1
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
2
|
Janssens R, Struyf S, Proost P. Pathological roles of the homeostatic chemokine CXCL12. Cytokine Growth Factor Rev 2018; 44:51-68. [PMID: 30396776 DOI: 10.1016/j.cytogfr.2018.10.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
Abstract
CXCL12 is a CXC chemokine that traditionally has been classified as a homeostatic chemokine. It contributes to physiological processes such as embryogenesis, hematopoiesis and angiogenesis. In contrast to these homeostatic functions, increased expression of CXCL12 in general, or of a specific CXCL12 splicing variant has been demonstrated in various pathologies. In addition to this increased or differential transcription of CXCL12, also upregulation of its receptors CXC chemokine receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) contributes to the onset or progression of diseases. Moreover, posttranslational modification of CXCL12 during disease progression, through interaction with locally produced molecules or enzymes, also affects CXCL12 activity, adding further complexity. As CXCL12, CXCR4 and ACKR3 are broadly expressed, the number of pathologies wherein CXCL12 is involved is growing. In this review, the role of the CXCL12/CXCR4/ACKR3 axis will be discussed for the most prevalent pathologies. Administration of CXCL12-neutralizing antibodies or small-molecule antagonists of CXCR4 or ACKR3 delays disease onset or prevents disease progression in cancer, viral infections, inflammatory bowel diseases, rheumatoid arthritis and osteoarthritis, asthma and acute lung injury, amyotrophic lateral sclerosis and WHIM syndrome. On the other hand, CXCL12 has protective properties in Alzheimer's disease and multiple sclerosis, has a beneficial role in wound healing and has crucial homeostatic properties in general.
Collapse
Affiliation(s)
- Rik Janssens
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, B-3000 Leuven, Belgium
| | - Sofie Struyf
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, B-3000 Leuven, Belgium
| | - Paul Proost
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, B-3000 Leuven, Belgium.
| |
Collapse
|
3
|
Fagone P, Mazzon E, Bramanti P, Bendtzen K, Nicoletti F. Gasotransmitters and the immune system: Mode of action and novel therapeutic targets. Eur J Pharmacol 2018; 834:92-102. [PMID: 30016662 DOI: 10.1016/j.ejphar.2018.07.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022]
Abstract
Gasotransmitters are a group of gaseous molecules, with pleiotropic biological functions. These molecules include nitric oxide (NO), hydrogen sulfide (H2S), and carbon monoxide (CO). Abnormal production and metabolism of these molecules have been observed in several pathological conditions. The understanding of the role of gasotransmitters in the immune system has grown significantly in the past years, and independent studies have shed light on the effect of exogenous and endogenous gasotransmitters on immune responses. Moreover, encouraging results come from the efficacy of NO-, CO- and H2S -donors in preclinical animal models of autoimmune, acute and chronic inflammatory diseases. To date, data on the influence of gasotransmitters in immunity and immunopathology are often scattered and partial, and the scarcity of clinical trials using NO-, CO- and H2S -donors, reveals that more effort is warranted. This review focuses on the role of gasotransmitters in the immune system and covers the evidences on the possible use of gasotransmitters for the treatment of inflammatory conditions.
Collapse
Affiliation(s)
- Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, Stada Statale 113, C.da Casazza, 98124 Messina, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, Stada Statale 113, C.da Casazza, 98124 Messina, Italy
| | - Klaus Bendtzen
- Institute for Inflammation Research, Rigshospitalet, Copenhagen, Denmark
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
4
|
Jevtić B, Djedović N, Stanisavljević S, Gašić U, Mišić D, Despotović J, Samardžić J, Miljković D, Timotijević G. Anti-encephalitogenic effects of cucumber leaf extract. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
5
|
Jevtić B, Djedović N, Stanisavljević S, Despotović J, Miljković D, Timotijević G. Cucurbitacin E Potently Modulates the Activity of Encephalitogenic Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:4900-7. [PMID: 27225664 DOI: 10.1021/acs.jafc.6b00951] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cucurbitacin E (CucE) is a highly oxidized steroid consisting of a tetracyclic triterpene. It is a member of a Cucurbitacin family of biomolecules that are predominantly found in Cucurbitaceae plants. CucE has already been identified as a potent anti-inflammatory compound. Here, its effects on CD4(+) T helper (Th) cells and macrophages, as the major encephalitogenic cells in the autoimmunity of the central nervous system, were investigated. Production of major pathogenic Th cell cytokines: interferon-gamma and interleukin-17 were inhibited under the influence of CucE. The effects of CucE on CD4(+) T cells were mediated through the modulation of aryl hydrocarbon receptor, STAT3, NFκB, p38 MAPK, and miR-146 signaling. Further, production of nitric oxide and reactive oxygen species, as well as phagocytic ability, were inhibited in macrophages treated with CucE. These results imply that CucE possesses powerful antiencephalitogenic activity.
Collapse
Affiliation(s)
- Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Neda Djedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Jovana Despotović
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade , Belgrade, Serbia
| | - Djordje Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Gordana Timotijević
- Laboratory for Plant Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade , Belgrade, Serbia
| |
Collapse
|
6
|
Predonzani A, Calì B, Agnellini AHR, Molon B. Spotlights on immunological effects of reactive nitrogen species: When inflammation says nitric oxide. World J Exp Med 2015; 5:64-76. [PMID: 25992321 PMCID: PMC4436941 DOI: 10.5493/wjem.v5.i2.64] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/09/2015] [Accepted: 02/04/2015] [Indexed: 02/06/2023] Open
Abstract
Over the last decades, nitric oxide (NO) has been definitively recognised as one of the key players involved in immunity and inflammation. NO generation was originally described in activated macrophages, which still represent the prototype of NO-producing cells. Notwithstanding, additional cell subsets belonging to both innate and adaptive immunity have been documented to sustain NO propagation by means of the enzymatic activity of different nitric oxide synthase isoforms. Furthermore, due to its chemical characteristics, NO could rapidly react with other free radicals to generate different reactive nitrogen species (RNS), which have been intriguingly associated with many pathological conditions. Nonetheless, the plethora of NO/RNS-mediated effects still remains extremely puzzling. The aim of this manuscript is to dig into the broad literature on the topic to provide intriguing insights on NO-mediated circuits within immune system. We analysed NO and RNS immunological clues arising from their biochemical properties, immunomodulatory activities and finally dealing with their impact on different pathological scenarios with far prompting intriguing perspectives for their pharmacological targeting.
Collapse
|
7
|
Pathologic role of glial nitric oxide in adult and pediatric neuroinflammatory diseases. Neurosci Biobehav Rev 2014; 45:168-82. [DOI: 10.1016/j.neubiorev.2014.06.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 05/28/2014] [Accepted: 06/05/2014] [Indexed: 01/22/2023]
|
8
|
Park HJ, Kim Y, Kim H, Ha E, Park H, Yoon S, Kim MJ, Choi S, Ryu Y, Park HK, Hong M. Neuroprotective effect by Dammishimgyu-herbal acupuncture against H2O2-induced apoptosis in human neuroblastoma, SH-SY5Y cells. Neurol Res 2013; 29 Suppl 1:S93-7. [PMID: 17359648 DOI: 10.1179/016164107x172329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
BACKGROUND The free radical is involved in neuronal cell death in human neurodegenerative disease. Dammishimgyu (DMSG)-herbal acupuncture has been used to treat neurological disorders in Korea. The present study was aimed to investigate the neuroprotective effect of DMSG-herbal acupuncture against H(2)O(2)-induced apoptosis in human neuroblastoma cell line, SH-SY5Y. METHODS The neuroprotective effect of DMSG-herbal acupuncture on H(2)O(2) induced apoptosis was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, 4,6-diamidino-2-phenylindole (DAPI) staining, reverse transcription-polymerase chain reaction (RT-PCR), western blots and nitrite assay. RESULTS In this study, 100 microM H(2)O(2)-treated cells decreased the cell viability with apoptotic features and increased the production of nitric oxide (NO). However, 0.1% DMSG treatment after exposure to 100 microM H(2)O(2) inhibited both H(2)O(2)-stimulated mRNA and protein expressions of BCL2-associated X protein (BAX) and caspase 3 apoptosis-related cysteine peptidase (CASP3). In addition, 0.1% DMSG treatment inhibited the increased NO production induced by H(2)O(2). CONCLUSION These results suggest that DMSG-herbal acupuncture shows protective effect against H(2)O(2)-induced neuronal damage.
Collapse
Affiliation(s)
- Hi-Joon Park
- TKM Research Group, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Petković F, Blaževski J, Momčilović M, Mostarica Stojkovic M, Miljković D. Nitric oxide inhibits CXCL12 expression in neuroinflammation. Immunol Cell Biol 2013; 91:427-34. [PMID: 23732617 DOI: 10.1038/icb.2013.23] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 05/07/2013] [Accepted: 05/08/2013] [Indexed: 02/07/2023]
Abstract
Chemokine CXCL12 (C-X-C motif chemokine ligand 12) restricts immune cell invasion of the central nervous system (CNS) and limits neuroinflammation in experimental autoimmune encephalomyelitis (EAE), an animal model of inflammatory and demyelinating disease of the CNS, multiple sclerosis (MS). Nitric oxide (NO), by contrast, predominantly contributes to CNS tissue destruction in MS and EAE. Thus, the influence of NO on CXCL12 in the inflamed CNS was investigated. Excess expression of inducible NO synthase was inversely correlated to CXCL12 gene expression in spinal cord homogenates of rats immunized to develop EAE. NO inhibited gene expression of CXCL12 in astrocytes and endothelial cells in vitro. The inhibition was paralleled with reduction of p38 mitogen-activated protein kinase (MAPK) phosphorylation and it was mimicked with inhibitors of p38 MAPK activation in astrocytes. In vivo suppression of nitric generation recovered CXCL12 expression in the CNS and attenuated EAE in Dark Agouti rats. On the contrary, in vivo NO donation decreased CXCL12 expression in the CNS of EAE-resistant Albino Oxford (AO) rats. However, the effect was not paralleled with induction of EAE in AO rats. It is suggested that NO acting through suppression of p38 MAPK inhibits CXCL12 expression in neuroinflammation. These results imply that downregulation of NO release and protection of CXCL12 expression within the CNS might present the potential approaches in MS therapy.
