Crime? Trends Immunol 2020; 41:820-835. [PMID: 32819809 DOI: 10.1016/j.it.2020.07.006] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 02/08/2023]
Abstract
Microglia-astrocyte interactions represent a delicate balance affecting neural cell functions in health and disease. Tightly controlled to maintain homeostasis during physiological conditions, rapid and prolonged departures during disease, infection, and following trauma drive multiple outcomes: both beneficial and detrimental. Recent sequencing studies at the bulk and single-cell level in humans and rodents provide new insight into microglia-astrocyte communication in homeostasis and disease. However, the complex changing ways these two cell types functionally interact has been a barrier to understanding disease initiation, progression, and disease mechanisms. Single cell sequencing is providing new insights; however, many questions remain. Here, we discuss how to bridge transcriptional states to specific functions so we can develop therapies to mediate negative effects of altered microglia-astrocyte interactions.
Collapse
Affiliation(s)
- Shane A Liddelow
- Neuroscience Institute, NYU School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, USA; Department of Ophthalmology, NYU School of Medicine, New York, NY, USA.
| | - Samuel E Marsh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA., USA.
| |
Collapse
|
12
|
Castellani G, Schwartz M. Immunological Features of Non-neuronal Brain Cells: Implications for Alzheimer's Disease Immunotherapy. Trends Immunol 2020; 41:794-804. [PMID: 32800704 DOI: 10.1016/j.it.2020.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/08/2020] [Accepted: 07/11/2020] [Indexed: 12/23/2022]
Abstract
An interaction network exists among cells within the brain, maintaining brain homeostasis and ensuring its functional plasticity. In addition to neurons, participating cells include astrocytes, oligodendrocytes, and microglia. Peripheral immune cells, such as monocytes and lymphocytes, have also been found to play an important role in supporting the brain in health and assisting in its repair. Here, we describe the multiple immune-specific modes of cellular dialogue among cells within the mammalian brain and their crosstalk with the periphery in both health and disease. We further suggest that interventions directed at boosting the peripheral immune response can restore the balance between the brain and the immune system and can rewire their communication to modify chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Giulia Castellani
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
13
|
Božić M, Verkhratsky A, Zorec R, Stenovec M. Exocytosis of large-diameter lysosomes mediates interferon γ-induced relocation of MHC class II molecules toward the surface of astrocytes. Cell Mol Life Sci 2020; 77:3245-3264. [PMID: 31667557 PMCID: PMC7391398 DOI: 10.1007/s00018-019-03350-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/01/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Abstract
Astrocytes are the key homeostatic cells in the central nervous system; initiation of reactive astrogliosis contributes to neuroinflammation. Pro-inflammatory cytokine interferon γ (IFNγ) induces the expression of the major histocompatibility complex class II (MHCII) molecules, involved in antigen presentation in reactive astrocytes. The pathway for MHCII delivery to the astrocyte plasma membrane, where MHCII present antigens, is unknown. Rat astrocytes in culture and in organotypic slices were exposed to IFNγ to induce reactive astrogliosis. Astrocytes were probed with optophysiologic tools to investigate subcellular localization of immunolabeled MHCII, and with electrophysiology to characterize interactions of single vesicles with the plasmalemma. In culture and in organotypic slices, IFNγ augmented the astrocytic expression of MHCII, which prominently co-localized with lysosomal marker LAMP1-EGFP, modestly co-localized with Rab7, and did not co-localize with endosomal markers Rab4A, EEA1, and TPC1. MHCII lysosomal localization was corroborated by treatment with the lysosomolytic agent glycyl-L-phenylalanine-β-naphthylamide, which reduced the number of MHCII-positive vesicles. The surface presence of MHCII was revealed by immunolabeling of live non-permeabilized cells. In IFNγ-treated astrocytes, an increased fraction of large-diameter exocytotic vesicles (lysosome-like vesicles) with prolonged fusion pore dwell time and larger pore conductance was recorded, whereas the rate of endocytosis was decreased. Stimulation with ATP, which triggers cytosolic calcium signaling, increased the frequency of exocytotic events, whereas the frequency of full endocytosis was further reduced. In IFNγ-treated astrocytes, MHCII-linked antigen surface presentation is mediated by increased lysosomal exocytosis, whereas surface retention of antigens is prolonged by concomitant inhibition of endocytosis.
Collapse
Affiliation(s)
- Mićo Božić
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia
| | - Alexei Verkhratsky
- Celica Biomedical, Tehnološki park 24, 1000, Ljubljana, Slovenia
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Achucarro Center for Neuroscience, IKERBASQUE, 48011, Bilbao, Spain
| | - Robert Zorec
- Celica Biomedical, Tehnološki park 24, 1000, Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia.
| | - Matjaž Stenovec
- Celica Biomedical, Tehnološki park 24, 1000, Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia.
| |
Collapse
|
14
|
Guerrero-García J. The role of astrocytes in multiple sclerosis pathogenesis. NEUROLOGÍA (ENGLISH EDITION) 2020. [DOI: 10.1016/j.nrleng.2017.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
15
|
Li K, Li J, Zheng J, Qin S. Reactive Astrocytes in Neurodegenerative Diseases. Aging Dis 2019; 10:664-675. [PMID: 31165009 PMCID: PMC6538217 DOI: 10.14336/ad.2018.0720] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/20/2018] [Indexed: 12/17/2022] Open
Abstract
Astrocytes, the largest and most numerous glial cells in the central nervous system (CNS), play a variety of important roles in regulating homeostasis, increasing synaptic plasticity and providing neuroprotection, thus helping to maintain normal brain function. At the same time, astrocytes can participate in the inflammatory response and play a key role in the progression of neurodegenerative diseases. Reactive astrocytes are strongly induced by numerous pathological conditions in the CNS. Astrocyte reactivity is initially characterized by hypertrophy of soma and processes, triggered by different molecules. Recent studies have demonstrated that neuroinflammation and ischemia can elicit two different types of reactive astrocytes, termed A1s and A2s. However, in the case of astrocyte reactivity in different neurodegenerative diseases, the recently published research issues remain a high level of conflict and controversy. So far, we still know very little about whether and how the function or reactivity of astrocytes changes in the progression of different neurodegenerative diseases. In this review, we aimed to briefly discuss recent studies highlighting the complex contribution of astrocytes in the process of various neurodegenerative diseases, which may provide us with new prospects for the development of an excellent therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Kunyu Li
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jiatong Li
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jialin Zheng
- 2Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Song Qin
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Yang Q, Zhou J. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia 2018; 67:1017-1035. [DOI: 10.1002/glia.23571] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Qiao‐qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
| | - Jia‐wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
- University of Chinese Academy of Sciences Shanghai 200031 China
| |
Collapse
|
17
|
Antigen-presenting cell diversity for T cell reactivation in central nervous system autoimmunity. J Mol Med (Berl) 2018; 96:1279-1292. [PMID: 30386908 DOI: 10.1007/s00109-018-1709-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022]
Abstract
Autoreactive T cells are considered the major culprits in the pathogenesis of many autoimmune diseases like multiple sclerosis (MS). Upon activation in the lymphoid organs, autoreactive T cells migrate towards the central nervous system (CNS) and target the myelin sheath-forming oligodendrocytes, resulting in detrimental neurological symptoms. Despite the availability of extensively studied systems like the experimental autoimmune encephalomyelitis (EAE) model, our understanding of this disease and the underlying pathogenesis is still elusive. One vividly discussed subject represents the T cell reactivation in the CNS. In order to exert their effector functions in the CNS, autoreactive T cells must encounter antigen-presenting cells (APCs). This interaction provides an antigen-restricted stimulus in the context of major histocompatibility complex class II (MHC-II) and other co-stimulatory molecules. Peripherally derived dendritic cells (DCs), B cells, border-associated macrophages (BAM), CNS-resident microglia, and astrocytes have the capacity to express molecules required for antigen presentation under inflammatory conditions. Also, endothelial cells can fulfill these prerequisites in certain situations. Which of these cells in fact act as APCs for T cell reactivation and to which extent they can exert this function has been studied intensively, but unfortunately with no firm conclusion. In this review, we will summarize the findings that support or question the antigen presenting capacities of the mentioned cell types of CNS-localized T cell reactivation.
