51
|
Peripheral elevation of TNF-α leads to early synaptic abnormalities in the mouse somatosensory cortex in experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 2013; 110:10306-11. [PMID: 23733958 DOI: 10.1073/pnas.1222895110] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Sensory abnormalities such as numbness and paresthesias are often the earliest symptoms in neuroinflammatory diseases including multiple sclerosis. The increased production of various cytokines occurs in the early stages of neuroinflammation and could have detrimental effects on the central nervous system, thereby contributing to sensory and cognitive deficits. However, it remains unknown whether and when elevation of cytokines causes changes in brain structure and function under inflammatory conditions. To address this question, we used a mouse model for experimental autoimmune encephalomyelitis (EAE) to examine the effect of inflammation and cytokine elevation on synaptic connections in the primary somatosensory cortex. Using in vivo two-photon microscopy, we found that the elimination and formation rates of dendritic spines and axonal boutons increased within 7 d of EAE induction--several days before the onset of paralysis--and continued to rise during the course of the disease. This synaptic instability occurred before T-cell infiltration and microglial activation in the central nervous system and was in conjunction with peripheral, but not central, production of TNF-α. Peripheral administration of a soluble TNF inhibitor prevented abnormal turnover of dendritic spines and axonal boutons in presymptomatic EAE mice. These findings indicate that peripheral production of TNF-α is a key mediator of synaptic instability in the primary somatosensory cortex and may contribute to sensory and cognitive deficits seen in autoimmune diseases.
Collapse
|
52
|
Development of experimental autoimmune encephalomyelitis critically depends on CD137 ligand signaling. J Neurosci 2013; 32:18246-52. [PMID: 23238738 DOI: 10.1523/jneurosci.2473-12.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Multiple sclerosis (MS) is a degenerative autoimmune disease of the CNS. Experimental autoimmune encephalomyelitis (EAE) is a commonly used murine model for MS. Here we report that CD137 ligand (CD137L, 4-1BB ligand, TNFS9), a member of the TNF superfamily, is critical for the development of EAE. EAE symptoms were significantly ameliorated in CD137L(-/-) mice. In the absence of CD137L, myelin oligodendrocyte glycoprotein (MOG)-specific T-cells secreted lower levels of T(h)1/T(h)17 cell-associated cytokines. MOG-specific T-cells also trafficked less efficiently to the CNS in CD137L(-/-) mice, possibly as a consequence of reduced expression of vascular cell adhesion molecule-1 (VCAM-1), which regulates leukocyte extravasation. Thus, CD137L regulates many functions of MOG-specific T-cells that contribute to EAE and may represent a novel therapeutic target for the treatment of MS.
Collapse
|
53
|
|
54
|
Abstract
Demyelinating diseases such as multiple sclerosis are chronic inflammatory autoimmune diseases with a heterogeneous clinical presentation and course. Both the adaptive and the innate immune systems have been suggested to contribute to their pathogenesis and recovery. In this review, we discuss the role of the innate immune system in mediating demyelinating diseases. In particular, we provide an overview of the anti-inflammatory or pro-inflammatory functions of dendritic cells, mast cells, natural killer (NK) cells, NK-T cells, γδ T cells, microglial cells, and astrocytes. We emphasize the interaction of astroctyes with the immune system and how this interaction relates to the demyelinating pathologies. Given the pivotal role of the innate immune system, it is possible that targeting these cells may provide an effective therapeutic approach for demyelinating diseases.
Collapse
Affiliation(s)
- Lior Mayo
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
55
|
Comparative spatiotemporal analysis of the intrathecal immune response in natural listeric rhombencephalitis of cattle and small ruminants. Comp Immunol Microbiol Infect Dis 2012; 35:429-41. [DOI: 10.1016/j.cimid.2012.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 03/20/2012] [Accepted: 03/28/2012] [Indexed: 12/29/2022]
|
56
|
Tayebati SK, Tomassoni D, Amenta F. Spontaneously hypertensive rat as a model of vascular brain disorder: microanatomy, neurochemistry and behavior. J Neurol Sci 2012; 322:241-9. [PMID: 22726353 DOI: 10.1016/j.jns.2012.05.047] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 05/23/2012] [Indexed: 11/28/2022]
Abstract
Arterial hypertension is the main risk factor for stroke and plays a role in the development of vascular cognitive impairment (VCI) and vascular dementia (VaD). An association between hypertension and reduced cerebral blood flow and VCI is documented and arterial hypertension in midlife is associated with a higher probability of cognitive impairment. These findings suggest that arterial hypertension is a main cause of vascular brain disorder (VBD). Spontaneously hypertensive rat (SHR) is the rat strain most extensively investigated and used for assessing hypertensive brain damage and treatment of it. They are normotensive at birth and at 6months they have a sustained hypertension. Time-dependent rise of arterial blood pressure, the occurrence of brain atrophy, loss of nerve cells and glial reaction are phenomena shared to some extent with hypertensive brain damage in humans. SHR present changes of some neurotransmitter systems that may have functional and behavioral relevance. An impaired cholinergic neurotransmission characterizes SHR, similarly as reported in patients affected by VaD. SHR are also characterized by a dopaminergic hypofunction and noradrenergic hyperactivity similarly as occurs in attention-deficit with hyperactivity disorder (ADHD). Microanatomical, neurochemical and behavioral data on SHR are in favor of the hypothesis that this strain is a suitable model of VBD. Changes in catecholaminergic transmission put forward SHR as a possible model of ADHD as well. Hence SHR could represent a multi-faced model of two important groups of pathologies, VBD and ADHD. As for most models, researchers should always consider that SHR offer some similarities with corresponding human pathologies, but they do not suffer from the same disease. This paper reviews the main microanatomical, neurochemical and behavioral characteristics of SHR with particular reference as an animal model of brain vascular injury.