Collapse
Affiliation(s)
- Filip Petković
- Department of Immunology, Institute for Biological Research, Siniša Stanković, University of Belgrade, Belgrade, Serbia
| | | | | | | | | |
Collapse
|
10
|
Bergman P, James T, Kular L, Ruhrmann S, Kramarova T, Kvist A, Supic G, Gillett A, Pivarcsi A, Jagodic M. Next-generation sequencing identifies microRNAs that associate with pathogenic autoimmune neuroinflammation in rats. THE JOURNAL OF IMMUNOLOGY 2013; 190:4066-75. [PMID: 23514736 DOI: 10.4049/jimmunol.1200728] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
MicroRNAs (miRNAs) are known to regulate most biological processes and have been found dysregulated in a variety of diseases, including multiple sclerosis (MS). In this study, we characterized miRNAs that associate with susceptibility to develop experimental autoimmune encephalomyelitis (EAE) in rats, a well-established animal model of MS. Using Illumina next-generation sequencing, we detected 544 miRNAs in the lymph nodes of EAE-susceptible Dark Agouti and EAE-resistant Piebald Virol Glaxo rats during immune activation. Forty-three miRNAs were found differentially expressed between the two strains, with 81% (35 out of 43) showing higher expression in the susceptible strain. Only 33% of tested miRNAs displayed differential expression in naive lymph nodes, suggesting that a majority of regulated miRNAs are EAE dependent. Further investigation of a selected six miRNAs indicates differences in cellular source and kinetics of expression. Several of the miRNAs, including miR-146a, miR-21, miR-181a, miR-223, and let-7, have previously been implicated in immune system regulation. Moreover, 77% (33 out of 43) of the miRNAs were associated with MS and other autoimmune diseases. Target genes likely regulated by the miRNAs were identified using computational predictions combined with whole-genome expression data. Differentially expressed miRNAs and their targets involve functions important for MS and EAE, such as immune cell migration through targeting genes like Cxcr3 and cellular maintenance and signaling by regulation of Prkcd and Stat1. In addition, we demonstrated that these three genes are direct targets of miR-181a. Our study highlights the impact of multiple miRNAs, displaying diverse kinetics and cellular sources, on development of pathogenic autoimmune inflammation.
Collapse
Affiliation(s)
- Petra Bergman
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Harrison DG, Guzik TJ, Lob HE, Madhur MS, Marvar PJ, Thabet SR, Vinh A, Weyand CM. Inflammation, immunity, and hypertension. Hypertension 2010; 57:132-40. [PMID: 21149826 DOI: 10.1161/hypertensionaha.110.163576] [Citation(s) in RCA: 611] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- David G Harrison
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN 37232-6602, USA.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Mix E, Meyer-Rienecker H, Hartung HP, Zettl UK. Animal models of multiple sclerosis--potentials and limitations. Prog Neurobiol 2010; 92:386-404. [PMID: 20558237 PMCID: PMC7117060 DOI: 10.1016/j.pneurobio.2010.06.005] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 06/01/2010] [Accepted: 06/07/2010] [Indexed: 12/17/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is still the most widely accepted animal model of multiple sclerosis (MS). Different types of EAE have been developed in order to investigate pathogenetic, clinical and therapeutic aspects of the heterogenic human disease. Generally, investigations in EAE are more suitable for the analysis of immunogenetic elements (major histocompatibility complex restriction and candidate risk genes) and for the study of histopathological features (inflammation, demyelination and degeneration) of the disease than for screening of new treatments. Recent studies in new EAE models, especially in transgenic ones, have in connection with new analytical techniques such as microarray assays provided a deeper insight into the pathogenic cellular and molecular mechanisms of EAE and potentially of MS. For example, it was possible to better delineate the role of soluble pro-inflammatory (tumor necrosis factor-α, interferon-γ and interleukins 1, 12 and 23), anti-inflammatory (transforming growth factor-β and interleukins 4, 10, 27 and 35) and neurotrophic factors (ciliary neurotrophic factor and brain-derived neurotrophic factor). Also, the regulatory and effector functions of distinct immune cell subpopulations such as CD4+ Th1, Th2, Th3 and Th17 cells, CD4+FoxP3+ Treg cells, CD8+ Tc1 and Tc2, B cells and γδ+ T cells have been disclosed in more detail. The new insights may help to identify novel targets for the treatment of MS. However, translation of the experimental results into the clinical practice requires prudence and great caution.
Collapse
Key Words
- apc, antigen-presenting cell
- at-eae, adoptive transfer eae
- bbb, blood–brain barrier
- bdnf, brain-derived neurotrophic factor
- cd, cluster of differentiation
- cns, central nervous system
- cntf, ciliary neurotrophic factor
- eae, experimental autoimmune encephalomyelitis
- hla, human leukocyte antigen
- ig, immunoglobulin
- il, interleukin
- ifn, interferon
- ivig, intravenous immunoglobulin
- mab, monoclonal antibody
- mbp, myelin basic protein
- mhc, major histocompatibility complex
- mog, myelin oligodendrocyte glycoprotein
- mp, methylprednisolone
- mri, magnetic resonance imaging
- ms, multiple sclerosis
- nk, natural killer
- odc, oligodendrocyte
- qtl, quantitative trait locus
- plp, proteolipid protein
- tc, cytotoxic t cell
- tcr, t cell receptor
- tgf, transforming growth factor
- th cell, helper t cell
- tnf, tumor necrosis factor
- animal model
- autoimmunity
- experimental autoimmune encephalomyelitis
- immunogenetics
- immunomodulatory therapy
- multiple sclerosis
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Clinical Trials as Topic
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Gene Expression Profiling
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Microarray Analysis
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/physiopathology
- Multiple Sclerosis/therapy
Collapse
Affiliation(s)
- Eilhard Mix
- Department of Neurology, University of Rostock, Germany
| | | | - Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine-University, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Uwe K. Zettl
- Department of Neurology, University of Rostock, Germany
| |
Collapse
|
13
|
Silveira-Lacerda EDP, Vilanova-Costa CAST, Pereira FDC, Hamaguchi A, Pavanin LA, Goulart LR, Homsi-Brandenburgo MI, Soares AM, dos Santos WB, Nomizo A. The ruthenium complex cis-(Dichloro)Tetraammineruthenium(III) chloride presents immune stimulatory activity on human peripheral blood mononuclear cells. Biol Trace Elem Res 2010; 133:270-83. [PMID: 19590831 DOI: 10.1007/s12011-009-8440-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 06/22/2009] [Indexed: 11/28/2022]
Abstract
Ruthenium compounds in general are well suited for medicinal applications. They have been investigated as immunosuppressants, nitric oxide scavengers, antimicrobial agents, and antimalarials. The aim of this study is to evaluate the immunomodulatory activity of cis-(dichloro)tetraammineruthenium(III) chloride (cis-[RuCl(2)(NH(3))(4)]Cl) on human peripheral blood mononuclear cells (PBMC). The cytotoxic studies performed here revealed that the ruthenium(III) complex presents a cytotoxic activity towards normal human PBMC, only at very high concentration. Results also showed that cis-[RuCl(2)(NH(3))(4)]Cl presents a dual role on PBMC stimulating proliferation and interleukin-2 (IL-2) production at low concentration and inducing cytotoxicity, inability to proliferate, and inhibiting IL-2 production at high concentration. The noncytotoxic activity of cis-[RuCl(2)(NH(3))(4)]Cl at low concentration towards PBMC, which correlates with the small number of annexin V positive cells and also the absence of DNA fragmentation, suggest that this compound does not induce apoptosis on PBMC. For the first time, we show that, at low concentration (10-100 microg L(-1)), the cis-[RuCl(2)(NH(3))(4)]Cl compound induces peripheral blood lymphocytes proliferation and also stimulates them to IL-2 production. These results open a new potential applicability of ruthenium(III) complexes as a possible immune regulatory compound acting as immune suppressor at high concentration and as immune stimulator at low concentration.
Collapse
Affiliation(s)
- Elisângela de Paula Silveira-Lacerda
- Laboratório de Genética Molecular e Citogenética, Instituto de Ciências Biológicas (ICB I), Universidade Federal de Goiás, Sala 200, Campus Samambaia (Campus II), Cx. Postal: 131, Goiânia, Goiás, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Connon RE, Geist J, Pfeiff J, Loguinov AV, D'Abronzo LS, Wintz H, Vulpe CD, Werner I. Linking mechanistic and behavioral responses to sublethal esfenvalerate exposure in the endangered delta smelt; Hypomesus transpacificus (Fam. Osmeridae). BMC Genomics 2009; 10:608. [PMID: 20003521 PMCID: PMC2806348 DOI: 10.1186/1471-2164-10-608] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 12/15/2009] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The delta smelt (Hypomesus transpacificus) is a pelagic fish species listed as endangered under both the USA Federal and Californian State Endangered Species Acts and considered an indicator of ecosystem health in its habitat range, which is limited to the Sacramento-San Joaquin estuary in California, USA. Anthropogenic contaminants are one of multiple stressors affecting this system, and among them, current-use insecticides are of major concern. Interrogative tools are required to successfully monitor effects of contaminants on the delta smelt, and to research potential causes of population decline in this species. We have created a microarray to investigate genome-wide effects of potentially causative stressors, and applied this tool to assess effects of the pyrethroid insecticide esfenvalerate on larval delta smelt. Selected genes were further investigated as molecular biomarkers using quantitative PCR analyses. RESULTS Exposure to esfenvalerate affected swimming behavior of larval delta smelt at concentrations as low as 0.0625 mug.L-1, and significant differences in expression were measured in genes involved in neuromuscular activity. Alterations in the expression of genes associated with immune responses, along with apoptosis, redox, osmotic stress, detoxification, and growth and development appear to have been invoked by esfenvalerate exposure. Swimming impairment correlated significantly with expression of aspartoacylase (ASPA), an enzyme involved in brain cell function and associated with numerous human diseases. Selected genes were investigated for their use as molecular biomarkers, and strong links were determined between measured downregulation in ASPA and observed behavioral responses in fish exposed to environmentally relevant pyrethroid concentrations. CONCLUSIONS The results of this study show that microarray technology is a useful approach in screening for, and generation of molecular biomarkers in endangered, non-model organisms, identifying specific genes that can be directly linked with sublethal toxicological endpoints; such as changes in expression levels of neuromuscular genes resulting in measurable swimming impairments. The developed microarrays were successfully applied on larval fish exposed to esfenvalerate, a known contaminant of the Sacramento-San Joaquin estuary, and has permitted the identification of specific biomarkers which could provide insight into the factors contributing to delta smelt population decline.