Collapse
|
18
|
Tanabe S, Yamashita T. The role of immune cells in brain development and neurodevelopmental diseases. Int Immunol 2018; 30:437-444. [DOI: 10.1093/intimm/dxy041] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Shogo Tanabe
- Department of Molecular Neuroscience, World Premier International Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, World Premier International Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka, Japan
- Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita-shi, Osaka, Japan
| |
Collapse
|
19
|
Guerrero-García JJ. The role of astrocytes in multiple sclerosis pathogenesis. Neurologia 2017; 35:400-408. [PMID: 28958395 DOI: 10.1016/j.nrl.2017.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 05/31/2017] [Accepted: 07/06/2017] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS), in which astrocytes play an important role as CNS immune cells. However, the activity of astrocytes as antigen-presenting cells (APC) continues to be subject to debate. DEVELOPMENT This review analyses the existing evidence on the participation of astrocytes in CNS inflammation in MS and on several mechanisms that modify astrocyte activity in the disease. CONCLUSIONS Astrocytes play a crucial role in the pathogenesis of MS because they express toll-like receptors (TLR) and major histocompatibility complex (MHC) classI andII. In addition, astrocytes participate in regulating the blood-brain barrier (BBB) and in modulating T cell activity through the production of cytokines. Future studies should focus on the role of astrocytes in order to find new therapeutic targets for the treatment of MS.
Collapse
Affiliation(s)
- J J Guerrero-García
- Doctorado en Ciencias Biomédicas (DCB), CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México; Unidad Médica de Alta Especialidad (UMAE), Hospital de Pediatría (HP), Centro Médico Nacional de Occidente (CMNO), IMSS, Guadalajara, Jalisco, México.
| |
Collapse
|
20
|
Khaibullin T, Ivanova V, Martynova E, Cherepnev G, Khabirov F, Granatov E, Rizvanov A, Khaiboullina S. Elevated Levels of Proinflammatory Cytokines in Cerebrospinal Fluid of Multiple Sclerosis Patients. Front Immunol 2017; 8:531. [PMID: 28572801 PMCID: PMC5435759 DOI: 10.3389/fimmu.2017.00531] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/20/2017] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune neurodegenerative disease characterized by chronic brain inflammation. Leukocyte infiltration of brain tissue causes inflammation, demyelination, and the subsequent formation of sclerotic plaques, which are a hallmark of MS. Activation of proinflammatory cytokines is essential for regulation of lymphocyte migration across the blood–brain barrier. We demonstrate increased levels of many cytokines, including IL-2RA, CCL5, CCL11, MIF, CXCL1, CXCL10, IFNγ, SCF, and TRAIL, were upregulated in cerebrospinal fluid (CSF), whereas IL-17, CCL2, CCL3, CCL4, and IL-12(p40) were activated in MS serum. Interaction analysis of cytokines in CSF demonstrated a connection between IFNγ and CCL5 as well as MIF. Many cells can contribute to production of these cytokines including CD8 and Th1 lymphocytes and astrocytes. Therefore, we suggest that IFNγ released by Th1 lymphocytes can activate astrocytes, which then produce chemoattractants, including CCL5 and MIF. These chemokines promote an inflammatory milieu and interact with multiple chemokines including CCL27 and CXCL1. Of special note, upregulation of CCL27 was found in CSF of MS cases. This observation is the first to demonstrate CCL27 as a potential contributor of brain pathology in MS. Our data suggest that CCL27 may be involved in activation and migration of autoreactive encephalitogenic immune effectors in the brain. Further, our data support the role of Th1 lymphocytes in the pathogenesis of brain inflammation in MS, with several cytokines playing a central role.
Collapse
Affiliation(s)
| | - Vilena Ivanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Georgy Cherepnev
- University Kazan Clinic, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | | | | | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation.,Nevada Center for Biomedical Research, Reno, NV, USA
| |
Collapse
|
21
|
Freeman L, Guo H, David CN, Brickey WJ, Jha S, Ting JPY. NLR members NLRC4 and NLRP3 mediate sterile inflammasome activation in microglia and astrocytes. J Exp Med 2017; 214:1351-1370. [PMID: 28404595 PMCID: PMC5413320 DOI: 10.1084/jem.20150237] [Citation(s) in RCA: 296] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 12/26/2016] [Accepted: 02/28/2017] [Indexed: 12/31/2022] Open
Abstract
Lysophosphatidylcholine is associated with neurodegeneration and demyelination. Freeman et al. demonstrate that lysophosphatidylcholine triggers NLRP3- and NLRC4-dependent inflammasome activation, and in a synergistic fashion, NLRP3 and NLRC4 contribute to a cuprizone-induced demyelination model in vivo. Inflammation in the brain accompanies several high-impact neurological diseases including multiple sclerosis (MS), stroke, and Alzheimer’s disease. Neuroinflammation is sterile, as damage-associated molecular patterns rather than microbial pathogens elicit the response. The inflammasome, which leads to caspase-1 activation, is implicated in neuroinflammation. In this study, we reveal that lysophosphatidylcholine (LPC), a molecule associated with neurodegeneration and demyelination, elicits NLRP3 and NLRC4 inflammasome activation in microglia and astrocytes, which are central players in neuroinflammation. LPC-activated inflammasome also requires ASC (apoptotic speck containing protein with a CARD), caspase-1, cathepsin-mediated degradation, calcium mobilization, and potassium efflux but not caspase-11. To study the physiological relevance, Nlrc4−/− and Nlrp3−/− mice are studied in the cuprizone model of neuroinflammation and demyelination. Mice lacking both genes show the most pronounced reduction in astrogliosis and microglial accumulation accompanied by decreased expression of the LPC receptor G2A, whereas MS patient samples show increased G2A. These results reveal that NLRC4 and NLRP3, which normally form distinct inflammasomes, activate an LPC-induced inflammasome and are important in astrogliosis and microgliosis.
Collapse
Affiliation(s)
- Leslie Freeman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Haitao Guo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Clément N David
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - W June Brickey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Sushmita Jha
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 .,Indian Institute of Technology Jodhpur, Rajasthan 342011, India
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 .,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Microbiology and Immunology, Institute of Inflammatory Diseases, Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
22
|
Olave MC, Vargas-Zambrano JC, Celis AM, Castañeda E, González JM. Infective capacity of Cryptococcus neoformans and Cryptococcus gattii in a human astrocytoma cell line. Mycoses 2017; 60:447-453. [PMID: 28338245 DOI: 10.1111/myc.12619] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 12/21/2022]
Abstract
Pathogenesis of cryptococcosis in the central nervous system (CNS) is a topic of ongoing research, including the mechanisms by which this fungus invades and infects the brain. Astrocytes, the most common CNS cells, play a fundamental role in the local immune response. Astrocytes might participate in cryptococcosis either as a host or by responding to fungal antigens. To determine the infectivity of Cryptococcus neoformans var. grubii and Cryptococcus gattii in a human astrocytoma cell line and the induction of major histocompatibility complex (MHC) molecules. A glioblastoma cell line was infected with C. neoformans var. grubii and C. gattii blastoconidia labelled with FUN-1 fluorescent stain. The percentage of infection and expression of HLA class I and II molecules were determined by flow cytometry. The interactions between the fungi and cells were observed by fluorescence microscopy. There was no difference between C. neoformans var. grubii and C. gattii in the percentage infection, but C. neoformans var. grubii induced higher expression of HLA class II than C. gattii. More blastoconidia were recovered from C. neoformans-infected cells than from C. gattii infected cells. Cryptococcus neoformans var. grubii may have different virulence mechanisms that allow its survival in human glia-derived cells.