Collapse
Affiliation(s)
- Seyed Khosrow Tayebati
- School of Medicinal Sciences and Health Products, University of Camerino, Camerino, Italy.
| | | | | |
Collapse
|
57
|
Miljković D, Timotijević G, Stojković MM. Astrocytes in the tempest of multiple sclerosis. FEBS Lett 2011; 585:3781-8. [DOI: 10.1016/j.febslet.2011.03.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Revised: 03/21/2011] [Accepted: 03/23/2011] [Indexed: 12/11/2022]
|
58
|
Cruz-Orengo L, Holman DW, Dorsey D, Zhou L, Zhang P, Wright M, McCandless EE, Patel JR, Luker GD, Littman DR, Russell JH, Klein RS. CXCR7 influences leukocyte entry into the CNS parenchyma by controlling abluminal CXCL12 abundance during autoimmunity. ACTA ACUST UNITED AC 2011; 208:327-39. [PMID: 21300915 PMCID: PMC3039853 DOI: 10.1084/jem.20102010] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During CNS autoimmunity, brain endothelial cell CXCR7 internalizes CXCL12 from the perivascular space, thereby permitting leukocyte migration into the CNS parenchyma. Loss of CXCL12, a leukocyte localizing cue, from abluminal surfaces of the blood–brain barrier occurs in multiple sclerosis (MS) lesions. However, the mechanisms and consequences of reduced abluminal CXCL12 abundance remain unclear. Here, we show that activation of CXCR7, which scavenges CXCL12, is essential for leukocyte entry via endothelial barriers into the central nervous system (CNS) parenchyma during experimental autoimmune encephalomyelitis (EAE), a model for MS. CXCR7 expression on endothelial barriers increased during EAE at sites of inflammatory infiltration. Treatment with a CXCR7 antagonist ameliorated EAE, reduced leukocyte infiltration into the CNS parenchyma and parenchymal VCAM-1 expression, and increased abluminal levels of CXCL12. Interleukin 17 and interleukin 1β increased, whereas interferon-γ decreased, CXCR7 expression on and CXCL12 internalization in primary brain endothelial cells in vitro. These findings identify molecular requirements for the transvascular entry of leukocytes into the CNS and suggest that CXCR7 blockade may have therapeutic utility for the treatment of MS.
Collapse
Affiliation(s)
- Lillian Cruz-Orengo
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Rubio N, Sanz-Rodriguez F, Arevalo MA. Up-regulation of the vascular cell adhesion molecule-1 (VCAM-1) induced by Theiler's murine encephalomyelitis virus infection of murine brain astrocytes. ACTA ACUST UNITED AC 2010; 17:57-68. [PMID: 20825262 DOI: 10.3109/15419061.2010.507827] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The present article reports the up-regulation of the expression of the vascular cell adhesion molecule-1 (VCAM-1) by SJL/J mouse brain astrocytes infected with Theiler's murine encephalomyelitis virus (TMEV). Complementary RNA (cRNA) from mock- and TMEV-infected cells was hybridized to the Affymetrix whole murine genome U74v2 DNA microarray. Hybridization data analysis revealed background expression in untreated cells and the up-regulation of three sequences coding for VCAM-1, as described by the SCOP (Structural Classification Of Proteins) database. The authors further studied its regulation, confirming and validating their mRNA increase by reverse transcriptase-polymerase chain reaction (RT-PCR) and quantitative real-time RT-PCR. The presence of the 100-kDa VCAM-1 protein in mock- and TMEV-infected cells was demonstrated in the cell membrane by a specific cell-based enzyme-linked immunosorbent assay (ELISA), in addition to flow cytometry and confocal immunohistochemistry. Further, Western blots were used to quantify the amount of VCAM-1 molecules in cell extracts. All these data demonstrated a mean 75% increase in the expression of VCAM-1 on the surface of TMEV-infected cells. Three inflammatory cytokines, interleukin-1alpha (IL-1alpha), interferon gamma (IFNgumma), and specially tumor necrosis factor alpha (TNF-α), some of which are also induced by TMEV in astrocytes (IL-1alpha and TNF-alpha), were potent inducers of VCAM-1 expression. To demonstrate whether the VCAM-1 molecules were biologically active, mediating adhesion to other cells as the integrin alpha4-expressing CD4+ T lymphocytes, the authors used a cell adhesion test. It was also demonstrated by immunohistochemistry that in vivo VCAM-1 expression is enhanced after TMEV intracraneal infection. The present data show a small but statistically significant overexpression of VCAM-1 after astrocyte infection with TMEV that could play a significant role in vivo.
Collapse
|
60
|
Mc Guire C, Volckaert T, Wolke U, Sze M, de Rycke R, Waisman A, Prinz M, Beyaert R, Pasparakis M, van Loo G. Oligodendrocyte-specific FADD deletion protects mice from autoimmune-mediated demyelination. THE JOURNAL OF IMMUNOLOGY 2010; 185:7646-53. [PMID: 21068410 DOI: 10.4049/jimmunol.1000930] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Apoptosis of oligodendrocytes (ODCs), the myelin-producing glial cells in the CNS, plays a central role in demyelinating diseases such as multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. To investigate the mechanism behind ODC apoptosis in EAE, we made use of conditional knockout mice lacking the adaptor protein FADD specifically in ODCs (FADD(ODC-KO)). FADD mediates apoptosis by coupling death receptors with downstream caspase activation. In line with this, ODCs from FADD(ODC-KO) mice were completely resistant to death receptor-induced apoptosis in vitro. In the EAE model, FADD(ODC-KO) mice followed an ameliorated clinical disease course in comparison with control littermates. Lymphocyte and macrophage infiltration into the spinal cord parenchyma was significantly reduced, as was the extent of demyelination and proinflammatory gene expression. Collectively, our data show that FADD is critical for ODC apoptosis and the development of autoimmune demyelinating disease.
Collapse
Affiliation(s)
- Conor Mc Guire
- Unit of Molecular Signal Transduction in Inflammation, Department for Molecular Biomedical Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
|
62
|
Lees JR, Sim J, Russell JH. Encephalitogenic T-cells increase numbers of CNS T-cells regardless of antigen specificity by both increasing T-cell entry and preventing egress. J Neuroimmunol 2010; 220:10-6. [PMID: 20167381 DOI: 10.1016/j.jneuroim.2009.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 11/17/2009] [Accepted: 11/24/2009] [Indexed: 01/26/2023]
Abstract
This study utilized an adoptive transfer model of experimental autoimmune encephalomyelitis (EAE) induction in mice to characterize the mechanisms involved in CNS accumulation of transferred and host T-cells. Using a flow cytometric technique, we examined phenotypic characteristics of CNS T-cells following disease initiation and the role of T-cell activation in CNS invasion and retention. Host T-cell activation increased cell recruitment and EAE severity. CNS antigen specific T-cells were required to induce T-cell retention within the CNS. Once retention was initiated, CNS T-cells were retained regardless of specificity. This study characterizes mechanisms involved in CNS accumulation of T-cells during EAE pathogenesis.