Collapse
Affiliation(s)
- Richard E Connon
- School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California 95616, USA
| | - Juergen Geist
- School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California 95616, USA
- Unit of Functional Aquatic Ecology and Fish Biology, Department of Animal Science, Technische Universität München, D-85350 Freising, Germany
| | - Janice Pfeiff
- School of Veterinary Medicine, Molecular Biosciences, University of California, Davis, California 95616, USA
| | - Alexander V Loguinov
- School of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA
| | - Leandro S D'Abronzo
- School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California 95616, USA
| | - Henri Wintz
- School of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA
- Biorad Laboratories, Life Science Research, Hercules, California, USA
| | - Christopher D Vulpe
- School of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA
| | - Inge Werner
- School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California, Davis, California 95616, USA
| |
Collapse
|
15
|
Mix E, Stefan K, Höppner J, Klauer T, Zettl UK, Strauss U, Meyer-Rienecker HJ, Rolfs A. Lymphocyte Subpopulations, Oxidative Burst and Apoptosis in Peripheral Blood Cells of Patients with Multiple Sclerosis–Effect of Interferon-β. Autoimmunity 2009; 36:291-305. [PMID: 14567559 DOI: 10.1080/0891693031000152697] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
At present, the most efficient therapeutical treatment of multiple sclerosis (MS) is achieved by IFN-beta. However, its in vivo effects remain incompletely understood. If applied parenterally, the hydrophobic IFN-beta acts primarily on blood cells with probable selectivity for functionally different lymphocyte subpopulations, monocytes and granulocytes. We have investigated the expression of the activation marker interleukin-2 receptor-alpha (CD25) on CD3+ T cells, CD19+ B cells, foetal-type gamma(delta)+CD3+ T cells and foetal-type CD5+CD19+ B cells of the peripheral blood. In addition, the oxidative burst activity and apoptosis have been determined in mononuclear and polymorphonuclear blood cells, respectively. The study accompanied a phase III trial with IFN-beta1b (BETAFERON, Schering). Two groups of MS patients with relapsing-remitting course of the disease have been investigated at 8 time points (days 0, 5, 15, 31, 60, 90, 180 and 270 after starting therapy): (1) verum group (n = 8) with application of 8 Mill. units IFN-beta1 b every other day, and (2) placebo group (n = 4) with application of placebo for 3 months and therapy as in (1) from day 90 onward. The main results were: (1) Activated T cells decreased until day 180 in the verum group and return thereafter to pre-treatment values, whereas in the placebo group the values remained relatively stable over the whole observation period. (2) Activated B cells increased between days 90 and 270 in both groups, i.e. after verum application in both groups. (3) Foetal-type B cells were more activated than total B and T cells with increase over time in both groups. (4) Foetal-type T cells exerted relatively stable intra-individual levels with generally low CD25 expression, but punctual CD25 peaks in both groups. (5) The spontaneous oxidative burst was higher in lymphocytes, more variable in monocytes and faster increasing in granulocytes in the verum group than in the placebo group. (6) Apoptosis of mononuclear cells and granulocytes showed similar variations in the verum and placebo groups with the exception of a selective increase over time of the proportion of granulocytes undergoing induced apoptosis in the verum group. It is concluded that IFN-beta has the following main effects on the immune system of MS patients: (1) the T cell immunity is systemically and reversibly suppressed, (2) the foetal-type lymphocytes, which are responsible for the first line of defence of infections, are stimulated in the long range, (3) the oxidative burst activity is increased in lymphocytes and granulocytes and instable in monocytes, and (4) the inducibility of apoptosis in granulocytes is increased. Re-examination of the altered blood cell parameters after long-term IFN-beta therapy is warranted.
Collapse
Affiliation(s)
- Eilhard Mix
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, P.O. Box 100888, 18055 Rostock, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Dello Russo C, Lisi L, Tringali G, Navarra P. Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem Pharmacol 2009; 78:1242-51. [PMID: 19576187 DOI: 10.1016/j.bcp.2009.06.097] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 06/19/2009] [Accepted: 06/22/2009] [Indexed: 01/31/2023]
Abstract
Neuroinflammation plays a prominent role in the pathophysiology of several neurodegenerative disorders, including Multiple Sclerosis. Reactive microglial cells are always found in areas of active demyelination as well as in normal-appearing white matter. Microglia contribute to initiating and maintaining brain inflammation, and once activated release pro-inflammatory mediators potentially cytotoxic, like nitric oxide (NO). It is now evident that the mTOR signaling pathway regulates different functions in the innate immune system, contributing to macrophage activation. More recently, mTOR has been found to enhance the survival of EOC2 microglia during oxygen-glucose deprivation and increase NO synthase 2 (NOS2) expression during hypoxia in BV2 microglial cell line, thus suggesting an involvement in microglial pro-inflammatory activation. In the present study, we detected mTOR activation in response to two different stimuli, namely LPS and a mixture of cytokines, in primary cultures of rat cortical microglia. Moreover, mTOR inhibitors reduced NOS activity and NOS2 expression induced by cytokines, but not those induced by LPS. The mTOR inhibitor RAD001, in combination with cytokines, also reduced microglial proliferation and the intracellular levels of cyclooxygenase. Under basal conditions mTOR inhibition significantly reduced microglial viability. Interestingly, mTOR inhibitors did not display any relevant effect on astrocyte NOS2 activity or cell viability. In conclusion, mTOR selectively controls microglial activation in response to pro-inflammatory cytokines and appears to play a crucial role in microglial viability; thus these drugs may be a useful pharmacological tool to reduce neuroinflammation.
Collapse
Affiliation(s)
- Cinzia Dello Russo
- Institute of Pharmacology, Catholic University Medical School, Largo F. Vito 1, Rome, Italy.
| | | | | | | |
Collapse
|
17
|
Kiang JG, Krishnan S, Lu X, Li Y. Inhibition of inducible nitric-oxide synthase protects human T cells from hypoxia-induced apoptosis. Mol Pharmacol 2008; 73:738-47. [PMID: 18079278 DOI: 10.1124/mol.107.041079] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sodium cyanide-induced chemical hypoxia triggers a series of biochemical alterations leading to apoptosis in many cell types, including T cells. It is known that chemical hypoxia promotes inducible nitric-oxide synthase (iNOS) gene transcription by activating its transcription factors. To determine whether iNOS and NO production are responsible for chemical hypoxia-induced apoptosis, we exposed human Jurkat T cells to sodium cyanide in the presence or absence of iNOS inhibitors. We found that iNOS expression is necessary for hypoxia-induced lipid peroxidation and leukotriene B(4) generation. The inhibition of iNOS limited T-cell apoptosis by decreasing the activity of caspase-3 without affecting the expression of Fas/Apo-1/CD95 on the surface membrane of T cells. These data suggest iNOS-mediated NO produced endogenously in the T cell alters overall T-cell function and results in apoptosis. Proper control of iNOS expressed in the T cell may represent a useful approach to immunomodulation.
Collapse
Affiliation(s)
- Juliann G Kiang
- Armed Forces Radiobiology Research Institute, Bldg. 46, Room 2423, 8901 Wisconsin Ave., Bethesda, MD 20889-5603, USA.
| | | | | | | |
Collapse
|
18
|
Pender MP. Treating autoimmune demyelination by augmenting lymphocyte apoptosis in the central nervous system. J Neuroimmunol 2007; 191:26-38. [PMID: 17931708 DOI: 10.1016/j.jneuroim.2007.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Accepted: 09/07/2007] [Indexed: 01/06/2023]
Abstract
The elimination of autoreactive T cells from the central nervous system (CNS) by apoptosis plays an important role in switching off autoimmune attack. B-cell apoptosis in the CNS probably also has a key role in downregulating autoimmunity. Augmenting lymphocyte apoptosis in the CNS is a potential strategy for treating autoimmune CNS diseases such as multiple sclerosis. These strategies involve modulation of the physiological pro-apoptotic and anti-apoptotic pathways that control lymphocyte fate in the CNS. In the case of T cells, apoptosis can be augmented by enhancing activation-induced T-cell apoptosis through the CD95 (Fas) pathway and by inhibiting costimulation-induced anti-apoptotic pathways mediated through BCL-2 and BCL-X L.
Collapse
Affiliation(s)
- Michael P Pender
- Neuroimmunology Research Centre, School of Medicine, The University of Queensland, Australia.
| |
Collapse
|
19
|
Abstract
Because of the anatomy, function, and nonregenerative nature of the myocardium, inflammation in this tissue is not well tolerated. Nevertheless, various diseases of the heart are characterized by inflammatory responses involving the effector mechanisms of innate and adaptive (lymphocyte-dependent) immunity. The innate immune response to ischemia-reperfusion injury is, by far, the most common cause of myocardial inflammation. Innate responses may have beneficial influences that preserve myocardial function in the short term but may be maladaptive in chronic states. Adaptive responses in the myocardium occur with infection or loss of tolerance, and lead to myocarditis. Given the narrow margin for benefit of cardiac inflammation, special regulatory mechanisms likely raise the threshold, compared to other tissues, for the induction and persistence of adaptive immune responses. These mechanisms include strong central and peripheral T cell tolerance to heart antigens and induction of anti-inflammatory feedback mechanisms involving cytokines such as interferon-gamma.