Collapse
Affiliation(s)
- M C Olave
- Grupo Ciencias Básicas Médicas, Facultad de Medicina, Universidad de los Andes, Bogotá DC, Colombia
| | - J C Vargas-Zambrano
- Grupo Ciencias Básicas Médicas, Facultad de Medicina, Universidad de los Andes, Bogotá DC, Colombia
| | - A M Celis
- Mycology and Phytopathology Laboratory, Department of Biological Sciences, Universidad de los Andes, Bogotá DC, Colombia
| | - E Castañeda
- Grupo de Micología, Instituto Nacional de Salud, Bogotá DC, Colombia
| | - J M González
- Grupo Ciencias Básicas Médicas, Facultad de Medicina, Universidad de los Andes, Bogotá DC, Colombia
| |
Collapse
|
23
|
Gilbert R, Al-Janabi A, Tomkins-Netzer O, Lightman S. Statins as anti-inflammatory agents: A potential therapeutic role in sight-threatening non-infectious uveitis. Porto Biomed J 2017; 2:33-39. [PMID: 32258583 PMCID: PMC6806973 DOI: 10.1016/j.pbj.2017.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/05/2017] [Indexed: 12/29/2022] Open
Abstract
In addition to the known lipid-lowering effects, statins are now widely accepted to have anti-inflammatory and immunomodulatory effects. Adjunctive use of statins has proven beneficial in the context of a wide range of inflammatory diseases, including rheumatoid arthritis. Evidence also suggests that statins may also have utility in the management of uveitis, a form of sight threatening inflammation which occurs in the eye. In this article, we outline our rationale behind a clinical trial of simvastatin as a steroid-sparing agent in uveitis, to which patient recruitment started last year. Potential risks associated with the clinical use of statins, including putative effects on the eyes, are discussed.
Collapse
Affiliation(s)
- Rose Gilbert
- Moorfields Eye Hospital, London, UK
- University College London (UCL) Institute of Ophthalmology, London, UK
| | - Ahmed Al-Janabi
- Moorfields Eye Hospital, London, UK
- University College London (UCL) Institute of Ophthalmology, London, UK
| | - Oren Tomkins-Netzer
- Moorfields Eye Hospital, London, UK
- University College London (UCL) Institute of Ophthalmology, London, UK
| | - Sue Lightman
- Moorfields Eye Hospital, London, UK
- University College London (UCL) Institute of Ophthalmology, London, UK
| |
Collapse
|
24
|
Cekanaviciute E, Buckwalter MS. Astrocytes: Integrative Regulators of Neuroinflammation in Stroke and Other Neurological Diseases. Neurotherapeutics 2016; 13:685-701. [PMID: 27677607 PMCID: PMC5081110 DOI: 10.1007/s13311-016-0477-8] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Astrocytes regulate neuroinflammatory responses after stroke and in other neurological diseases. Although not all astrocytic responses reduce inflammation, their predominant function is to protect the brain by driving the system back to homeostasis after injury. They receive multidimensional signals within the central nervous system and between the brain and the systemic circulation. Processing this information allows astrocytes to regulate synapse formation and maintenance, cerebral blood flow, and blood-brain barrier integrity. Similarly, in response to stroke and other central nervous system disorders, astrocytes detect and integrate signals of neuronal damage and inflammation to regulate the neuroinflammatory response. Two direct regulatory mechanisms in the astrocyte arsenal are the ability to form both physical and molecular barriers that seal the injury site and localize the neuroinflammatory response. Astrocytes also indirectly regulate the inflammatory response by affecting neuronal health during the acute injury and axonal regrowth. This ability to regulate the location and degree of neuroinflammation after injury, combined with the long time course of neuroinflammation, makes astrocytic signaling pathways promising targets for therapies.
Collapse
Affiliation(s)
- Egle Cekanaviciute
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, 94305, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, 94305, USA.
- Department of Neurosurgery, Stanford Medical School, Stanford, CA, 94305, USA.
- Stanford Stroke Center, Stanford Medical School, Stanford, CA, 94305, USA.
| |
Collapse
|
25
|
Sphingosine 1-phosphate signaling in astrocytes: Implications for progressive multiple sclerosis. J Neurol Sci 2015; 361:60-5. [PMID: 26810518 DOI: 10.1016/j.jns.2015.12.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/26/2015] [Accepted: 12/14/2015] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis is an autoimmune disorder characterized by recurrent attacks against the central nervous system. After many years, certain patients enter a progressive disease phase, characterized by steady clinical deterioration. However, in 10-15% of patients, the disease is progressive from the beginning, and thus diagnosed as Primary Progressive Multiple Sclerosis. Unlike relapsing-remitting forms, progressive MS lacks effective therapy. Astrocytes are a major component of glial cells and are now thought to play a role in disease progression. Sphingosine 1-phophate is a molecule with extensive receptor expression on both immune and glial cells and is also a target of fingolimod, a drug used in relapsing remitting patients that sequesters lymphocytes within lymph nodes. However, because sphingosine 1-phosphate receptors are also expressed in astrocytes, and also because modification of this pathway has shown interesting benefits in animal models of Multiple Sclerosis, this astrocyte pathway has become an interesting target for developing potential new therapeutic approaches for Multiple Sclerosis.