Collapse
Affiliation(s)
- Jason R Lees
- Department of Surgery, 400 MSTF, 10 S. Pine St, University of Maryland School of Medicine, Baltimore 21201-1509, USA.
| | | | | |
Collapse
|
63
|
Noradrenaline reuptake inhibitors inhibit expression of chemokines IP-10 and RANTES and cell adhesion molecules VCAM-1 and ICAM-1 in the CNS following a systemic inflammatory challenge. J Neuroimmunol 2010; 220:34-42. [PMID: 20061033 DOI: 10.1016/j.jneuroim.2009.12.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 12/12/2009] [Accepted: 12/14/2009] [Indexed: 11/20/2022]
Abstract
Evidence suggests that noradrenaline has a tonic anti-inflammatory action in the central nervous system (CNS) via its ability to inhibit expression of inflammatory mediators from glial cells. Consequently it is suggested that noradrenaline may play an endogenous neuroprotective role in CNS disorders where inflammatory events contribute to pathology. Infiltration of peripheral immune cells into the brain is driven by increased chemokine and cell adhesion molecule (CAM) expression, and is known to exacerbate neuroinflammation and thereby contribute to the disease process in a number of neurodegenerative disease states. Here we demonstrate that treatment of rats with the noradrenaline reuptake inhibitors (NRIs) desipramine and atomoxetine, agents that increase extracellular noradrenaline in the CNS, suppressed chemokine and cell adhesion molecule (CAM) expression in rat brain following a systemic challenge with bacterial lipopolysaccharide (LPS). Specifically, these agents reduced expression of the chemokines, interferon-inducible protein-10 (IP-10, CXCL-10) and regulated upon activation normal T-cell expressed and secreted (RANTES, CCL-5), and the CAMs, vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule (ICAM-1) in cortex and hippocampus. The inhibitory action of NRIs on chemokines and CAM expression was mimicked by in vitro exposure of cultured glial cells to noradrenaline, but not to the NRIs themselves. These data indicate that the suppressive action of NRIs on chemokine and CAM expression that occurs in vivo is due to increased noradrenaline availability at glial cells, as opposed to a direct action of the drugs on glial cells per se. These results support the theory that noradrenaline has anti-inflammatory properties, and agents that increase noradrenaline availability in vivo can play a role in combating brain inflammation by reducing expression of chemokines and CAMs; molecules that facilitate leucocyte influx into the CNS.
Collapse
|
64
|
Therapeutic effect of PEGylated TNFR1-selective antagonistic mutant TNF in experimental autoimmune encephalomyelitis mice. J Control Release 2009; 149:8-14. [PMID: 20036293 DOI: 10.1016/j.jconrel.2009.12.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 12/07/2009] [Accepted: 12/16/2009] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease, the pathogenesis of which is related to elevated serum levels of tumor necrosis factor-α (TNF). Although anti-TNF therapy has been tested as a potential treatment for MS, no remission of symptoms was observed. Recent reports indicated that the TNFR1 signal was responsible for the pathogenesis of murine experimental autoimmune encephalomyelitis (EAE), while the TNFR2 signal was responsible for recovery of the pathogenesis of EAE. Therefore, selective blocking of TNFR1 appears to be a promising strategy for the treatment of MS. In this regard, we previously succeeded in developing a novel TNFR1-selective antagonistic TNF mutant (R1antTNF) by using phage display technology. Here, we have examined the therapeutic potential of R1antTNF using EAE mice. Treatment with PEGylated R1antTNF (PEG-R1antTNF) significantly improved the clinical score and cerebral demyelination at the onset of EAE. Considerable suppression of Th1 and Th17-type response was also observed in spleen and lymph node cells of mice given PEG-R1antTNF. Moreover, the administration of PEG-R1antTNF suppressed the infiltration of inflammatory cells containing Th1 and Th17 cells into the spinal cord. These results suggest that selective blocking of TNFR1 by PEG-R1antTNF could be an effective therapeutic strategy against MS.
Collapse
|
65
|
van Loo G, Sze M, Bougarne N, Praet J, Mc Guire C, Ullrich A, Haegeman G, Prinz M, Beyaert R, De Bosscher K. Antiinflammatory properties of a plant-derived nonsteroidal, dissociated glucocorticoid receptor modulator in experimental autoimmune encephalomyelitis. Mol Endocrinol 2009; 24:310-22. [PMID: 19965930 DOI: 10.1210/me.2009-0236] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Compound A (CpdA), a plant-derived phenyl aziridine precursor, was recently characterized as a fully dissociated nonsteroidal antiinflammatory agent, acting via activation of the glucocorticoid receptor, thereby down-modulating nuclear factor-kappaB-mediated transactivation, but not supporting glucocorticoid response element-driven gene expression. The present study demonstrates the effectiveness of CpdA in inhibiting the disease progress in experimental autoimmune encephalomyelitis (EAE), a well-characterized animal model of multiple sclerosis. CpdA treatment of mice, both early and at the peak of the disease, markedly suppressed the clinical symptoms of EAE induced by myelin oligodendrocyte glycoprotein peptide immunization. Attenuation of the clinical symptoms of EAE by CpdA was accompanied by reduced leukocyte infiltration in the spinal cord, reduced expression of inflammatory cytokines and chemokines, and reduced neuronal damage and demyelination. In vivo CpdA therapy suppressed the encephalogenicity of myelin oligodendrocyte glycoprotein peptide-specific T cells. Moreover, CpdA was able to inhibit TNF- and lipopolysaccharide-induced nuclear factor-kappaB activation in primary microglial cells in vitro, in a differential mechanistic manner as compared with dexamethasone. Finally, in EAE mice the therapeutic effect of CpdA, in contrast to that of dexamethasone, occurred in the absence of hyperinsulinemia and in the absence of a suppressive effect on the hypothalamic-pituitary-adrenal axis. Based on these results, we propose CpdA as a compound with promising antiinflammatory characteristics useful for therapeutic intervention in multiple sclerosis and other neuroinflammatory diseases.