Collapse
Affiliation(s)
- Viviany R Taqueti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
20
|
Vairetti M, Ferrigno A, Bertone R, Richelmi P, Bertè F, Freitas I. Apoptosis vs. necrosis: glutathione-mediated cell death during rewarming of rat hepatocytes. Biochim Biophys Acta Mol Basis Dis 2005; 1740:367-74. [PMID: 15949704 DOI: 10.1016/j.bbadis.2004.11.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Revised: 11/18/2004] [Accepted: 11/19/2004] [Indexed: 12/16/2022]
Abstract
Hypothermia induces injury in its own right, but the mechanisms involved in the cell damage are still unclear. The aim of this study was to test the effects that glutathione (GSH) depletion induces on cell death in isolated rat hepatocytes, kept at 4 degrees C for 20 h, by modulating intracellular GSH concentration with diethylmaleate and buthionine sulfoximine (DEM and BSO). Untreated hepatocytes showed Annexin V stained cells (AnxV(+)), scarce propidium iodide stained cells (PI(+)) and presented a low level of lactate dehydrogenase (LDH) leakage after 20 h at 4 degrees C and rewarming at 37 degrees C. When DEM and BSO were added before cold storage, we observed a few AnXV(+) cells and an increase in PI(+) cells associated with LDH release in the incubation medium. Conversely, the addition of DEM and BSO only during rewarming caused a marked increase in cell death by apoptosis. Production of reactive oxygen species (ROS) and thiobarbituric acid species (TBARS), associated with a decrease in GSH concentrations, was higher when DEM and BSO were added before cold storage. Cells treated with DEM and BSO before cold storage showed lower ATP energy stores than hepatocytes treated with DEM and BSO only during rewarming. Pretreatment of hepatocytes with deferoxamine protected against apoptotic and necrotic morphology in conditions of GSH depletion. These results suggest that pretreatment of hepatocytes with DEM and BSO before cold storage induces necrosis, while the treatment of hepatocytes only during rewarming increases apoptosis. In both conditions, iron represents a crucial mediator of cell death.
Collapse
Affiliation(s)
- Mariapia Vairetti
- Department of Internal Medicine and Therapeutics, University of Pavia, Italy.
| | | | | | | | | | | |
Collapse
|
21
|
Apoptotic Cell Death in Experimental Autoimmune Encephalomyelitis. EXPERIMENTAL MODELS OF MULTIPLE SCLEROSIS 2005. [PMCID: PMC7122661 DOI: 10.1007/0-387-25518-4_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Particularly in the vulnerable CNS with a low capacity for regeneration specialized mechanisms must be active for the fast and gentle elimination of dysregulated autoaggressive immune cells. In EAE, local apoptosis of autoimmune T-cells has been identified as a safe means for the removal of these unwanted cells. T-cell apoptosis in situ followed by phagocytic clearance of apoptotic remnants by glia assures a minimum of detrimental bystander damage to the local parenchyma and down-regulates the local inflammatory reaction. The pharmacological augmentation of local apoptosis of inflammatory effector cells might gain therapeutic importance also in human neuroimmunological diseases such as multiple sclerosis.
Collapse
|
22
|
Dalton DK, Wittmer S. Nitric-oxide-dependent and independent mechanisms of protection from CNS inflammation during Th1-mediated autoimmunity: evidence from EAE in iNOS KO mice. J Neuroimmunol 2004; 160:110-21. [PMID: 15710464 DOI: 10.1016/j.jneuroim.2004.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2004] [Revised: 11/10/2004] [Accepted: 11/10/2004] [Indexed: 10/26/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) disease was accelerated iNOS-deficient (KO) mice: coinciding with greatly increased numbers of Ag-specific Th1 cells in the periphery that appeared to rapidly shift from the spleen to the CNS during onset of disease symptoms. iNOS KO mice had significantly increased Th1 cells in the CNS versus wild-type mice. Apoptosis of CNS-infiltrating CD4(+) T cells was impaired in iNOS KO mice at peak of disease; consequently, these mice had more CNS-infiltrating CD4(+) T cells. Subsequently, iNOS KO mice up-regulated apoptosis of CNS-CD4(+) T cells. During chronic EAE, CNS macrophages were greatly decreased, suggesting elimination of CNS-infiltrating CD4(+) T cells and activated macrophages by iNOS-independent mechanisms. INOS is not only required for apoptosis of CNS-CD4(+) T cells but also prevents overexpansion of autoreactive Th1 cells in the periphery and the CNS.
Collapse
MESH Headings
- Adjuvants, Immunologic/physiology
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- CD4 Lymphocyte Count
- CD4-Positive T-Lymphocytes/enzymology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Cell Movement/genetics
- Cell Movement/immunology
- Cells, Cultured
- Central Nervous System/enzymology
- Central Nervous System/immunology
- Central Nervous System/pathology
- Chronic Disease
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Epitopes, T-Lymphocyte/immunology
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Macrophage Activation/genetics
- Macrophage Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin Proteins
- Myelin-Associated Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein
- Nitric Oxide/physiology
- Nitric Oxide Synthase/deficiency
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase/physiology
- Nitric Oxide Synthase Type II
- Severity of Illness Index
- Th1 Cells/enzymology
- Th1 Cells/immunology
- Th1 Cells/pathology
Collapse
Affiliation(s)
- Dyana K Dalton
- Trudeau Institute Inc., 154 Algonquin Ave, Saranac Lake, NY 12983, USA.
| | | |
Collapse
|
23
|
Correale J, Villa A. The neuroprotective role of inflammation in nervous system Injuries. J Neurol 2004; 251:1304-16. [PMID: 15592725 DOI: 10.1007/s00415-004-0649-z] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Accepted: 09/02/2004] [Indexed: 12/16/2022]
Abstract
The contribution of inflammation to the pathogenesis of several nervous system disorders has long been established. Other observations, however, indicate that both inflammatory cells and mediators may also have beneficial functions, assisting in repair and recovery processes. There is compelling evidence to indicate that in the injured nervous system, as in other tissues, macrophages are needed at an early stage after injury in order for healing to take place. Likewise, activated T cells of a particular specificity can reduce the spread of damage. This neuroprotective effect of T cells may be caused, at least in part, by the production of neurotrophic factors such as neurotrophin-3 or brain-derived neurotrophic factor. Interestingly, recent findings indicate that immune cells are able to produce a variety of neurotrophic factors which promote neuronal survival and may also mediate anti-inflammatory effects. Numerous cytokines are induced after nervous system injuries. Some cytokines, such as TNF-alpha, IL-1 and IFN-gamma, are well known for their promotion of inflammatory responses. However, these cytokines also have immunosuppressive functions and their subsequent expression also assists in repair or recovery processes, suggesting a dual role for some pro-inflammatory cytokines. This should be clarified, as it may be crucial in the design of therapeutic strategies to target specific cytokine(s). Finally, there is a growing body of evidence to show that autoreactive IgM antibodies may constitute an endogenous system of tissue repair, and therefore prove of value as a therapeutic strategy. Available evidence would appear to indicate that the inflammatory response observed in several neurological conditions is more complex than previously thought. Therefore, the design of more effective therapies depends on a clear delineation of the beneficial and detrimental effects of inflammation.
Collapse
Affiliation(s)
- Jorge Correale
- Raúl Carrea Institute for Neurological Research, FLENI, Montañeses 2325, 1428, Buenos Aires, Argentina.
| | | |
Collapse
|
24
|
Ochi H, Feng-Jun M, Osoegawa M, Minohara M, Murai H, Taniwaki T, Kira JI. Time-dependent cytokine deviation toward the Th2 side in Japanese multiple sclerosis patients with interferon beta-1b. J Neurol Sci 2004; 222:65-73. [PMID: 15240198 DOI: 10.1016/j.jns.2004.04.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2003] [Revised: 04/09/2004] [Accepted: 04/12/2004] [Indexed: 11/23/2022]
Abstract
To address the immune mechanism sustaining interferon beta (IFNbeta) efficacy in multiple sclerosis (MS), we longitudinally analyzed expressions of IFN-gamma, IL-4, IL-5 and IL-13 in CD4+ T cells and CD8+ T cells in 22 Japanese MS patients (16 patients with conventional MS and 6 with opticospinal MS) undergoing IFNbeta using flow cytometry. During the 48-week observation period, five opticospinal MS patients (83%) relapsed compared to only four conventional MS patients (25%); the frequency of relapsed patients was significantly higher in the former (p=0.046). The effects of IFNbeta on individual cytokines were time-dependent and altered cytokine productions were particularly evident in CD4+ rather than CD8+ T cells. A decreased intracellular IFN-gamma/IL-4 ratio in CD4+ T cells was thus evident soon after the initiation of therapy, and persisted for the entire 1 year follow-up period, regardless of whether or not the patient relapsed (p<0.01). IFNbeta treatment resulted in a rapid increase in the percentage of IFN-gamma- IL-4+ and IL-13+ CD4+ T cells 1 week after the initiation of therapy and high values were sustained for 6 months but declined to the baseline over 1 year. Later, the percentage of IFN-gamma+ IL-4- CD4+ T cells decreased significantly from weeks 24 through 48 of therapy (p<0.01). When comparisons with the pretreatment values were made for each subtype of MS, a significant reduction of IFN-gamma+ IL-4- CD4+ T cell percentages was shown in conventional MS (p<0.0001), but not in opticospinal MS. Moreover, when such a comparison was made by the presence or absence of relapse during therapy, a significant reduction of IFN-gamma+ IL-4- CD4+ T cell percentages was observed in MS patients without relapse (p<0.01). Thus, a reduction of IFN-gamma+ IL-4- CD4+ T cell percentages in the late phase of therapy is considered important for reducing relapse in conventional MS. When the expression patterns of IFN-gamma, IL-4, IL-5 and IL-13 in CD4+ T cells and CD8+ T cells were compared between patients with and without relapse during therapy, the only significant difference was an increase in the IL-13+ CD4+ T cell percentages in patients with relapse compared to those without (p<0.05). The results indicate that in CD4+ T cells IL-4 was preferentially up-regulated in the early course and IFN-gamma was down-regulated in the late phase of IFNbeta therapy. The net effect of IFNbeta on the immune balance was entirely toward type 2 immune deviation, possibly contributing to its beneficial effects on MS.