Collapse
|
26
|
Ludewig P, Gallizioli M, Urra X, Behr S, Brait VH, Gelderblom M, Magnus T, Planas AM. Dendritic cells in brain diseases. Biochim Biophys Acta Mol Basis Dis 2015; 1862:352-67. [PMID: 26569432 DOI: 10.1016/j.bbadis.2015.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mattia Gallizioli
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic, Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sarah Behr
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa H Brait
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
27
|
Sosa RA, Murphey C, Robinson RR, Forsthuber TG. IFN-γ ameliorates autoimmune encephalomyelitis by limiting myelin lipid peroxidation. Proc Natl Acad Sci U S A 2015; 112:E5038-47. [PMID: 26305941 PMCID: PMC4568689 DOI: 10.1073/pnas.1505955112] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Evidence has suggested both a pathogenic and a protective role for the proinflammatory cytokine IFN-γ in experimental autoimmune encephalomyelitis (EAE). However, the mechanisms underlying the protective role of IFN-γ in EAE have not been fully resolved, particularly in the context of CNS antigen-presenting cells (APCs). In this study we examined the role of IFN-γ in myelin antigen uptake by CNS APCs during EAE. We found that myelin antigen colocalization with APCs was decreased substantially and that EAE was significantly more severe and showed a chronic-progressive course in IFN-γ knockout (IFN-γ-/-) or IFN-γ receptor knockout (IFN-γR-/-) mice as compared with WT animals. IFN-γ was a critical regulator of phagocytic/activating receptors on CNS APCs. Importantly, "free" myelin debris and lipid peroxidation activity at CNS lesions was increased in mice lacking IFN-γ signaling. Treatment with N-acetyl-l-cysteine, a potent antioxidant, abolished lipid peroxidation activity and ameliorated EAE in IFN-γ-signaling-deficient mice. Taken together the data suggest a protective role for IFN-γ in EAE by regulating the removal of myelin debris by CNS APCs and thereby limiting the substrate available for the generation of neurotoxic lipid peroxidation products.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Apoptosis/genetics
- Apoptosis/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Flow Cytometry
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Lipid Peroxidation/immunology
- Lymphocyte Activation/immunology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myelin Sheath/immunology
- Myelin Sheath/metabolism
- Phagocytosis/genetics
- Phagocytosis/immunology
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Interferon/immunology
- Signal Transduction/genetics
- Signal Transduction/immunology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Rebecca A Sosa
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249
| | - Cathi Murphey
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249
| |
Collapse
|
28
|
Hussain RZ, Hayardeny L, Cravens PC, Yarovinsky F, Eagar TN, Arellano B, Deason K, Castro-Rojas C, Stüve O. Immune surveillance of the central nervous system in multiple sclerosis--relevance for therapy and experimental models. J Neuroimmunol 2014; 276:9-17. [PMID: 25282087 PMCID: PMC4301841 DOI: 10.1016/j.jneuroim.2014.08.622] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/15/2014] [Accepted: 08/20/2014] [Indexed: 12/25/2022]
Abstract
Treatment of central nervous system (CNS) autoimmune disorders frequently involves the reduction, or depletion of immune-competent cells. Alternatively, immune cells are being sequestered away from the target organ by interfering with their movement from secondary lymphoid organs, or their migration into tissues. These therapeutic strategies have been successful in multiple sclerosis (MS), the most prevalent autoimmune inflammatory disorder of the CNS. However, many of the agents that are currently approved or in clinical development also have severe potential adverse effects that stem from the very mechanisms that mediate their beneficial effects by interfering with CNS immune surveillance. This review will outline the main cellular components of the innate and adaptive immune system that participate in host defense and maintain immune surveillance of the CNS. Their pathogenic role in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also discussed. Furthermore, an experimental model is introduced that may assist in evaluating the effect of therapeutic interventions on leukocyte homeostasis and function within the CNS. This model or similar models may become a useful tool in the repertoire of pre-clinical tests of pharmacological agents to better explore their potential for adverse events.
Collapse
Affiliation(s)
- Rehana Z Hussain
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | | | - Petra C Cravens
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Felix Yarovinsky
- Department of Immunology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Todd N Eagar
- Histocompatibility and Transplant Immunology, Department of Pathology and Genomic Medicine, The Methodist Hospital Physician Organization, Houston, TX, USA
| | - Benjamine Arellano
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Krystin Deason
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Cyd Castro-Rojas
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany.
| |
Collapse
|
29
|
Duraes FV, Thelemann C, Sarter K, Acha-Orbea H, Hugues S, Reith W. Role of major histocompatibility complex class II expression by non-hematopoietic cells in autoimmune and inflammatory disorders: facts and fiction. ACTA ACUST UNITED AC 2014; 82:1-15. [PMID: 23745569 DOI: 10.1111/tan.12136] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is well established that interactions between CD4(+) T cells and major histocompatibility complex class II (MHCII) positive antigen-presenting cells (APCs) of hematopoietic origin play key roles in both the maintenance of tolerance and the initiation and development of autoimmune and inflammatory disorders. In sharp contrast, despite nearly three decades of intensive research, the functional relevance of MHCII expression by non-hematopoietic tissue-resident cells has remained obscure. The widespread assumption that MHCII expression by non-hematopoietic APCs has an impact on autoimmune and inflammatory diseases has in most instances neither been confirmed nor excluded by indisputable in vivo data. Here we review and put into perspective conflicting in vitro and in vivo results on the putative impact of MHCII expression by non-hematopoietic APCs--in both target organs and secondary lymphoid tissues--on the initiation and development of representative autoimmune and inflammatory disorders. Emphasis will be placed on the lacunar status of our knowledge in this field. We also discuss new mouse models--developed on the basis of our understanding of the molecular mechanisms that regulate MHCII expression--that constitute valuable tools for filling the severe gaps in our knowledge on the functions of non-hematopoietic APCs in inflammatory conditions.
Collapse
Affiliation(s)
- F V Duraes
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
30
|
Sosa RA, Murphey C, Ji N, Cardona AE, Forsthuber TG. The kinetics of myelin antigen uptake by myeloid cells in the central nervous system during experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 191:5848-57. [PMID: 24227784 DOI: 10.4049/jimmunol.1300771] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Induction of experimental autoimmune encephalomyelitis (EAE) in susceptible animals requires reactivation of encephalitogenic CD4(+) T cells by APCs in the CNS. However, it has remained unresolved from where APCs in the CNS acquire myelin Ag for T cell activation and under which conditions, that is, whether only during EAE or also in the naive CNS. In this study, we investigated the kinetics of myelin Ag uptake by CNS APCs during EAE and in the naive CNS. Our results show that during EAE CX3CR1(+)CD11b(+) microglia were the first APCs in the CNS to contain myelin Ag upon induction of disease, albeit in very small numbers. Dendritic cells (DCs) arrived in the CNS in sizable numbers significantly later (day 5 postimmunization), without detectable myelin Ag, but acquired it by day 7 postimmunization. Furthermore, a sharp increase in neuroantigen-containing DCs coincided with the onset of EAE symptoms. Importantly, in naive mice a low but consistent number of microglia contained myelin Ag, suggesting release by oligodendrocytes under steady state conditions. Although microglia isolated from naive brain and spinal cord did not elicit a strong CD4(+) T cell response in vitro, myelin Ag-containing microglia may still play a local role in modulating encephalitogenic CD4(+) T cell responses in early EAE prior to the arrival of other professional APCs, such as DCs. Finally, newly arriving DCs in the CNS not yet loaded with myelin Ag before the onset of EAE may be a potential therapeutic target.
Collapse
Affiliation(s)
- Rebecca A Sosa
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249
| | | | | | | | | |
Collapse
|
31
|
Astrocyte regulation of CNS inflammation and remyelination. Brain Sci 2013; 3:1109-27. [PMID: 24961523 PMCID: PMC4061872 DOI: 10.3390/brainsci3031109] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/12/2013] [Accepted: 07/12/2013] [Indexed: 01/17/2023] Open
Abstract
Astrocytes regulate fundamentally important functions to maintain central nervous system (CNS) homeostasis. Altered astrocytic function is now recognized as a primary contributing factor to an increasing number of neurological diseases. In this review, we provide an overview of our rapidly developing understanding of the basal and inflammatory functions of astrocytes as mediators of CNS responsiveness to inflammation and injury. Specifically, we elaborate on ways that astrocytes actively participate in the pathogenesis of demyelinating diseases of the CNS through their immunomodulatory roles as CNS antigen presenting cells, modulators of blood brain barrier function and as a source of chemokines and cytokines. We also outline how changes in the extracellular matrix can modulate astrocytes phenotypically, resulting in dysregulation of astrocytic responses during inflammatory injury. We also relate recent studies describing newly identified roles for astrocytes in leukodystrophies. Finally, we describe recent advances in how adapting this increasing breadth of knowledge on astrocytes has fostered new ways of thinking about human diseases, which offer potential to modulate astrocytic heterogeneity and plasticity towards therapeutic gain. In summary, recent studies have provided improved insight in a wide variety of neuroinflammatory and demyelinating diseases, and future research on astrocyte pathophysiology is expected to provide new perspectives on these diseases, for which new treatment modalities are increasingly necessary.