Collapse
Affiliation(s)
- Geert van Loo
- Department for Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent University, B-9052 Ghent, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Ricci G, Volpi L, Pasquali L, Petrozzi L, Siciliano G. Astrocyte-neuron interactions in neurological disorders. J Biol Phys 2009; 35:317-36. [PMID: 19669420 PMCID: PMC2750745 DOI: 10.1007/s10867-009-9157-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2008] [Accepted: 04/15/2009] [Indexed: 12/12/2022] Open
Abstract
Astrocytes have long been considered as just providing trophic support for neurons in the central nervous system, but recently several studies have highlighted their importance in many functions such as neurotransmission, metabolite and electrolyte homeostasis, cell signaling, inflammation, and synapse modulation. Astrocytes are, in fact, part of a bidirectional crosstalk with neurons. Moreover, increasing evidence is stressing the emerging role of astrocyte dysfunction in the pathophysiology of neurological disorders, including neurodegenerative disease, stroke, epilepsy, migraine, and neuroinflammatory diseases.
Collapse
Affiliation(s)
- G Ricci
- Neurologic Clinic, University of Pisa, Pisa, Italy.
| | | | | | | | | |
Collapse
|
67
|
McCandless EE, Budde M, Lees JR, Dorsey D, Lyng E, Klein RS. IL-1R signaling within the central nervous system regulates CXCL12 expression at the blood-brain barrier and disease severity during experimental autoimmune encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:613-20. [PMID: 19535637 PMCID: PMC2892701 DOI: 10.4049/jimmunol.0802258] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the CNS characterized by disruption of the blood-brain barrier (BBB). This breach in CNS immune privilege allows undeterred trafficking of myelin-specific lymphocytes into the CNS where they induce demyelination. Although the mechanism of BBB compromise is not known, the chemokine CXCL12 has been implicated as a molecular component of the BBB whose pattern of expression is specifically altered during MS and which correlates with disease severity. The inflammatory cytokine IL-1beta has recently been shown to contribute not only to BBB permeability but also to the development of IL-17-driven autoimmune responses. Using experimental autoimmune encephalomyelitis, the rodent model of MS, we demonstrate that IL-1beta mediates pathologic relocation of CXCL12 during the induction phase of the disease, before the development of BBB disruption. We also show that CD4, CD8, and, surprisingly gammadelta T cells are all sources of IL-1beta. In addition, gammadelta T cells are also targets of this cytokine, contributing to IL-1beta-mediated production of IL-17. Finally, we show that the level of CNS IL-1R determines the clinical severity of experimental autoimmune encephalomyelitis. These data suggest that T cell-derived IL-1beta contributes to loss of immune privilege during CNS autoimmunity via pathologic alteration in the expression of CXCL12 at the BBB.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/immunology
- Chemokine CXCL12/biosynthesis
- Chemokine CXCL12/genetics
- Chemokine CXCL12/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Protein Transport/genetics
- Protein Transport/immunology
- Receptors, Interleukin-1/deficiency
- Receptors, Interleukin-1/genetics
- Receptors, Interleukin-1/physiology
- Severity of Illness Index
- Signal Transduction/genetics
- Signal Transduction/immunology
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
Collapse
Affiliation(s)
- Erin E. McCandless
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis MO 63110
| | - Matthew Budde
- Department of Radiology, Washington University School of Medicine, St Louis MO 63110
| | - Jason R. Lees
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St Louis MO 63110
| | - Denise Dorsey
- Department of Internal Medicine, Washington University School of Medicine, St Louis MO 63110
| | - Eric Lyng
- Department of Internal Medicine, Washington University School of Medicine, St Louis MO 63110
| | - Robyn S. Klein
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis MO 63110
- Department of Internal Medicine, Washington University School of Medicine, St Louis MO 63110
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St Louis MO 63110
| |
Collapse
|
68
|
Focosi D, Pelosini M, Palla P, Galimberti S, Caracciolo F, Benedetti E, Papineschi F, Petrini M. Hypercytokinemia-induced metabolic encephalopathy in a multiple myeloma patient on hemodialysis undergoing autologous stem cell transplantation: clinical response after plasma exchange. Transpl Immunol 2009; 21:240-3. [PMID: 19539028 DOI: 10.1016/j.trim.2009.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/02/2009] [Accepted: 06/04/2009] [Indexed: 01/20/2023]
Abstract
We report here a 50-years old female with multiple myeloma-associated chronic renal failure who underwent high-dose chemotherapy supported by autologous hematopoietic stem cell transplantation. She developed progressive encephalopathy on day 5 progressing to coma despite hemodialysis and no obvious organ failure. She finally recovered after a single 1-liter plasma exchange. The final diagnosis was metabolic encephalopathy due to hypercytokinemia, particularly high serum TNF levels. We discuss here the pathogenesis and raise an alert for monitoring cytokine levels in patients with renal failure undergoing high-dose chemotherapy.
Collapse
Affiliation(s)
- Daniele Focosi
- Division of Hematology, Department of Oncology, Transplants and Advances in Medicine, University of Pisa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Brambilla R, Persaud T, Hu X, Karmally S, Shestopalov VI, Dvoriantchikova G, Ivanov D, Nathanson L, Barnum SR, Bethea JR. Transgenic inhibition of astroglial NF-kappa B improves functional outcome in experimental autoimmune encephalomyelitis by suppressing chronic central nervous system inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:2628-40. [PMID: 19234157 PMCID: PMC4291126 DOI: 10.4049/jimmunol.0802954] [Citation(s) in RCA: 217] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the CNS, the transcription factor NF-kappaB is a key regulator of inflammation and secondary injury processes. Following trauma or disease, the expression of NF-kappaB-dependent genes is activated, leading to both protective and detrimental effects. In this study, we show that transgenic inactivation of astroglial NF-kappaB (glial fibrillary acidic protein-IkappaB alpha-dominant-negative mice) resulted in reduced disease severity and improved functional recovery following experimental autoimmune encephalomyelitis. At the chronic stage of the disease, transgenic mice exhibited an overall higher presence of leukocytes in spinal cord and brain, and a markedly higher percentage of CD8(+)CD122(+) T regulatory cells compared with wild type, which correlated with the timing of clinical recovery. We also observed that expression of proinflammatory genes in both spinal cord and cerebellum was delayed and reduced, whereas the loss of neuronal-specific molecules essential for synaptic transmission was limited compared with wild-type mice. Furthermore, death of retinal ganglion cells in affected retinas was almost abolished, suggesting the activation of neuroprotective mechanisms. Our data indicate that inhibiting NF-kappaB in astrocytes results in neuroprotective effects following experimental autoimmune encephalomyelitis, directly implicating astrocytes in the pathophysiology of this disease.