Collapse
Affiliation(s)
- Hirofumi Ochi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Kahl KG, Schmidt HHHW, Jung S, Sherman P, Toyka KV, Zielasek J. Experimental autoimmune encephalomyelitis in mice with a targeted deletion of the inducible nitric oxide synthase gene: increased T-helper 1 response. Neurosci Lett 2004; 358:58-62. [PMID: 15016434 DOI: 10.1016/j.neulet.2003.12.095] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2003] [Revised: 12/15/2003] [Accepted: 12/15/2003] [Indexed: 11/26/2022]
Abstract
Mice with a targeted deletion of the cytokine-inducible nitric oxide synthase gene (iNOS(-/-)) show increased severity of experimental autoimmune encephalomyelitis (EAE). We studied the mechanisms of susceptibility to myelin-basic protein-induced 'active' EAE in iNOS(-/-) mice. Spleen cells and lymph node cells from iNOS(-/-) mice with EAE showed a significantly enhanced ex vivo proliferation and production of T-helper 1 (Th1) cytokines (interferon-gamma by 157 and 57% and tumor-necrosis-factor-alpha by 86 and 27%, respectively). We conclude that NO produced by iNOS plays a protective role in EAE probably by inhibiting the production of Th1 cytokines and T cell proliferation.
Collapse
Affiliation(s)
- Kai G Kahl
- Department of Neurology, Julius-Maximilians-University, Josef-Schneider Str. 11, D-97080 Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Jagtap JC, Chandele A, Chopde BA, Shastry P. Sodium pyruvate protects against H(2)O(2) mediated apoptosis in human neuroblastoma cell line-SK-N-MC. J Chem Neuroanat 2004; 26:109-18. [PMID: 14599660 DOI: 10.1016/s0891-0618(03)00037-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Free radicals are involved in neuronal damage. The present study was aimed to investigate the protective effect of sodium pyruvate-a free radical scavenger against hydrogen peroxide (H(2)O(2)) induced apoptosis in human neuroblastoma cell line-SK-N-MC. On exposure to H(2)O(2) (0.025 mM) cells exhibited apoptosis within 24 h, demonstrating a high caspase 3 activity by 3 h followed by cleavage of PARP that was maximum at 24 h. A break down in the mitochondrial membrane potential was observed 3 h onwards. Sodium pyruvate protected cells significantly (P<0.05) against apoptosis in a dose dependent manner as assessed for cell viability by dye exclusion method and apoptosis by TUNEL. Sodium pyruvate significantly inhibited caspase 3 activity, cleavage of PARP and breakdown of mitochondrial membrane potential. These data suggest that sodium pyruvate protects neuronal damage caused by H(2)O(2).
Collapse
Affiliation(s)
- Jayashree C Jagtap
- Department of Biotechnology, National Center for Cell Science, Government of India An Autonomous Institution, Pune University Campus, Ganeshkhind, 411007 Pune, India
| | | | | | | |
Collapse
|
27
|
Taniguchi Y, Kohno K, Inoue SI, Koya-Miyata S, Okamoto I, Arai N, Iwaki K, Ikeda M, Kurimoto M. Oral administration of royal jelly inhibits the development of atopic dermatitis-like skin lesions in NC/Nga mice. Int Immunopharmacol 2003; 3:1313-24. [PMID: 12890429 DOI: 10.1016/s1567-5769(03)00132-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We have shown previously that in addition to IL-4, IL-5 and IL-10, antigen-specific interferon-gamma (IFN-gamma) production by spleen cells from ovalbumin (OVA)/Alum-immunized mice is inhibited by the administration of royal jelly (RJ). Since it has been shown that both Th1 and Th2 cytokines play pathogenic roles in the generation of atopic dermatitis (AD), we have examined whether RJ suppresses the development of AD-like skin lesions in NC/Nga mice induced by repeated application of picryl chloride (PiCl) under specific pathogen-free (SPF) conditions. Oral administration of RJ to the PiCl-treated NC/Nga mice inhibited the development of AD-like skin lesions in these mice as exemplified by the significant decrease in the total skin severity scores and the decrease in hypertrophy, hyperkeratosis, and infiltration of the epidermis and corium by inflammatory cells. IFN-gamma production by spleen cells from PiCl-treated NC/Nga mice in response to TNP-KLH was partially but significantly inhibited by the oral administration of RJ, while IFN-gamma production by Con A-stimulated spleen cells was not affected. Since inducible nitric oxide (NO) synthase (iNOS)-derived NO has been suggested as an important immunoregulatory mediator in inflammatory autoimmune diseases, we have also examined the expression of iNOS in the dorsal skin lesions of PiCl-treated NC/Nga mice. Interestingly, the expression of iNOS was significantly increased in the skin lesions of RJ-administered mice compared with those of control PBS-administered mice. Thus, our results suggest that RJ suppresses the development of AD-like skin lesions in PiCl-treated NC/Nga mice, possibly by a combination of down-regulating TNP-specific IFN-gamma production and up-regulating iNOS expression.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/therapeutic use
- Administration, Oral
- Animals
- Dermatitis, Atopic/chemically induced
- Dermatitis, Atopic/genetics
- Dermatitis, Atopic/pathology
- Dermatitis, Atopic/prevention & control
- Dermatitis, Contact/etiology
- Dermatitis, Contact/genetics
- Dermatitis, Contact/pathology
- Dermatitis, Contact/prevention & control
- Disease Models, Animal
- Drug Eruptions/etiology
- Drug Eruptions/genetics
- Drug Eruptions/pathology
- Drug Eruptions/prevention & control
- Drug Evaluation, Preclinical
- Fatty Acids/administration & dosage
- Fatty Acids/therapeutic use
- Female
- Haptens/toxicity
- Hypertrophy
- Immunoglobulin E/blood
- Immunoglobulin E/immunology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Mice
- Mice, Mutant Strains
- Nitric Oxide Synthase/biosynthesis
- Nitric Oxide Synthase Type II
- Picryl Chloride/toxicity
- Skin/drug effects
- Skin/immunology
- Skin/pathology
- Specific Pathogen-Free Organisms
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
Collapse
Affiliation(s)
- Yoshifumi Taniguchi
- Fujisaki Institute, Hayashibara Biochemical Laboratories, Inc, Fujisaki 675-1, Okayama 702-8006, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kahl KG, Zielasek J, Uttenthal LO, Rodrigo J, Toyka KV, Schmidt HHHW. Protective role of the cytokine-inducible isoform of nitric oxide synthase induction and nitrosative stress in experimental autoimmune encephalomyelitis of the DA rat. J Neurosci Res 2003; 73:198-205. [PMID: 12836162 DOI: 10.1002/jnr.10649] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The pathogenic role of nitric oxide (NO) in multiple sclerosis (MS) remains controversial. Some groups have reported a pathogenic role of NO in experimental autoimmune encephalomyelitis (EAE), an animal model of some aspects of MS, whereas we and others have found a disease-suppressive effect of NO in EAE. Because the previously used EAE models have a mainly monophasic inflammatory disease course, distinct from MS, we here studied EAE in the DA rat, which better models the demyelinating and relapsing disease course of human MS. The induction of EAE in DA rats led to 1) severe inflammatory infiltrates mainly in the lumbar spinal cord; 2) an up-regulation of the activity of the cytokine-inducible isoform of NO synthases (NOS-II); and 3) increased tissue protein tyrosine nitration, as indicated by peroxynitrite (ONOO(-)), as a marker of nitrosative stress. Sources of superoxide metabolism, i.e., NADPH oxidase, myeloperoxidase, and superoxide dismutase, remained unchanged. Early treatment of animals with aminoguanidine, a relatively selective inhibitor of NOS-II, lowered nitrotyrosine immunoreactivity but at the same time led to more severe disease and pronounced inflammatory infiltrates in the lumbar spinal cord. Our results suggest a rather protective role of NOS-II induction and nitrosative stress in EAE in DA rats and support the hypothesis of a disease-mitigating immunomodulatory role of NO in this animal model of MS.
Collapse
Affiliation(s)
- Kai G Kahl
- Department of Neurology, Julius-Maximilians-University, Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Staykova MA, Berven LA, Cowden WB, Willenborg DO, Crouch MF. Nitric oxide induces polarization of actin in encephalitogenic T cells and inhibits their in vitro trans-endothelial migration in a p70S6 kinase-independent manner. FASEB J 2003; 17:1337-9. [PMID: 12759332 DOI: 10.1096/fj.02-0577fje] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Nitric oxide (NO) inhibits both actively induced and transferred autoimmune encephalomyelitis. To explore potential mechanisms, we examined the ability of NO to inhibit migration of T lymphoblasts through both collagen matrices and monolayers of rat brain endothelial cells. The NO donor 1-hydroxy-2-oxo-3, 3-bis (2-aminoethyl)-1-triazene (HOBAT) inhibited migration in a concentration-dependent manner. NO pretreatment of T cells inhibited migration through untreated endothelial cells, but NO pretreatment of endothelial cells had no inhibitory effect on untreated T cells. Therefore NO's migration inhibitory action was mediated through its effect on T cells and not endothelial cells. HOBAT did not inhibit migration by inducing T-cell death but rather by polarizing the T cells, resulting in a morphology suggestive of migrating cells. P70S6 kinase, shown to have a role in NO-induced migration inhibition in fibroblasts, had no role in the inhibitory effect of NO on T-cell migration. Thus, HOBAT did not alter p70S6K activity nor did rapamycin, a specific inhibitor of p70S6K, inhibit HOBAT-induced T-cell morphological changes or T-cell migration. We suggest that NO-induced morphological changes result in T cells with predefined migratory directionality, thus limiting the ability of these cells to respond to other migratory signals.
Collapse
|
30
|
Djerbi M, Abdul-Majid KB, Abedi-Valugerdi M, Olsson T, Harris RA, Grandien A. Expression of the long form of human FLIP by retroviral gene transfer of hemopoietic stem cells exacerbates experimental autoimmune encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:2064-73. [PMID: 12574377 DOI: 10.4049/jimmunol.170.4.2064] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Subsidence of inflammation and clinical recovery in experimental autoimmune encephalomyelitis (EAE) is postulated to involve apoptosis of inflammatory cells. To test this concept, we examined the effects of overexpressing the long form of human FLICE-inhibitory protein, a potent inhibitor of death receptor-mediated apoptosis, in myelin oligodendrocyte glycoprotein-induced EAE in DBA/1 mice. We found that overexpression of the long form of human FLICE-inhibitory protein by retroviral gene transfer of hemopoietic stem cells led to a clinically more severe EAE in these mice compared with control mice receiving the retroviral vector alone. The exacerbated disease was evident by an enhanced and prolonged inflammatory reaction in the CNS of these animals compared with control mice. The acute phase of EAE was characterized by a massive infiltration of macrophages and granulocytes and a simultaneous increase in TNF-alpha production in the CNS. In the chronic phase of the disease, there was a prolonged inflammatory response in the form of persistent CD4(+) T and B cells in the CNS and a peripheral Th1 cytokine bias caused by elevated levels of IFN-gamma and reduced levels of IL-4 in the spleen. Our findings demonstrate that death receptor-mediated apoptosis can be important in the pathogenesis of EAE and further emphasize the need for effective apoptotic elimination of inflammatory cells to achieve disease remission.