Collapse
|
32
|
Hertzenberg D, Lehmann-Horn K, Kinzel S, Husterer V, Cravens PD, Kieseier BC, Hemmer B, Brück W, Zamvil SS, Stüve O, Weber MS. Developmental maturation of innate immune cell function correlates with susceptibility to central nervous system autoimmunity. Eur J Immunol 2013; 43:2078-88. [PMID: 23637087 DOI: 10.1002/eji.201343338] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 03/28/2013] [Accepted: 04/25/2013] [Indexed: 11/08/2022]
Abstract
MS is an inflammatory CNS disorder, which typically occurs in early adulthood and rarely in children. Here we tested whether functional maturation of innate immune cells may determine susceptibility to CNS autoimmune disease in EAE. Two-week-old mice were resistant to active EAE, which causes fulminant paralysis in adult mice; this resistance was associated with an impaired development of Th1 and Th17 cells. Resistant, young mice had higher frequencies of myeloid-derived suppressor cells and plasma-cytoid DCs. Furthermore, myeloid APCs and B cells from young mice expressed lower levels of MHC class II and CD40, produced decreased amounts of proinflammatory cytokines, and released enhanced levels of anti-inflammatory IL-10. When used as APCs, splenocytes from 2-week-old mice failed to differentiate naive T cells into Th1 and Th17 cells irrespective of the T-cell donor's age, and promoted development of Treg cells and Th2 cells instead. Adoptive transfer of adult APCs restored the ability of 2-week-old mice to generate encephalitogenic T cells and develop EAE. Collectively, these findings indicate that the innate immune compartment functionally matures during development, which may be a prerequisite for development of T-cell-mediated CNS autoimmune disease.
Collapse
Affiliation(s)
- Deetje Hertzenberg
- Department of Neurology, Technische Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cravens PD, Kieseier BC, Hussain R, Herndon E, Arellano B, Ben LH, Timmons BC, Castro-Rojas C, Hartung HP, Hemmer B, Weber MS, Zamvil SS, Stüve O. The neonatal CNS is not conducive for encephalitogenic Th1 T cells and B cells during experimental autoimmune encephalomyelitis. J Neuroinflammation 2013; 10:67. [PMID: 23705890 PMCID: PMC3679999 DOI: 10.1186/1742-2094-10-67] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/06/2013] [Indexed: 01/07/2023] Open
Abstract
Multiple sclerosis (MS) is thought to be a CD4+ T cell mediated autoimmune demyelinating disease of the central nervous system (CNS) that is rarely diagnosed during infancy. Cellular and molecular mechanisms that confer disease resistance in this age group are unknown. We tested the hypothesis that a differential composition of immune cells within the CNS modulates age-associated susceptibility to CNS autoimmune disease. C57BL/6 mice younger than eight weeks were resistant to experimental autoimmune encephalomyelitis (EAE) following active immunization with myelin oligodendrocyte glycoprotein (MOG) peptide (p) 35-55. Neonates also developed milder EAE after transfer of adult encephalitogenic T cells primed by adult or neonate antigen presenting cells (APC). There was a significant increase in CD45+ hematopoietic immune cells and CD45+ high side scatter granulocytes in the CNS of adults, but not in neonates. Within the CD45+ immune cell compartment of adults, the accumulation of CD4+ T cells, Gr-1+ and Gr-1- monocytes and CD11c+ dendritic cells (DC) was identified. A significantly greater percentage of CD19+ B cells in the adult CNS expressed MHC II than neonate CNS B cells. Only in the adult CNS could IFNγ transcripts be detected 10 days post immunization for EAE. IFNγ is highly expressed by adult donor CD4+ T cells that are adoptively transferred but not by transferred neonate donor cells. In contrast, IL-17 transcripts could not be detected in adult or neonate CNS in this EAE model, and neither adult nor neonate donor CD4+ T cells expressed IL-17 at the time of adoptive transfer.
Collapse
Affiliation(s)
- Petra D Cravens
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9036, USA
| | - Bernd C Kieseier
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | - Rehana Hussain
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9036, USA
| | - Emily Herndon
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Benjamine Arellano
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9036, USA
| | - Li-Hong Ben
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9036, USA
| | - Brenda C Timmons
- Hamon Center for Therapeutic Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Cyd Castro-Rojas
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9036, USA
| | - Hans-Peter Hartung
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, München, 81675, Germany
| | - Martin S Weber
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, München, 81675, Germany
- Department of Neurology, University Medical Center, Georg August University, Göttingen, 37075, Germany
- Department of Neuropathology, University Medical Center, Georg August University, Göttingen, 37975, Germany
| | - Scott S Zamvil
- Department of Neurology, University of California, San Francisco, CA, 94143, USA
- Program in Immunology, University of California, San Francisco, CA, 94143, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9036, USA
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, München, 81675, Germany
- Neurology Section, VA North Texas Health Care System, Medical Service, 4500 South Lancaster Rd, Dallas, TX, 75216, USA
| |
Collapse
|
34
|
Lehmann-Horn K, Kronsbein HC, Weber MS. Targeting B cells in the treatment of multiple sclerosis: recent advances and remaining challenges. Ther Adv Neurol Disord 2013; 6:161-73. [PMID: 23634189 DOI: 10.1177/1756285612474333] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Recent years have substantially broadened our view on the pathogenesis of multiple sclerosis (MS). While earlier concepts focused predominantly on T lymphocytes as the key cell type to mediate inflammatory damage within central nervous system (CNS) lesions, emerging evidence suggests that B lymphocytes may play a comparably important role both as precursors of antibody-secreting plasma cells and as antigen-presenting cells (APCs) for the activation of T cells. With greater appreciation of this pathogenic B-cell function in MS, B-cell-directed therapies, and in particular B-cell-depleting monoclonal antibodies targeting the CD20 molecule, have gained enormous interest over recent years. Clinical trials demonstrated that anti-CD20 treatment, which depletes immature and mature B cells but spares CD20 negative plasma cells, rapidly reduces formation of new inflammatory CNS lesions. While these findings clearly corroborate a pathogenic contribution of B cells, recent experimental but also clinical findings indicate that not all B cells contribute in an equally pathogenic manner and that certain subsets may in contrast mediate anti-inflammatory effects. In this review, we summarize current findings in support of pathogenic B-cell function in MS, including the encouraging clinical data which derived from anti-CD20 MS trials. Further, we review novel findings suggestive of regulatory properties of B-cell subsets which may be collaterally abolished by pan-CD20 depletion. In conclusion, we aim to provide an outlook on how this currently differentiating concept of pro- and anti-inflammatory B-cell function could be harnessed to further improve safety and effectiveness of B-cell-directed therapeutic approaches in MS.
Collapse
Affiliation(s)
- Klaus Lehmann-Horn
- Department of Neurology, Technische Universität München, Munich, Germany
| | | | | |
Collapse
|
35
|
Fagone P, Mangano K, Coco M, Perciavalle V, Garotta G, Romao CC, Nicoletti F. Therapeutic potential of carbon monoxide in multiple sclerosis. Clin Exp Immunol 2012; 167:179-87. [PMID: 22235993 DOI: 10.1111/j.1365-2249.2011.04491.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Carbon monoxide (CO) is produced during the catabolism of free haem, catalyzed by haem oxygenase (HO) enzymes, and its physiological roles include vasodilation, neurotransmission, inhibition of platelet aggregation and anti-proliferative effects on smooth muscle. In vivo preclinical studies have shown that exogenously administered quantities of CO may represent an effective treatment for conditions characterized by a dysregulated immune response. The carbon monoxide-releasing molecules (CORMs) represent a group of compounds capable of carrying and liberating controlled quantities of CO in the cellular systems. This review covers the physiological and anti-inflammatory properties of the HO/CO pathway in the central nervous system. It also discusses the effects of CORMs in preclinical models of inflammation. The accumulating data discussed herein support the possibility that CORMs may represent a novel class of drugs with disease-modifying properties in multiple sclerosis.