Collapse
Affiliation(s)
- Roberta Brambilla
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Trikaldarshi Persaud
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Xianchen Hu
- Departments of Microbiology and Neurology, University of Alabama, Birmingham, AL 35294
| | - Shaffiat Karmally
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Valery I. Shestopalov
- Bascom Palmer Eye Institute, Miller School of Medicine, University of Miami, Miami, FL 33136
- Department of Cell Biology and Anatomy, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Galina Dvoriantchikova
- Bascom Palmer Eye Institute, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Dmitry Ivanov
- Bascom Palmer Eye Institute, Miller School of Medicine, University of Miami, Miami, FL 33136
- Vavilov Institute of General Genetics, Russian Academy of Science, Moscow, Russian Federation
| | - Lubov Nathanson
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Scott R. Barnum
- Departments of Microbiology and Neurology, University of Alabama, Birmingham, AL 35294
| | - John R. Bethea
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136
- Neuroscience Program, Miller School of Medicine, University of Miami, Miami, FL 33136
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136
| |
Collapse
|
70
|
Lees JR, Iwakura Y, Russell JH. Host T cells are the main producers of IL-17 within the central nervous system during initiation of experimental autoimmune encephalomyelitis induced by adoptive transfer of Th1 cell lines. THE JOURNAL OF IMMUNOLOGY 2008; 180:8066-72. [PMID: 18523270 DOI: 10.4049/jimmunol.180.12.8066] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, has long been thought to be mediated by Th1 CD4(+) T cells. Using adoptive transfer techniques, transfer of CNS specific Th1 T cells was sufficient to induce EAE in naive mice. However, recent studies found a vital role for IL-17 in induction of EAE. These studies suggested that a fraction of IL-17-producing T cells that contaminate Th1 polarized cell lines are largely responsible for initiation of EAE. In this study, we tracked the appearance and cytokine production capacity of adoptively transferred cells within the CNS of mice throughout EAE disease. IL-17-producing, adoptively transferred cells were not enriched over the low percentages present in vitro. Thus, there was no selective recruitment and/or preferential proliferation of adoptively transferred IL-17-producing cells during the induction of EAE. Instead a large number of CNS infiltrating host T cells in mice with EAE were capable of producing IL-17 following ex vivo stimulation. The IL-17-producing T cells contained both alphabeta and gammadelta TCR(+) T cells with a CD4(+)CD8(-) or CD4(-)CD8(-) phenotype. These cells concentrated within the CNS within 3 days of adoptive transfer, and appeared to play a role in EAE induction as adoptive transfer of Th1 lines derived from wild-type mice into IL-17-deficient mice induced reduced EAE clinical outcomes. This study demonstrates that an encephalitogenic Th1 cell line induces recruitment of host IL-17-producing T cells to the CNS during the initiation of EAE and that these cells contribute to the incidence and severity of disease.
Collapse
Affiliation(s)
- Jason R Lees
- Department of Molecular Biology and Pharmacology, Washington University, St Louis, MO 63110, USA
| | | | | |
Collapse
|
71
|
Abstract
It has long been thought that astrocytes, like other glial cells, simply provide a support mechanism for neuronal function in the healthy and inflamed central nervous system (CNS). However, recent evidence suggests that astrocytes play an active and dual role in CNS inflammatory diseases such as multiple sclerosis (MS). Astrocytes not only have the ability to enhance immune responses and inhibit myelin repair, but they can also be protective and limit CNS inflammation while supporting oligodendrocyte and axonal regeneration. The particular impact of these cells on the pathogenesis and repair of an inflammatory demyelinating process is dependent upon a number of factors, including the stage of the disease, the type and microenvironment of the lesion, and the interactions with other cell types and factors that influence their activation. In this review, we summarize recent data supporting the idea that astrocytes play a complex role in the regulation of CNS autoimmunity.
Collapse
Affiliation(s)
- A. Nair
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Fienberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611 USA
| | - T. J. Frederick
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Fienberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611 USA
| | - S. D. Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Fienberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611 USA
| |
Collapse
|
72
|
Carpentier PA, Getts MT, Miller SD. Pro-inflammatory functions of astrocytes correlate with viral clearance and strain-dependent protection from TMEV-induced demyelinating disease. Virology 2008; 375:24-36. [PMID: 18289626 PMCID: PMC2397444 DOI: 10.1016/j.virol.2008.01.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2007] [Revised: 01/03/2008] [Accepted: 01/15/2008] [Indexed: 12/11/2022]
Abstract
Intracerebral infection of susceptible strains of mice, e.g. SJL/J, with Theiler's murine encephalomyelitis virus (TMEV) leads to a persistent CNS infection accompanied by development of a chronic-progressive inflammatory CNS autoimmune demyelinating disease which is clinically and pathologically similar to human multiple sclerosis. In contrast, resistant strains of mice, e.g. C57BL/6 (B6), effectively clear TMEV from the CNS and do not develop demyelinating disease. Although CD8(+) T cells are crucial for viral clearance in B6 mice, SJL mice also mount potent CD8(+) T cell responses against virus, thus the reason for the viral persistence in the CNS in these mice is unclear. Here, we examined innate anti-viral responses of CNS-resident astrocytes as a potential determinant of viral persistence and disease susceptibility. We demonstrate that B6 astrocytes produce significantly higher levels of cytokines, chemokines and adhesion molecules in response to TMEV infection, or stimulation with IFN-gamma and TNF-alpha or poly I:C than SJL mice. In addition, TMEV more effectively induces MHC I molecules on B6 astrocytes than SJL, corresponding with an increased ability to activate TMEV-specific CD8(+) T cells directly ex vivo. These results suggest that enhanced anti-viral responses of B6 astrocytes contribute to the ability of these mice to clear TMEV from the CNS and therefore to their resistance to the development of autoimmune demyelinating disease.