Collapse
MESH Headings
- Acute Disease
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/toxicity
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- Bone Marrow Cells/immunology
- CASP8 and FADD-Like Apoptosis Regulating Protein
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Carrier Proteins/toxicity
- Cells, Cultured
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Chronic Disease
- Cytokines/biosynthesis
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Genetic Vectors
- Growth Inhibitors/genetics
- Growth Inhibitors/physiology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Hematopoietic Stem Cell Transplantation/methods
- Humans
- Inflammation/genetics
- Inflammation/immunology
- Intracellular Signaling Peptides and Proteins
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred DBA
- Myelin Proteins
- Myelin-Associated Glycoprotein/administration & dosage
- Myelin-Associated Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- Protein Isoforms/toxicity
- Retroviridae/genetics
- Retroviridae/immunology
- Stem Cells/immunology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Transduction, Genetic
- Tumor Necrosis Factor-alpha/biosynthesis
- fas Receptor/physiology
Collapse
Affiliation(s)
- Mounira Djerbi
- Department of Immunology, Wenner-Gren Institute, University of Stockholm, S-10691 Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
31
|
Ochi H, Osoegawa M, Wu XM, Minohara M, Horiuchi I, Murai H, Furuya H, Kira JI. Increased IL-13 but not IL-5 production by CD4-positive T cells and CD8-positive T cells in multiple sclerosis during relapse phase. J Neurol Sci 2002; 201:45-51. [PMID: 12163193 DOI: 10.1016/s0022-510x(02)00189-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the present study, we flow cytometrically analyzed the intracellular production of interleukin (IL)-5 and IL-13 in peripheral blood CD4(+) and CD8(+) T cells from patients with multiple sclerosis (MS), human T-lymphotropic virus type I-associated myelopathy/tropical spastic paraparesis (HAM/TSP) and healthy controls. IL-13-producing T cells were significantly increased in both T cell subsets in MS at relapse, markedly in the conventional form of MS and modestly in the optocospinal form of MS, and returned to normal at remission. However, IL-5-producing T cells did not vary regardless of clinical phase or type. HAM/TSP showed no significant change in the number of IL-5- and IL-13-producing cells. A distinct profile of IL-13 and IL-5 production by disease and by phase of MS suggests an active involvement of these type 2 cytokines in central nervous system (CNS) inflammation.
Collapse
Affiliation(s)
- Hirofumi Ochi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Nitric oxide (NO) is a free radical found at higher than normal concentrations within inflammatory multiple sclerosis (MS) lesions. These high concentrations are due to the appearance of the inducible form of nitric oxide synthase (iNOS) in cells such as macrophages and astrocytes. Indeed, the concentrations of markers of NO production (eg, nitrate and nitrite) are raised in the CSF, blood, and urine of patients with MS. Circumstantial evidence suggests that NO has a role in several features of the disease, including disruption of the blood-brain barrier, oligodendrocyte injury and demyelination, axonal degeneration, and that it contributes to the loss of function by impairment of axonal conduction. However, despite these considerations, the net effect of NO production in MS is not necessarily deleterious because it also has several beneficial immunomodulatory effects. These dual effects may help to explain why iNOS inhibition has not provided reliable and encouraging results in animal models of MS, but alternative approaches based on the inhibition of superoxide production, partial sodium-channel blockade, or the replacement of lost immunomodulatory function, may prove beneficial.
Collapse
Affiliation(s)
- Kenneth J Smith
- Neuroinflammation Research Group, Guy's, King's, and St Thomas' School of Medicine, King's College, London SE1 1UL, UK.
| | | |
Collapse
|
33
|
Mäurer M, Toyka KV, Gold R. Immune mechanisms in acquired demyelinating neuropathies: lessons from animal models. Neuromuscul Disord 2002; 12:405-14. [PMID: 12062260 DOI: 10.1016/s0960-8966(01)00302-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The peripheral nervous system (PNS) is the target for a heterogenous immune attack mediated by T-cells, B-cells, and macrophages. The interaction of the humoral and cellular immune system with the structural components in the peripheral nervous system may determine the extent of inflammation and possibly repair mechanisms. The animal model experimental autoimmune neuritis (EAN) allows detailed study of the various effector pathways and tests novel therapeutic strategies in vivo. Unexpectedly, involvement of the immune system is also found in animal models for inherited neuropathies and in its human counterpart Charcot-Marie-Tooth (CMT) disease, suggesting an autoimmune reaction triggered by the genetically determined demyelinating disorder. A better understanding of immune regulation and its failure in the peripheral nervous system may help to develop more specific and more effective immunotherapies.
Collapse
Affiliation(s)
- Mathias Mäurer
- Department of Neurology, Section of Developmental Neurobiology, Julius-Maximilians-Universität, Würzburg, Germany
| | | | | |
Collapse
|
34
|
Abstract
Reactive oxygen intermediates (ROI) and nitric oxide (NO(.)) are produced in abundance in the inflammatory muscle diseases of autoimmune origin polymyositis (PM), dermatomyositis (DM), and inclusion body myositis (IBM). However, their role in the pathogenesis of these diseases is so far not clear. In contrast to demyelinating neuropathies, there is no convincing evidence for oxide-induced apoptosis either in myocytes or in lymphocytes and phagocytes in inflammatory myopathies. On the contrary, NO(.) released at low concentrations at target sites may even have cell-protective effects. A major mechanism of protection from apoptosis in both myocytes and inflammatory cells seems to be the upregulation of anti-apoptotic proteins like Bcl-2. Caution is warranted to apply antioxidative and anti-apoptotic agents to patients with inflammatory myopathies as long as the pathogenic role of oxides and apoptosis in the individual case is not resolved.
Collapse
Affiliation(s)
- M Stangel
- Department of Neurology, Universitätsklinikum Benjamin Franklin, Free University Berlin, D-12200 Berlin, Germany
| | | | | | | |
Collapse
|
35
|
Pender MP, Rist MJ. Apoptosis of inflammatory cells in immune control of the nervous system: role of glia. Glia 2001; 36:137-44. [PMID: 11596122 DOI: 10.1002/glia.1103] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The elimination of inflammatory cells within the central nervous system (CNS) by apoptosis plays an important role in protecting the CNS from immune-mediated damage. T cells, B cells, macrophages, and microglia all undergo apoptosis in the CNS. The apoptotic elimination of CNS-reactive T cells is particularly important, as these cells can recruit and activate other inflammatory cells. T-cell apoptosis contributes to the resolution of CNS inflammation and clinical recovery from attacks of experimental autoimmune encephalomyelitis (EAE), an animal model of the demyelinating disease multiple sclerosis (MS). T-cell apoptosis in the CNS in EAE occurs in both an antigen-specific and an antigen-nonspecific manner. In antigen-specific T-cell apoptosis, it is proposed that T cells that recognize their antigen in the CNS, such as CNS-reactive T cells, are deleted by the process of activation-induced apoptosis after activation of the T-cell receptor. This may result from the ligation of T-cell death receptors (such as CD95 (Fas) or tumor necrosis factor (TNF) receptor 1) by CD95 ligand (CD95L) or TNF expressed by the same T cell or possibly by microglia, astrocytes or neurons. Inadequate costimulation of the T cell by antigen-presenting glial cells may render T cells susceptible to activation-induced apoptosis. T cells expressing CD95 may also die in an antigen-nonspecific manner after interacting with glial cells expressing CD95L. Other mechanisms for antigen-nonspecific T-cell apoptosis include the endogenous release of glucocorticosteroids, deprivation of interleukin-2, and the release of nitric oxide by macrophages or glia. Apoptosis of autoreactive T cells in the CNS is likely to be important in preventing the development of autoimmune CNS diseases such as MS.
Collapse
Affiliation(s)
- M P Pender
- Department of Medicine, University of Queensland, Brisbane, Australia
| | | |
Collapse
|
36
|
Vairetti M, Griffini P, Pietrocola G, Richelmi P, Freitas I. Cold-induced apoptosis in isolated rat hepatocytes: protective role of glutathione. Free Radic Biol Med 2001; 31:954-61. [PMID: 11595380 DOI: 10.1016/s0891-5849(01)00670-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Liver conservation for transplantation is usually made at 2-4 degrees C. We studied the effect of rewarming to 37 degrees C for up to 3 h of rat hepatocytes kept at 4 degrees C for 20 h, modulating intracellular glutathione (GSH) concentration either with a GSH precursor (N-acetyl-L-cysteine, NAC), or with GSH depleting agents (diethylmaleate and buthionine sulfoximine, DEM/BSO). Untreated hepatocytes showed time-dependent production of reactive oxygen species (ROS), lipid peroxidation, chromatin condensation and membrane blebbing, decrease in GSH concentration, and protein sulfhydryl groups. Fluorochromatization with Propidium Iodide (PI) and Annexin V (AnxV) of cells rewarmed for 1 h caused an increase of AnxV-positive cells without PI staining and any observed lactate dehydrogenase leakage. TUNEL and DNA-laddering tests were negative for all times and treatments, indicating that apoptosis may occur without DNA fragmentation. Cold preservation and rewarming in the presence of NAC induced a significant improvement in the morphology, less oxidative stress and apoptosis. Conversely, DEM/BSO caused a marked deterioration of morphology, increase of oxidative stress and apoptosis. These results suggested that marked changes in GSH status might play a critical role in triggering apoptosis during cold preservation of isolated rat hepatocytes. NAC, added before rewarming, might represent a therapeutic approach for preventing the early events of apoptosis during cold storage.