Collapse
Affiliation(s)
- P Fagone
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Cravens PD, Hussain RZ, Zacharias TE, Ben LH, Herndon E, Vinnakota R, Lambracht-Washington D, Nessler S, Zamvil SS, Eagar TN, Stüve O. Lymph node-derived donor encephalitogenic CD4+ T cells in C57BL/6 mice adoptive transfer experimental autoimmune encephalomyelitis highly express GM-CSF and T-bet. J Neuroinflammation 2011; 8:73. [PMID: 21702922 PMCID: PMC3161869 DOI: 10.1186/1742-2094-8-73] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 06/24/2011] [Indexed: 01/20/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a relevant animal model for the human demyelinating inflammatory disorder of the central nervous system (CNS), multiple sclerosis (MS). Induction of EAE by adoptive transfer allows studying the role of the donor T lymphocyte in disease pathogenesis. It has been challenging to reliably induce adoptive transfer EAE in C57BL/6 (H-2b) mice. The goal of this study was to develop a reproducible and high yield protocol for adoptive transfer EAE in C57BL/6 mice. A step-wise experimental approach permitted us to develop a protocol that resulted in a consistent relatively high disease incidence of ~70% in recipient mice. Donor mice were immunized with myelin oligodendrocyte glycoprotein (MOG)p35-55 in complete Freund's adjuvant (CFA) followed by pertussis toxin (PT). Only lymph node cells (LNC) isolated at day 12 post immunization, and restimulated in vitro for 72 hours with 10 μg/mL of MOGp35-55 and 0.5 ng/mL of interleukin-12 (IL-12) were able to transfer disease. The ability of LNC to transfer disease was associated with the presence of inflammatory infiltrates in the CNS at day 12. Interferon gamma (IFNγ) was produced at comparable levels in cell cultures prepared from mice at both day 6 and day 12 post immunization. By contrast, there was a trend towards a negative association between IL-17 and disease susceptibility in our EAE model. The amount of GM-CSF secreted was significantly increased in the culture supernatants from cells collected at day 12 post immunization versus those collected at day 6 post-immunization. Activated CD4+ T cells present in the day 12 LNC cultures maintained expression of the transcription factor T-bet, which has been shown to regulate the expression of the IL-23 receptor. Also, there was an increased prevalence of MOGp35-55-specific CD4+ T cells in day 12 LNC after in vitro re-stimulation. In summary, encephalitogenic LNC that adoptively transfer EAE in C57BL/6 mice were not characterized by a single biomarker in our study, but by a composite of inflammatory markers. Our data further suggest that GM-CSF expression by CD4+ T cells regulated by IL-23 contributes to their encephalitogenicity in our EAE model.
Collapse
Affiliation(s)
- Petra D Cravens
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Moliné-Velázquez V, Cuervo H, Vila-Del Sol V, Ortega MC, Clemente D, de Castro F. Myeloid-derived suppressor cells limit the inflammation by promoting T lymphocyte apoptosis in the spinal cord of a murine model of multiple sclerosis. Brain Pathol 2011; 21:678-91. [PMID: 21507122 DOI: 10.1111/j.1750-3639.2011.00495.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multiple Sclerosis (MS) is a demyelinating/inflammatory disease of the central nervous system. Relapsing-remitting MS is characterized by a relapsing phase with clinical symptoms and the production of inflammatory cell infiltrates, and a period of remission during which patients recover partially. Myeloid-derived suppressor cells (MDSCs) are immature cells capable of suppressing the inflammatory response through Arginase-I (Arg-I) activity, among other mechanisms. Here, we have identified Arg-I(+) -MDSCs in the spinal cord during experimental autoimmune encephalomyelitis (EAE), cells that were largely restricted to the demyelinating plaque and that always exhibited the characteristic MDSC surface markers Arg-I/CD11b/Gr-1/M-CSF1R. The presence and density of Arg-I(+) -cells, and the proportion of apoptotic but not proliferative T cells, were correlated with the EAE time course: peaked in parallel with the clinical score, decreased significantly during the remitting phase and completely disappeared during the chronic phase. Spinal cord-isolated MDSCs of EAE animals augmented the cell death when co-cultured with stimulated control splenic CD3 T cells. These data point to an important role for MDSCs in limiting inflammatory damage in MS, favoring the relative recovery in the remitting phase of the disease. Thus, the MDSC population should be considered as a potential therapeutic target to accelerate the recovery of MS patients.
Collapse
Affiliation(s)
- Verónica Moliné-Velázquez
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda" s/n, Toledo, Spain
| | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Cripps JG, Gorham JD. MDSC in autoimmunity. Int Immunopharmacol 2011; 11:789-93. [PMID: 21310255 DOI: 10.1016/j.intimp.2011.01.026] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 01/22/2011] [Accepted: 01/24/2011] [Indexed: 12/22/2022]
Abstract
Myeloid derived suppressor cells (MDSC) were first described nearly two decades ago. Until recently, however, descriptions of MDSC populations were found almost exclusively in animal models of cancer or in cancer patients. Over the last few years, an increasing number of reports have been published describing populations of myeloid cells with MDSC-like properties in murine models of autoimmune disease. In contrast to the proposed deleterious role of MDSC in cancer--where these cells likely inhibit tumor immunity--in the context of autoimmunity, MDSC have the potential to suppress the autoimmune response, thereby limiting tissue injury. A logical corollary of this hypothesis is that a failure of endogenous MDSC to appropriately control autoimmune T cell responses in vivo may actually contribute to the pathogenesis of autoimmune disease.
Collapse
Affiliation(s)
- James G Cripps
- Department of Microbiology & Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | | |
Collapse
|
40
|
Nelson PA, Khodadoust M, Prodhomme T, Spencer C, Patarroyo JC, Varrin-Doyer M, Ho JD, Stroud RM, Zamvil SS. Immunodominant T cell determinants of aquaporin-4, the autoantigen associated with neuromyelitis optica. PLoS One 2010; 5:e15050. [PMID: 21151500 PMCID: PMC2994828 DOI: 10.1371/journal.pone.0015050] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 10/14/2010] [Indexed: 01/02/2023] Open
Abstract
Autoantibodies that target the water channel aquaporin-4 (AQP4) in neuromyelitis optica (NMO) are IgG1, a T cell-dependent Ig subclass. However, a role for AQP4-specific T cells in this CNS inflammatory disease is not known. To evaluate their potential role in CNS autoimmunity, we have identified and characterized T cells that respond to AQP4 in C57BL/6 and SJL/J mice, two strains that are commonly studied in models of CNS inflammatory diseases. Mice were immunized with either overlapping peptides or intact hAQP4 protein encompassing the entire 323 amino acid sequence. T cell determinants identified from examination of the AQP4 peptide (p) library were located within AQP4 p21-40, p91-110, p101-120, p166-180, p231-250 and p261-280 in C57BL/6 mice, and within p11-30, p21-40, p101-120, p126-140 and p261-280 in SJL/J mice. AQP4-specific T cells were CD4+ and MHC II-restricted. In recall responses to immunization with intact AQP4, T cells responded primarily to p21-40, indicating this region contains the immunodominant T cell epitope(s) for both strains. AQP4 p21-40-primed T cells secreted both IFN-γ and IL-17. The core immunodominant AQP4 21-40 T cell determinant was mapped to residues 24-35 in C57BL/6 mice and 23-35 in SJL/J mice. Our identification of the AQP4 T cell determinants and characterization of its immunodominant determinant should permit investigators to evaluate the role of AQP4-specific T cells in vivo and to develop AQP4-targeted murine NMO models.