Collapse
Affiliation(s)
| | - Meghann Teague Getts
- Northwestern University Feinberg School of Medicine, Department of Microbiology-Immunology, Interdepartmental Immunobiology Center, and Northwestern University Institute for Neuroscience, 303 E. Chicago Avenue, Chicago, IL 60611
| | - Stephen D. Miller
- Northwestern University Feinberg School of Medicine, Department of Microbiology-Immunology, Interdepartmental Immunobiology Center, and Northwestern University Institute for Neuroscience, 303 E. Chicago Avenue, Chicago, IL 60611
| |
Collapse
|
73
|
Lin ST, Wang Y, Xue Y, Feng DC, Xu Y, Xu LY. Tetrandrine suppresses LPS-induced astrocyte activation via modulating IKKs-IκBα-NF-κB signaling pathway. Mol Cell Biochem 2008; 315:41-9. [DOI: 10.1007/s11010-008-9787-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 05/05/2008] [Indexed: 11/24/2022]
|
74
|
A Preparation of Herbal Medicine Salvia miltiorrhiza Reduces Expression of Intercellular Adhesion Molecule-1 and Development of Atherosclerosis in Apolipoprotein E-Deficient Mice. J Cardiovasc Pharmacol 2008; 51:38-44. [DOI: 10.1097/fjc.0b013e31815a9575] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
75
|
A promising therapeutic approach for multiple sclerosis: recombinant T-cell receptor ligands modulate experimental autoimmune encephalomyelitis by reducing interleukin-17 production and inhibiting migration of encephalitogenic cells into the CNS. J Neurosci 2007; 27:12531-9. [PMID: 18003831 DOI: 10.1523/jneurosci.3599-07.2007] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Recombinant T-cell receptor ligands (RTLs) can prevent and reverse clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) in an antigen-specific manner and are currently in clinical trials for treatment of subjects with multiple sclerosis (MS). To evaluate regulatory mechanisms, we designed and tested RTL551, containing the alpha1 and beta1 domains of the I-A(b) class II molecule covalently linked to the encephalitogenic MOG-35-55 peptide in C57BL/6 mice. Treatment of active or passive EAE with RTL551 after disease onset significantly reduced clinical signs and spinal cord lesions. Moreover, RTL551 treatment strongly and selectively reduced secretion of interleukin-17 and tumor necrosis factor alpha by transferred green fluorescent protein-positive (GFP+) MOG-35-55-reactive T-cells and almost completely abrogated existent GFP+ cellular infiltrates in affected spinal cord sections. Reduced inflammation in spinal cords of RTL551-treated mice was accompanied by a highly significant downregulation of chemokines and their receptors and inhibition of VCAM-1 (vascular cell adhesion molecule-1) and ICAM-1 (intercellular adhesion molecule-1) expression by endothelial cells. Thus, RTL therapy cannot only inhibit systemic production of encephalitogenic cytokines by the targeted myelin oligodendrocyte glycoprotein-reactive T-cells but also impedes downstream local recruitment and retention of inflammatory cells in the CNS. These findings indicate that targeted immunotherapy of antigen-specific T-cells can result in a reversal of CNS lesion formation and lend strong support to the application of the RTL approach for therapy in MS.
Collapse
|
76
|
Collagen-induced arthritis as a model of hyperalgesia: Functional and cellular analysis of the analgesic actions of tumor necrosis factor blockade. ACTA ACUST UNITED AC 2007; 56:4015-23. [DOI: 10.1002/art.23063] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
77
|
Abstract
In multiple sclerosis (MS), the presence of demyelinating plaques has concentrated researchers' minds on the role of the oligodendrocyte in its pathophysiology. Recently, with the rediscovery of early and widespread loss of axons in the disease, new emphasis has been put on the role of axons and axon-oligodendrocyte interactions in MS. Despite the fact that, in 1904, Müller claimed that MS was a disease of astrocytes, more recently, astrocytes have taken a back seat, except as the cells that form the final glial scar after all hope of demyelination is over. However, perhaps it is time for the return of the astrocyte to popularity in the pathogenesis of MS, with recent reports on the dual role of astrocytes in aiding degeneration and demyelination, by promoting inflammation, damage of oligodendrocytes and axons, and glial scarring, but also in creating a permissive environment for remyelination by their action on oligodendrocyte precursor migration, oligodendrocyte proliferation, and differentiation. We review these findings to try to provide a cogent view of astrocytes in the pathology of MS.
Collapse
Affiliation(s)
- Anna Williams
- Inserm, U711, Université Pierre and Marie Curie, Faculté de médecine, IFR 70, Paris F-75013, France, and Department of Clinical Neurosciences, Western General Hospital, Edinburgh, UK.
| | | | | |
Collapse
|
78
|
Chauveau F, Aissouni Y, Hamm J, Boutin H, Libri D, Ducongé F, Tavitian B. Binding of an aptamer to the N-terminal fragment of VCAM-1. Bioorg Med Chem Lett 2007; 17:6119-22. [PMID: 17905582 DOI: 10.1016/j.bmcl.2007.09.046] [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] [Received: 07/26/2007] [Revised: 09/10/2007] [Accepted: 09/11/2007] [Indexed: 02/07/2023]
Abstract
In vitro selection of 2'-fluoropyrimidine oligonucleotide aptamers was performed against the N-terminal two-domain fragment of mouse VCAM-1. The SELEX procedure enriched the starting pool in a family of homologous sequences. High binding affinity (10nM) of one member of this family, aptamer 12.11, was demonstrated in a filter binding assay.