Collapse
Affiliation(s)
- M Vairetti
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy.
| | | | | | | | | |
Collapse
|
37
|
Hancock JT, Desikan R, Neill SJ. Does the redox status of cytochrome C act as a fail-safe mechanism in the regulation of programmed cell death? Free Radic Biol Med 2001; 31:697-703. [PMID: 11522455 DOI: 10.1016/s0891-5849(01)00646-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
It has now become recognized that one of the key events in the induction of apoptosis, or programmed cell death, in both plants and animals is the release of cytochrome c from mitochondria. It is also known that oxidative stress imposed on cells can have a profound effect on the onset or progression of apoptosis. Here, we discuss how the redox status of cytochrome c, and thus its structure, can be altered by the presence of reactive oxygen species (ROS) and reduced glutathione (GSH). We suggest that cytochrome c will only induce programmed cell death if present in the cytoplasm in the oxidized state, and that the presence of high levels of cytoplasmic GSH maintain cytochrome c in an inactive (reduced) state, thus behaving as a fail-safe mechanism if cytochrome c is released by mitochondria when programmed cell death is not the required outcome. If the redox status of the cell is disturbed however, perhaps in the presence of hydrogen peroxide, GSH concentrations will drop, the cellular E(h) will rise, and cytochrome c will tend towards the oxidized state, allowing programmed cell death to proceed. Therefore, we propose that the redox state of cytoplasmic cytochrome c may be a key regulator of programmed cell death.
Collapse
Affiliation(s)
- J T Hancock
- Centre for Research in Plant Science, Faculty of Applied Science, University of the West of England, Bristol, UK.
| | | | | |
Collapse
|
38
|
Xiao BG, Huang YM, Yang JS, Xu LY, Link H. Bone marrow-derived dendritic cells from experimental allergic encephalomyelitis induce immune tolerance to EAE in Lewis rats. Clin Exp Immunol 2001; 125:300-9. [PMID: 11529923 PMCID: PMC1906114 DOI: 10.1046/j.1365-2249.2001.01573.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have previously shown that dendritic cells (DC), upon being pulsed in vitro with encephalitogenic myelin basic protein peptide 68-86 (MBP 68-86) and injected subcutaneously (s.c.) back to healthy Lewis rats, transfer immune tolerance to experimental allergic encephalomyelitis (EAE) induced by immunization with MBP 68-86 and Freund's complete adjuvant (FCA). We here assumed that DC become pulsed in EAE rats, and that expansion in vitro of such 'in vivo pulsed EAE-DC' might also have the capacity to induce immune tolerance to EAE, thereby eliminating the need for in vitro pulsing of DC with autoantigens which are still unknown in many autoimmune diseases in the human. In the present study, EAE-DC were generated from bone marrow of Lewis rats, with EAE induced with MBP 68-86 + FCA, and expanded in vitro by culture with GM-CSF and IL-4. In comparison with DC from normal rats, EAE-DC exhibited higher viability in the absence of growth factors, and presented specific antigen to naïve T cells in vitro. The DC derived from both EAE and healthy rats stimulated strong proliferation in an antigen-independent manner, lasting for 4 weeks after DC were s.c. injected into healthy rats. During this time, injection of EAE-DC did not induce clinical EAE. However, when these rats were immunized with MBP 68-86 + FCA, subsequent EAE was dramatically suppressed, and was associated with increased IFN-gamma expression, nitric oxide production, gradually reduced proliferation and cell apoptosis, compared with PBS-injected control EAE rats. LPS-treated DC did not induce tolerance, suggesting that the tolerance is mediated by an immature stage of DC. These observations support the hypothesis that EAE-DC can transfer immune tolerance to EAE, thereby omitting the step of characterizing specific autoantigen. Omitting the step of loading DC with antigen not only eliminates the extremely complex procedure of defining pathogenically-relevant autoantigens, but also avoids the risk of inducing immunogenicity of DC in the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- B G Xiao
- Experimental Neurology and Neuroimmunology Units, Division of Neurology, Huddinge University Hospital, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Steven Zeisel
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
40
|
Kiefer R, Kieseier BC, Stoll G, Hartung HP. The role of macrophages in immune-mediated damage to the peripheral nervous system. Prog Neurobiol 2001; 64:109-27. [PMID: 11240209 DOI: 10.1016/s0301-0082(00)00060-5] [Citation(s) in RCA: 220] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Macrophage-mediated segmental demyelination is the pathological hallmark of autoimmune demyelinating polyneuropathies, including the demyelinating form of Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy. Macrophages serve a multitude of functions throughout the entire pathogenetic process of autoimmune neuropathy. Resident endoneurial macrophages are likely to act as local antigen-presenting cells by their capability to express major histocompatibility complex antigens and costimulatory B7-molecules, and may thus be critical in triggering the autoimmune process. Hematogenous infiltrating macrophages then find their way into the peripheral nerve together with T-cells by the concerted action of adhesion molecules, matrix metalloproteases and chemotactic signals. Within the nerve, macrophages regulate inflammation by secreting several pro-inflammatory cytokines including IL-1, IL-6, IL-12 and TNF-alpha. Autoantibodies are likely to guide macrophages towards their myelin or primarily axonal targets, which then attack in a complement-dependent and receptor-mediated manner. In addition, non-specific tissue damage occurs through the secretion of toxic mediators and cytokines. Later, macrophages contribute to the termination of inflammation by promoting T-cell apoptosis and expressing anti-inflammatory cytokines including TGF-beta1 and IL-10. During recovery, they are tightly involved in allowing Schwann cell proliferation, remyelination and axonal regeneration to proceed. Macrophages, thus, play dual roles in autoimmune neuropathy, being detrimental in attacking nervous tissue but also salutary, when aiding in the termination of the inflammatory process and the promotion of recovery.
Collapse
Affiliation(s)
- R Kiefer
- Department of Neurology, Westfälische Wilhelms-Universität, Albert-Schweitzer-Strasse 33, D-48129 Münster, Germany.
| | | | | | | |
Collapse
|
41
|
Karp DR, Shimooku K, Lipsky PE. Expression of gamma-glutamyl transpeptidase protects ramos B cells from oxidation-induced cell death. J Biol Chem 2001; 276:3798-804. [PMID: 11080500 DOI: 10.1074/jbc.m008484200] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ectoenzyme, gamma-glutamyl transpeptidase (GGT, EC ) cleaves glutathione (GSH) to facilitate the recapture of cysteine for synthesis of intracellular GSH. The impact of GGT expression on cell survival during oxidative stress was investigated using the human B cell lymphoblastoid cell line, Ramos. Ramos cells did not express surface GGT and exhibited no GGT enzyme activity. In contrast, Ramos cells stably transfected with the human GGT cDNA expressed high levels of surface GGT and enzymatic activity. GGT-transfected Ramos cells were protected from apoptosis when cultured in cyst(e)ine-deficient medium. The GGT-expressing cells also had lower levels of intracellular reactive oxygen species (ROS). Homocysteic acid and alanine, inhibitors of cystine and cysteine uptake, respectively, caused increased ROS content and diminished viability of GGT expressing cells. Exogenous GSH increased the viability of the GGT-transfected cells more effectively than that of control cells, whereas the products of GSH metabolism prevented death of both the control and GGT-transfected cells comparably. These data indicate that GGT cleavage of GSH and the subsequent recapture of cysteine and cystine allow cells to maintain low levels of cellular ROS and thereby avoid apoptosis induced by oxidative stress.
Collapse
Affiliation(s)
- D R Karp
- Harold C. Simmons Arthritis Research Center, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| | | | | |
Collapse
|
42
|
Weishaupt A, Jander S, Brück W, Kuhlmann T, Stienekemeier M, Hartung T, Toyka KV, Stoll G, Gold R. Molecular mechanisms of high-dose antigen therapy in experimental autoimmune encephalomyelitis: rapid induction of Th1-type cytokines and inducible nitric oxide synthase. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:7157-63. [PMID: 11120847 DOI: 10.4049/jimmunol.165.12.7157] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
High-dose Ag administration induces apoptotic death of autoreactive T cells and is an effective therapy of experimental autoimmune diseases of the nervous system. To explore the role of cytokines in Ag-specific immunotherapy, we analyzed mRNA induction and protein expression for the proinflammatory cytokines TNF-alpha and IFN-gamma, the anti-inflammatory cytokine IL-10, and the cytokine-inducible NO synthase (iNOS) during high-dose Ag therapy of adoptive transfer experimental autoimmune encephalomyelitis (AT-EAE) in the Lewis rat. Using semiquantitative and competitive RT-PCR, we found 5- to 6-fold induction of TNF-alpha mRNA and 3-fold induction of IFN-gamma mRNA in the spinal cord that occurred within 1 h after i.v. injection of Ag and was accompanied by a 2-fold increase of iNOS mRNA. Both IFN-gamma and iNOS mRNA remained elevated for at least 6 h, whereas TNF-alpha mRNA was already down-regulated 6 h after Ag injection. A comparable time course was found for circulating serum levels of TNF-alpha and IFN-gamma. IL-10 mRNA levels did not change significantly following Ag injection. Neutralization of TNF-alpha by anti-TNF-alpha antiserum in vivo led to a significant decrease in the rate of T cell and oligodendrocyte apoptosis induced by high-dose Ag administration, but did not change the beneficial clinical effect of Ag therapy. Our data suggest profound activation of proinflammatory but not of anti-inflammatory cytokine gene expression by high-dose Ag injection. Functionally, TNF-alpha contributes to increased apoptosis of both autoaggressive T cells and oligodendrocytes in the target organ and may thereby play a dual role in this model of Ag-specific therapy of CNS autoimmune diseases.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- Autoantigens/administration & dosage
- Autoantigens/therapeutic use
- Cell Movement/immunology
- Cytokines/biosynthesis
- Cytokines/blood
- Disease Progression
- Dose-Response Relationship, Immunologic
- Drug Administration Schedule
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Enzyme Induction/genetics
- Enzyme Induction/immunology
- Female
- Gene Expression Regulation/immunology
- Guinea Pigs
- Immune Sera/administration & dosage
- Injections, Intravenous
- Myelin Basic Protein/administration & dosage
- Myelin Basic Protein/immunology
- Myelin Basic Protein/therapeutic use
- Nitric Oxide Synthase/biosynthesis
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase Type II
- RNA, Messenger/biosynthesis
- Rats
- Rats, Inbred Lew
- Spinal Cord/enzymology
- Spinal Cord/immunology
- Spinal Cord/pathology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- A Weishaupt
- Department of Neurology, Neuroimmunology Branch and Clinical Research Group for Multiple Sclerosis, Julius-Maximilians Universität, Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Huang YM, Yang JS, Xu LY, Link H, Xiao BG. Autoantigen-pulsed dendritic cells induce tolerance to experimental allergic encephalomyelitis (EAE) in Lewis rats. Clin Exp Immunol 2000; 122:437-44. [PMID: 11122252 PMCID: PMC1905816 DOI: 10.1046/j.1365-2249.2000.01398.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dendritic cells (DC) can modulate the nature of immune responses in a stimulatory or tolerogenic fashion. Great attention has been given to the induction of immunity to tumour and infection. In this study, bone marrow-derived DC from healthy Lewis rats were pulsed in vitro with encephalitogenic myelin basic protein peptide 68-86 (MBP 68-86), and injected subcutaneously (1 x 106/rat) into normal Lewis rats. Upon observation of the rats pretreated in this way for 4 weeks, when no clinical signs of EAE occurred, these rats were immunized with MBP 68-86 and Freund's complete adjuvant. The pretreated rats failed to develop clinical EAE. This tolerance was associated with augmented proliferative responses, interferon-gamma secretion, inducible nitric oxide synthase (iNOS) expression and NO production. The frequency of apoptotic cells was increased in the rats receiving MBP 68-86-pulsed DC compared with unpulsed control DC. Few infiltrating inflammatory cells were observed in spinal cord sections from rats that had received MBP 68-86-pulsed DC. The data are compatible with the interpretation that MBP 68-86-pulsed DC induce tolerance to EAE possibly through up-regulation of iNOS expression and NO production, which mediate cell apoptosis, thereby reducing infiltration of inflammatory cells within the central nervous system.