Collapse
Affiliation(s)
- Patricia A. Nelson
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Mojgan Khodadoust
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Thomas Prodhomme
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Collin Spencer
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Juan Carlos Patarroyo
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Michel Varrin-Doyer
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Joseph D. Ho
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Robert M. Stroud
- Department of Biochemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Scott S. Zamvil
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Tufekci KU, Oner MG, Genc S, Genc K. MicroRNAs and Multiple Sclerosis. Autoimmune Dis 2010; 2011:807426. [PMID: 21188194 PMCID: PMC3003960 DOI: 10.4061/2011/807426] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 10/16/2010] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) have recently emerged as a new class of modulators of gene expression. miRNAs control protein synthesis by targeting mRNAs for translational repression or degradation at the posttranscriptional level. These noncoding RNAs are endogenous, single-stranded molecules approximately 22 nucleotides in length and have roles in multiple facets of immunity, from regulation of development of key cellular players to activation and function in immune responses. Recent studies have shown that dysregulation of miRNAs involved in immune responses leads to autoimmunity. Multiple sclerosis (MS) serves as an example of a chronic and organ-specific autoimmune disease in which miRNAs modulate immune responses in the peripheral immune compartment and the neuroinflammatory process in the brain. For MS, miRNAs have the potential to serve as modifying drugs. In this review, we summarize current knowledge of miRNA biogenesis and mode of action and the diverse roles of miRNAs in modulating the immune and inflammatory responses. We also review the role of miRNAs in autoimmunity, focusing on emerging data regarding miRNA expression patterns in MS. Finally, we discuss the potential of miRNAs as a disease marker and a novel therapeutic target in MS. Better understanding of the role of miRNAs in MS will improve our knowledge of the pathogenesis of this disease.
Collapse
Affiliation(s)
- Kemal Ugur Tufekci
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | | | | | | |
Collapse
|
42
|
Slavin A, Kelly-Modis L, Labadia M, Ryan K, Brown ML. Pathogenic mechanisms and experimental models of multiple sclerosis. Autoimmunity 2010; 43:504-13. [DOI: 10.3109/08916931003674733] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
43
|
Hu W, Nessler S, Hemmer B, Eagar TN, Kane LP, Leliveld SR, Müller-Schiffmann A, Gocke AR, Lovett-Racke A, Ben LH, Hussain RZ, Breil A, Elliott JL, Puttaparthi K, Cravens PD, Singh MP, Petsch B, Stitz L, Racke MK, Korth C, Stüve O. Pharmacological prion protein silencing accelerates central nervous system autoimmune disease via T cell receptor signalling. ACTA ACUST UNITED AC 2010; 133:375-88. [PMID: 20145049 PMCID: PMC2822628 DOI: 10.1093/brain/awp298] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The primary biological function of the endogenous cellular prion protein has remained unclear. We investigated its biological function in the generation of cellular immune responses using cellular prion protein gene-specific small interfering ribonucleic acid in vivo and in vitro. Our results were confirmed by blocking cellular prion protein with monovalent antibodies and by using cellular prion protein-deficient and -transgenic mice. In vivo prion protein gene-small interfering ribonucleic acid treatment effects were of limited duration, restricted to secondary lymphoid organs and resulted in a 70% reduction of cellular prion protein expression in leukocytes. Disruption of cellular prion protein signalling augmented antigen-specific activation and proliferation, and enhanced T cell receptor signalling, resulting in zeta-chain-associated protein-70 phosphorylation and nuclear factor of activated T cells/activator protein 1 transcriptional activity. In vivo prion protein gene-small interfering ribonucleic acid treatment promoted T cell differentiation towards pro-inflammatory phenotypes and increased survival of antigen-specific T cells. Cellular prion protein silencing with small interfering ribonucleic acid also resulted in the worsening of actively induced and adoptively transferred experimental autoimmune encephalomyelitis. Finally, treatment of myelin basic protein1–11 T cell receptor transgenic mice with prion protein gene-small interfering ribonucleic acid resulted in spontaneous experimental autoimmune encephalomyelitis. Thus, central nervous system autoimmune disease was modulated at all stages of disease: the generation of the T cell effector response, the elicitation of T effector function and the perpetuation of cellular immune responses. Our findings indicate that cellular prion protein regulates T cell receptor-mediated T cell activation, differentiation and survival. Defects in autoimmunity are restricted to the immune system and not the central nervous system. Our data identify cellular prion protein as a regulator of cellular immunological homoeostasis and suggest cellular prion protein as a novel potential target for therapeutic immunomodulation.
Collapse
Affiliation(s)
- Wei Hu
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Direct and consensual murine pupillary reflex metrics: establishing normative values. Auton Neurosci 2009; 151:164-7. [PMID: 19683968 DOI: 10.1016/j.autneu.2009.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/25/2009] [Accepted: 07/20/2009] [Indexed: 11/20/2022]
Abstract
Pupillometry is a non-invasive technique, based on well-established neurophysiologic principles, that can be utilized to objectively characterize pathophysiologic demyelinating and neurodegenerative changes involving the pupillary reflex pathway. In animal models of human disorders, pupillometry derived reflex metrics could potentially be used to longitudinally monitor disease activity and responses to pharmacotherapies. These investigations would have important implications for translational initiatives focused on the identification and application of novel neuroprotective and restorative treatments for human diseases such as multiple sclerosis. Here, we have established normal reference values for various pupillary reflex metrics across different mouse strains. Ultimately, we anticipate that this new data will help to catalyze unique lines of inquiry using pupillometry methods.
Collapse
|
45
|
Hu W, Metselaar J, Ben LH, Cravens PD, Singh MP, Frohman EM, Eagar TN, Racke MK, Kieseier BC, Stüve O. PEG minocycline-liposomes ameliorate CNS autoimmune disease. PLoS One 2009; 4:e4151. [PMID: 19127301 PMCID: PMC2613526 DOI: 10.1371/journal.pone.0004151] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Accepted: 11/21/2008] [Indexed: 11/18/2022] Open
Abstract
Background Minocycline is an oral tetracycline derivative with good bioavailability in the central nervous system (CNS). Minocycline, a potent inhibitor of matrix metalloproteinase (MMP)-9, attenuates disease activity in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Potential adverse effects associated with long-term daily minocycline therapy in human patients are concerning. Here, we investigated whether less frequent treatment with long-circulating polyethylene glycol (PEG) minocycline liposomes are effective in treating EAE. Findings Performing in vitro time kinetic studies of PEG minocycline-liposomes in human peripheral blood mononuclear cells (PBMCs), we determined that PEG minocycline-liposome preparations stabilized with CaCl2 are effective in diminishing MMP-9 activity. Intravenous injections of PEG minocycline-liposomes every five days were as effective in ameliorating clinical EAE as daily intraperitoneal injections of minocycline. Treatment of animals with PEG minocycline-liposomes significantly reduced the number of CNS-infiltrating leukocytes, and the overall expression of MMP-9 in the CNS. There was also a significant suppression of MMP-9 expression and proteolytic activity in splenocytes of treated animals, but not in CNS-infiltrating leukocytes. Thus, leukocytes gaining access to the brain and spinal cord require the same absolute amount of MMP-9 in all treatment groups, but minocycline decreases the absolute cell number. Conclusions Our data indicate that less frequent injections of PEG minocycline-liposomes are an effective alternative pharmacotherapy to daily minocycline injections for the treatment of CNS autoimmune diseases. Also, inhibition of MMP-9 remains a promising treatment target in EAE and patients with MS.