Collapse
Affiliation(s)
- Fabien Chauveau
- Commissariat á l'Energie Atomique, DSV, I(2)BM, SHFJ, Laboratoire d'Imagerie Moléculaire Expérimentale, 4 place du général Leclerc, Orsay, France
| | | | | | | | | | | | | |
Collapse
|
79
|
Alexander JJ, Jacob A, Cunningham P, Hensley L, Quigg RJ. TNF is a key mediator of septic encephalopathy acting through its receptor, TNF receptor-1. Neurochem Int 2007; 52:447-56. [PMID: 17884256 PMCID: PMC3191465 DOI: 10.1016/j.neuint.2007.08.006] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Revised: 07/17/2007] [Accepted: 08/09/2007] [Indexed: 12/21/2022]
Abstract
In this study, we demonstrate that mice deficient in TNFR1 (TNFR1(-/-)) were resistant to LPS-induced encephalopathy. Systemic administration of lipopolysaccharide (LPS) induces a widespread inflammatory response similar to that observed in sepsis. Following LPS administration TNFR1(-/-) mice had less caspase-dependent apoptosis in brain cells and fewer neutrophils infiltrating the brain (p<0.039), compared to control C57Bl6 (TNFR1(+/+)) mice. TNFR1-dependent increase in aquaporin (AQP)-4 mRNA and protein expression was observed with a concomitant increase in water content, in brain (18% increase in C57Bl6 mice treated with LPS versus those treated with saline), similar to cerebral edema observed in sepsis. Furthermore, absence of TNFR1 partially but significantly reduced the activation of astrocytes, as shown by immunofluorescence and markedly inhibited iNOS mRNA expression (p<0.01). Septic encephalopathy is a devastating complication of sepsis. Although, considerable work has been done to identify the mechanism causing the pathological alterations in this setting, the culprit still remains an enigma. Our results demonstrate for the first time that endotoxemia leads to inflammation in brain, with alteration in blood-brain barrier, up-regulation of AQP4 and associated edema, neutrophil infiltration, astrocytosis, as well as apoptotic cellular death, all of which appear to be mediated by TNF-alpha signaling through TNFR1.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Aquaporin 4/genetics
- Aquaporin 4/metabolism
- Astrocytes/metabolism
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/physiopathology
- Brain/metabolism
- Brain/physiopathology
- Brain Diseases, Metabolic/metabolism
- Brain Diseases, Metabolic/pathology
- Brain Diseases, Metabolic/physiopathology
- Brain Edema/genetics
- Brain Edema/metabolism
- Brain Edema/physiopathology
- Chemotaxis, Leukocyte/genetics
- Encephalitis/metabolism
- Encephalitis/microbiology
- Encephalitis/physiopathology
- Gliosis/genetics
- Gliosis/metabolism
- Gliosis/physiopathology
- Inflammation Mediators/metabolism
- Lipopolysaccharides
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide Synthase Type II/genetics
- RNA, Messenger/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Sepsis/complications
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Jessy J Alexander
- Department of Medicine, University of Chicago, 5841 South Maryland Avenue, MC5100, Chicago, IL 60637, USA.
| | | | | | | | | |
Collapse
|
80
|
McCandless EE, Klein RS. Molecular targets for disrupting leukocyte trafficking during multiple sclerosis. Expert Rev Mol Med 2007; 9:1-19. [PMID: 17637110 DOI: 10.1017/s1462399407000397] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AbstractAutoimmune diseases of the central nervous system (CNS) involve the migration of abnormal numbers of self-directed leukocytes across the blood–brain barrier that normally separates the CNS from the immune system. The cardinal lesion associated with neuroinflammatory diseases is the perivascular infiltrate, which comprises leukocytes that have traversed the endothelium and have congregated in a subendothelial space between the endothelial-cell basement membrane and the glial limitans. The exit of mononuclear cells from this space can be beneficial, as when virus-specific lymphocytes enter the CNS for pathogen clearance, or might induce CNS damage, such as in the autoimmune disease multiple sclerosis when myelin-specific lymphocytes invade and induce demyelinating lesions. The molecular mechanisms involved in the movement of lymphocytes through these compartments involve multiple signalling pathways between these cells and the microvasculature. In this review, we discuss adhesion, costimulatory, cytokine, chemokine and signalling molecules involved in the dialogue between lymphocytes and endothelial cells that leads to inflammatory infiltrates within the CNS, and the targeting of these molecules as therapies for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Erin E McCandless
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
81
|
Sanders P, De Keyser J. Janus faces of microglia in multiple sclerosis. ACTA ACUST UNITED AC 2007; 54:274-85. [PMID: 17383006 DOI: 10.1016/j.brainresrev.2007.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 03/02/2007] [Accepted: 03/02/2007] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is the most common cause of neurological disability in young adults. The disease is characterized by inflammatory reactions, demyelination and axonal loss in the brain, spinal cord and optic nerves. Microglia seem to play an important role in the inflammatory processes in MS, since they are found in actively demyelinating lesions. Their role in the differentiation of T cells could led to the expansion of inflammation and tissue destruction. However, microglia are also involved in the termination of an inflammatory response and produce protective factors. To be able to therapeutically manipulate microglia, their exact function in the onset and development of MS needs to be clarified. This review provides an overview of the functions of the most important microglia-associated molecules in MS, being CD40, B7-1 and B7-2, interferon-gamma, tumor necrosis factor-alpha, chemokines, prostanoids, and nitric oxide.
Collapse
Affiliation(s)
- Patricia Sanders
- Department of Neurology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
82
|
Archambault AS, Sim J, McCandless EE, Klein RS, Russell JH. Region-specific regulation of inflammation and pathogenesis in experimental autoimmune encephalomyelitis. J Neuroimmunol 2006; 181:122-32. [PMID: 17030428 DOI: 10.1016/j.jneuroim.2006.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Revised: 08/25/2006] [Accepted: 08/28/2006] [Indexed: 11/20/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis and is characterized by an infiltrate of predominantly T cells and macrophages in the spinal cord and brain. In both the spinal cord and the cerebellum, Th1 cells direct inflammation to antigen-rich white matter tracts, and there is a TNFR1-dependent recruitment of CD11b(hi) cells in both regions. In the spinal cord, parenchymal invasion, demyelination and clinical symptoms are associated with TNFR1-dependant parenchymal induction (especially astrocytes) of VCAM-1 and CXCL2. None of these events occur in the cerebellum despite the fact that an inflammatory infiltrate accumulates in the perivascular space. Therefore regional specificity in astrocyte responses to inflammatory cytokines may regulate regional parenchymal infiltration and pathogenesis.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD11b Antigen/metabolism
- Cell Movement/immunology
- Cerebellum/immunology
- Cerebellum/metabolism
- Cerebellum/pathology
- Chemokine CXCL2
- Chemokines/metabolism
- Coloring Agents
- Demyelinating Diseases/immunology
- Demyelinating Diseases/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endothelial Cells/metabolism
- Macrophages/immunology
- Macrophages/pathology
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Monocytes/immunology
- Monocytes/pathology
- Myelin Proteins
- Myelin-Associated Glycoprotein/immunology
- Myelin-Associated Glycoprotein/metabolism
- Myelin-Oligodendrocyte Glycoprotein
- Nerve Fibers, Myelinated/immunology
- Nerve Fibers, Myelinated/pathology
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Thy-1 Antigens/genetics
- Tolonium Chloride
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- Angela S Archambault
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
83
|
Abstract
Central nervous system (CNS) immune privilege is an experimentally defined phenomenon. Tissues that are rapidly rejected by the immune system when grafted in sites, such as the skin, show prolonged survival when grafted into the CNS. Initially, CNS immune privilege was construed as CNS isolation from the immune system by the blood-brain barrier (BBB), the lack of draining lymphatics, and the apparent immunoincompetence of microglia, the resident CNS macrophage. CNS autoimmunity and neurodegeneration were presumed automatic consequences of immune cell encounter with CNS antigens. Recent data have dramatically altered this viewpoint by revealing that the CNS is neither isolated nor passive in its interactions with the immune system. Peripheral immune cells can cross the intact BBB, CNS neurons and glia actively regulate macrophage and lymphocyte responses, and microglia are immunocompetent but differ from other macrophage/dendritic cells in their ability to direct neuroprotective lymphocyte responses. This newer view of CNS immune privilege is opening the door for therapies designed to harness autoreactive lymphocyte responses and also implies (i) that CNS autoimmune diseases (i.e. multiple sclerosis) may result as much from neuronal and/or glial dysfunction as from immune system dysfunctions and (ii) that the severe neuronal and glial dysfunction associated with neurodegenerative disorders (i.e. Alzheimer's disease) likely alters CNS-specific regulation of lymphocyte responses affecting the utility of immune-based therapies (i.e. vaccines).