Collapse
Affiliation(s)
- Y M Huang
- Experimental Neurology and Neuroimmunology Units, Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
44
|
Schluesener HJ, Seid K. Heme oxygenase-1 in lesions of rat experimental autoimmune encephalomyelitis and neuritis. J Neuroimmunol 2000; 110:114-20. [PMID: 11024540 DOI: 10.1016/s0165-5728(00)00352-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The enzyme heme oxygenase-1 (HO-1) is reducing heme to the gaseous mediator carbon monoxide, to iron and the antioxidant biliverdin. The inducible expression of HO-1 is considered a protective cellular mechanism against reactive oxygen intermediates. Further, carbon monoxide (CO) is a regulator of cGMP synthesis, of NO-synthetases and cyclooxygenases, thereby indirectly modulating reactive processes. Here we report expression of HO-1 in rat experimental autoimmune encephalomyelitis (EAE) and neuritis (EAN). With both models, similar results were obtained: HO-1 was localized predominantly to infiltrating, monocytic, but only rarely to ramified microglial cells or astrocytes surrounding the inflammatory lesions. Prominent expression by monocytic cells was seen from day 11 after immunization correlating with the development of neurologic disease. Further, local expression is persistent for long after cessation of neurologic signs. Thus, HO-1 could be considered a factor in the formation and resolution of inflammatory autoimmune lesions of the nervous system.
Collapse
Affiliation(s)
- H J Schluesener
- Institute of Brain Research, University of Tübingen, Calwer Str. 3, D-72076, Tübingen, Germany.
| | | |
Collapse
|
45
|
Young TE, Gallie DR. Programmed cell death during endosperm development. PLANT MOLECULAR BIOLOGY 2000; 44:283-301. [PMID: 11199389 DOI: 10.1007/978-94-010-0934-8_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The endosperm of cereals functions as a storage tissue in which the majority of starch and seed storage proteins are synthesized. During its development, cereal endosperm initiates a cell death program that eventually affects the entire tissue with the exception of the outermost cells, which differentiate into the aleurone layer and remain living in the mature seed. To date, the cell death program has been described for maize and wheat endosperm, which exhibits common and unique elements for each species. The progression of endosperm programmed cell death (PCD) in both species is accompanied by an increase in nuclease activity and the internucleosomal degradation of nuclear DNA, hallmarks of apoptosis in animals. Moreover, ethylene and abscisic acid are key to mediating PCD in cereal endosperm. The progression of the cell death program in developing maize endosperm follows a highly organized pattern whereas in wheat endosperm, PCD initiates stochastically. Although the essential characteristics of cereal endosperm PCD are now known, the molecular mechanisms responsible for its execution remain to be identified.
Collapse
Affiliation(s)
- T E Young
- Department of Biochemistry, University of California, Riverside 92521-0129, USA.
| | | |
Collapse
|
46
|
Chu CQ, Wittmer S, Dalton DK. Failure to suppress the expansion of the activated CD4 T cell population in interferon gamma-deficient mice leads to exacerbation of experimental autoimmune encephalomyelitis. J Exp Med 2000; 192:123-8. [PMID: 10880533 PMCID: PMC1887710 DOI: 10.1084/jem.192.1.123] [Citation(s) in RCA: 342] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mice deficient in interferon (IFN)-gamma or IFN-gamma receptor develop progressive and fatal experimental autoimmune encephalomyelitis (EAE). We demonstrate that CD4 T cells lacking IFN-gamma production were required to passively transfer EAE, indicating that they were disease-mediating cells in IFN-gamma knockout (KO) mice. IFN-gamma KO mice accumulated 10-16-fold more activated CD4 T cells (CD4(+)CD44(hi)) than wild-type mice in the central nervous system during EAE. CD4(+)CD44(hi) T cells in the spleen and central nervous system of IFN-gamma KO mice during EAE showed markedly increased in vivo proliferation and significantly decreased ex vivo apoptosis compared with those of wild-type mice. IFN-gamma KO CD4(+)CD44(hi) T cells proliferated extensively to antigen restimulation in vitro and accumulated larger numbers of live CD4(+) CD44(hi) T cells. IFN-gamma completely suppressed proliferation and significantly induced apoptosis of CD4(+)CD44(hi) T cells responding to antigen and hence inhibited accumulation of live, activated CD4 T cells. We thus present novel in vivo and in vitro evidence that IFN-gamma may limit the extent of EAE by suppressing expansion of activated CD4 T cells.
Collapse
Affiliation(s)
- Cong-Qiu Chu
- From The Trudeau Institute, Saranac Lake, New York 12983
| | - Susan Wittmer
- From The Trudeau Institute, Saranac Lake, New York 12983
| | | |
Collapse
|
47
|
Petitto JM, Streit WJ, Huang Z, Butfiloski E, Schiffenbauer J. Interleukin-2 gene deletion produces a robust reduction in susceptibility to experimental autoimmune encephalomyelitis in C57BL/6 mice. Neurosci Lett 2000; 285:66-70. [PMID: 10788709 DOI: 10.1016/s0304-3940(00)00996-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dysregulation of interleukin-2 (IL-2), the prototypical T cell growth factor and immunoregulatory cytokine, may modify self-tolerance and predisposition to autoimmunity. The available literature suggested that IL-2 could be hypothesized to either propagate or inhibit the development autoimmune demyelinating disorders of the central nervous system such as multiple sclerosis. Thus, the present study sought to test these competing hypotheses by examining whether disrupting one or both IL-2 gene alleles would render mice more or less vulnerable to experimental autoimmune encephalomyelitis (EAE). Myelin oligodendrocyte glycoprotein was used to induce EAE in C57BL/6-IL-2(-/-) knockout, C57BL/6-IL-2(+/-) heterozygote and C57BL/6-IL-2(+/+) wild-type mice. All of the wild-type and heterozygote mice developed signs of EAE compared with only 23% of the IL-2 knockout mice. Histopathological examination of lumbar spinal cord sections confirmed that subpial perivascular inflammatory infiltrates found in wild-type and heterozygote mice were absent in the unaffected IL-2 knockout mice. These data demonstrate that vulnerability to EAE is markedly reduced in C57BL/6 mice lacking IL-2, and suggest that this cytokine may play a critical role in autoimmune processes of the central nervous system.
Collapse
Affiliation(s)
- J M Petitto
- Department of Psychiatry, UF Brain Institute, University of Florida College of Medicine, Gainesville, FL 32610-0256, USA.
| | | | | | | | | |
Collapse
|
48
|
Jin YX, Xu LY, Guo H, Ishikawa M, Link H, Xiao BG. TGF-beta1 inhibits protracted-relapsing experimental autoimmune encephalomyelitis by activating dendritic cells. J Autoimmun 2000; 14:213-20. [PMID: 10756083 DOI: 10.1006/jaut.2000.0364] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protracted-relapsing experimental autoimmune encephalomyelitis (PR-EAE) in DA rats is an animal model closely related to multiple sclerosis (MS). Previous studies showed that nasal administration of TGF-beta1 suppressed the development and relapse of PR-EAE clinically and pathologically. Here we demonstrate that this suppressive effect was associated with activation of dendritic cells (DC), showing elevated proliferative response and IFN-gamma and nitric oxide (NO) production by DC. DC derived from TGF-beta1-treated rats with PR-EAE also showed high expression of nitric oxide synthase 2 (NOS2) at both mRNA and protein levels. Apoptotic cells were increased in spleen sections of TGF-beta1-treated rats compared to control rats. In studying mechanisms of apoptosis in TGF-beta1-treated rats, in vitro experiments demonstrated that TGF-beta1-treated DC induced apoptosis of CD4(+)T cells by a NO pathway after co-culture with T cells. These results support the hypothesis that TGF-beta1-induced suppression of PR-EAE is associated with apoptosis of CD4(+)T cells induced by DC-derived NO.
Collapse
Affiliation(s)
- Y X Jin
- Division of Neurology, Units of Experimental Neurology and Neuroimmunology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
49
|
Wohlleben G, Hartung HP, Gold R. Humoral and cellular immune functions of cytokine-treated Schwann cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 468:151-6. [PMID: 10635026 DOI: 10.1007/978-1-4615-4685-6_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Affiliation(s)
- G Wohlleben
- Department of Neurology, University of Würzburg, Germany
| | | | | |
Collapse
|
50
|
|