Collapse
Affiliation(s)
- Wei Hu
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Josbert Metselaar
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Li-Hong Ben
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Petra D. Cravens
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Mahendra P. Singh
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Elliot M. Frohman
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Ophthalmology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Todd N. Eagar
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Center for Immunology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Michael K. Racke
- Department of Neurology, Ohio State University, Columbus, Ohio, United States of America
| | - Bernd C. Kieseier
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany
- * E-mail: (BCK); (OS)
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Center for Immunology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany
- Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, Texas, United States of America
- * E-mail: (BCK); (OS)
| |
Collapse
|
46
|
Weber MS, Hemmer B. Cooperation of B cells and T cells in the pathogenesis of multiple sclerosis. Results Probl Cell Differ 2009; 51:115-26. [PMID: 19582406 DOI: 10.1007/400_2009_21] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
B cells and T cells are two major players in the pathogenesis of multiple sclerosis (MS) and cooperate at various check points. B cells, besides serving as a source for antibody-secreting plasma cells, are efficient antigen presenting cells for processing of intact myelin antigen and subsequent activation and pro-inflammatory differentiation of T cells. This notion is supported by the immediate clinical benefit of therapeutic B cell depletion in MS, presumably abrogating development of encephalitogenic T cells. However, different B cell subsets strongly vary in their respective effect on T cell differentiation which may relate to B cell phenotype, activation status, antigen specificity and the immunological environment where a B cell encounters a naïve T cell in. In this regard, some B cells also have anti-inflammatory properties producing regulatory cytokines and facilitating development and maintenance of other immunomodulatory immune cells, such as regulatory T cells. Reciprocally, differentiated T cells influence T cell polarizing B cell properties establishing a positive feedback loop of joint pro- or anti-inflammatory B and T cell developments. Further, under the control of activated T helper cells, antigen-primed B cells can switch immunoglobulin isotype, terminally commit to the plasma cell pathway or enter the germinal center reaction to memory B Cell development. Taken together, B cells and T cells thus closely support one another to participate in the pathogenesis of MS in an inflammatory but also in a regulatory manner.
Collapse
Affiliation(s)
- Martin S Weber
- Department of Neurology, Technische Universität München, Ismaningerstrasse 22, 81675, Munich, Germany
| | | |
Collapse
|
47
|
CIITA versus IFN-gamma induced MHC class II expression in head and neck cancer cells. Arch Dermatol Res 2008; 301:189-93. [PMID: 19104823 DOI: 10.1007/s00403-008-0922-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Revised: 11/07/2008] [Accepted: 12/06/2008] [Indexed: 01/01/2023]
Abstract
A growing body of evidence suggests that optimal induction of systemic anti-tumor immunity requires priming of both the CD4+ and CD8+ T cells that are specific for tumor-associated antigens (TAA). Recently, it was shown that MHC class II positive tumor cells are able to induce tumor-specific CD4+ T cells, and that this event may improve clinical outcome. This has rekindled the interest in modulating MHC class II expression in nonprofessional antigen presenting tumor cells. The class II transactivator (CIITA) is a major regulator of MHC class I and class II expression. We compared, in head and neck cancer cell lines, the effect of stable overexpression of CIITA to treatment with IFN-gamma on the cell surface expression profile of MHC class I and II molecules. Here, we provide evidence that CIITA transfection is more effective than IFN-gamma in inducing MHC class II expression. To more thoroughly explore the mechanisms of MHC class II induction in this context, we used RT-PCR to measure the mRNA expression pattern of HLA-DR, HLA-DM, cathepsin S, and the invariant chain. In contrast to the effect of treatment with IFN-gamma, CIITA transfection did not induce cathepsin S, an important protease responsible for the degradation of the invariant chain, and thus for binding of the peptides to the MHC class II binding groove. These findings may have a significant impact on practical and clinical aspects of tumor immunotherapeutic strategies.
Collapse
|
48
|
Singh MP, Meyer zu Hörste G, Hu W, Mausberg AK, Cravens PD, Eagar T, Löber S, Klingenstein R, Gmeiner P, Korth C, Kieseier BC, Stüve O. Quinpramine is a novel compound effective in ameliorating brain autoimmune disease. Exp Neurol 2008; 215:397-400. [PMID: 18996373 DOI: 10.1016/j.expneurol.2008.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Revised: 09/30/2008] [Accepted: 10/06/2008] [Indexed: 11/20/2022]
Abstract
Acridine-iminodibenzyl chimeric compounds were previously introduced as a class of cholesterol-redistributing substances with antiprion effects. Here, we show that administration of the lead compound quinpramine to mice with experimental autoimmune encephalitis, an animal model of multiple sclerosis (MS), significantly ameliorates disease in preventive and therapeutic paradigms. Quinpramine treatment decreased the number of inflammatory CNS lesions, antigen-specific T-cell proliferation, and pro-inflammatory cytokines IFNgamma and IL-17. Quinpramine is thus an immunoregulatory drug that is a candidate pharmaceutical for MS.
Collapse
Affiliation(s)
- Mahendra P Singh
- Department of Neurology, University of Texas Southwestern, Dallas, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
It has long been thought that astrocytes, like other glial cells, simply provide a support mechanism for neuronal function in the healthy and inflamed central nervous system (CNS). However, recent evidence suggests that astrocytes play an active and dual role in CNS inflammatory diseases such as multiple sclerosis (MS). Astrocytes not only have the ability to enhance immune responses and inhibit myelin repair, but they can also be protective and limit CNS inflammation while supporting oligodendrocyte and axonal regeneration. The particular impact of these cells on the pathogenesis and repair of an inflammatory demyelinating process is dependent upon a number of factors, including the stage of the disease, the type and microenvironment of the lesion, and the interactions with other cell types and factors that influence their activation. In this review, we summarize recent data supporting the idea that astrocytes play a complex role in the regulation of CNS autoimmunity.
Collapse
Affiliation(s)
- A. Nair
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Fienberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611 USA
| | - T. J. Frederick
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Fienberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611 USA
| | - S. D. Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Fienberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611 USA
| |
Collapse
|
50
|
Harnesk K, Swanberg M, Ockinger J, Diez M, Lidman O, Wallström E, Lobell A, Olsson T, Piehl F. Vra4 congenic rats with allelic differences in the class II transactivator gene display altered susceptibility to experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2008; 180:3289-96. [PMID: 18292553 DOI: 10.4049/jimmunol.180.5.3289] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Presentation of Ag bound to MHC class II (MHC II) molecules to CD4+ T cells is a key event in adaptive immune responses. Genetic differences in MHC II expression in the rat CNS were recently positioned to allelic variability in the CIITA gene (Mhc2ta), located within the Vra4 locus on rat chromosome 10. In this study, we have examined reciprocal Vra4-congenic strains on the DA and PVGav1 backgrounds, respectively. After experimental nerve injury the strain-specific MHC II expression on microglia was reversed in the congenic strains. Similar findings were obtained after intraparenchymal injection of IFN-gamma in the brain. Expression of MHC class II was also lower on B cells and dendritic cells from the DA.PVGav1-Vra4- congenic strain compared with DA rats after in vitro stimulation with IFN-gamma. We next explored whether Vra4 may affect the outcome of experimental autoimmune disease. In experimental autoimmune encephalomyelitis induced by immunization with myelin oligodendrocyte glycoprotein, DA.PVGav1-Vra4 rats displayed a lower disease incidence and milder disease course compared with DA, whereas both PVGav1 and PVGav1.DA-Vra4 rats were completely protected. These results demonstrate that naturally occurring allelic differences in Mhc2ta have profound effects on the quantity of MHC II expression in the CNS and on immune cells and that this genetic variability also modulates susceptibility to autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Karin Harnesk
- Department of Clinical Neurosciences, Neuroimmunology Unit, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|