Collapse
Affiliation(s)
- Monica J Carson
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA.
| | | | | | | | | |
Collapse
|
84
|
van Loo G, De Lorenzi R, Schmidt H, Huth M, Mildner A, Schmidt-Supprian M, Lassmann H, Prinz MR, Pasparakis M. Inhibition of transcription factor NF-κB in the central nervous system ameliorates autoimmune encephalomyelitis in mice. Nat Immunol 2006; 7:954-61. [PMID: 16892069 DOI: 10.1038/ni1372] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Accepted: 07/12/2006] [Indexed: 12/31/2022]
Abstract
Activation of transcription factor NF-kappaB in the central nervous system (CNS) has been linked to autoimmune demyelinating disease; however, it remains unclear whether its function is protective or pathogenic. Here we show that CNS-restricted ablation of 'upstream' NF-kappaB activators NEMO or IKK2 but not IKK1 ameliorated disease pathology in a mouse model of multiple sclerosis, suggesting that 'canonical' NF-kappaB activation in cells of the CNS has a mainly pathogenic function in autoimmune demyelinating disease. NF-kappaB inhibition prevented the expression of proinflammatory cytokines, chemokines and the adhesion molecule VCAM-1 from CNS-resident cells. Thus, NF-kappaB-dependent gene expression in non-microglial cells of the CNS provides a permissive proinflammatory milieu that is critical for CNS inflammation and tissue damage in autoimmune demyelinating disease.
Collapse
Affiliation(s)
- Geert van Loo
- European Molecular Biology Laboratory Mouse Biology Unit, I-00016 Monterotondo, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Yang P, Baker KA, Hagg T. The ADAMs family: coordinators of nervous system development, plasticity and repair. Prog Neurobiol 2006; 79:73-94. [PMID: 16824663 DOI: 10.1016/j.pneurobio.2006.05.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 05/18/2006] [Accepted: 05/23/2006] [Indexed: 10/24/2022]
Abstract
A disintegrin and metalloprotease (ADAM) transmembrane proteins have metalloprotease, integrin-binding, intracellular signaling and cell adhesion activities. In contrast to other metalloproteases, ADAMs are particularly important for cleavage-dependent activation of proteins such as Notch, amyloid precursor protein (APP) and transforming growth factor alpha (TGFalpha), and can bind integrins. Not surprisingly, ADAMs have been shown or suggested to play important roles in the development of the nervous system, where they regulate proliferation, migration, differentiation and survival of various cells, as well as axonal growth and myelination. On the eleventh anniversary of the naming of this family of proteins, the relatively unknown ADAMs are emerging as potential therapeutic targets for neural repair. For example, over-expression of ADAM10, one of the alpha-secretases for APP, can prevent amyloid formation and hippocampal defects in an Alzheimer mouse model. Another example of this potential neural repair role is the finding that ADAM21 is uniquely associated with neurogenesis and growing axons of the adult brain. This comprehensive review will discuss the growing literature about the roles of ADAMs in the developing and adult nervous system, and their potential roles in neurological disorders. Most excitingly, the expanding understanding of their normal roles suggests that they can be manipulated to promote neural repair in the degenerating and injured adult nervous system.
Collapse
Affiliation(s)
- Peng Yang
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, Louisville, KY 40292, USA
| | | | | |
Collapse
|
86
|
Lees JR, Archambault AS, Russell JH. T-cell trafficking competence is required for CNS invasion. J Neuroimmunol 2006; 177:1-10. [PMID: 16822552 DOI: 10.1016/j.jneuroim.2006.05.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Revised: 05/05/2006] [Accepted: 05/09/2006] [Indexed: 01/06/2023]
Abstract
T-cell invasion of the CNS is critical for the induction of a variety of autoimmune mediated neuronal diseases. We utilized blood-brain barrier (BBB) mediated exclusion of anti-CD4 antibody to define populations of encephalitogenic T-cells recovered from mouse CNS preparations as either CNS invasive or non-invasive. This separation of cells allowed flow cytometric examination of the kinetics of encephalitogenic T-cell entry past the BBB. Further experiments examined the relative contribution of EAE inflammatory conditioning of the BBB to the kinetics of T-cell adherence and migration into the CNS. Inflammatory conditioning was found to have no effect on accumulation of T-cells at the vascular interface of the BBB, but was found to increase the entry of adoptively transferred T-cells into the CNS following their initial adherence to the BBB.
Collapse
Affiliation(s)
- Jason R Lees
- Department of Molecular Biology and Pharmacology, Campus Box 8103, 660 South Euclid, Washington University, St. Louis 63110, USA
| | | | | |
Collapse
|
87
|
Wheeler RD, Zehntner SP, Kelly LM, Bourbonnière L, Owens T. Elevated interferon gamma expression in the central nervous system of tumour necrosis factor receptor 1-deficient mice with experimental autoimmune encephalomyelitis. Immunology 2006; 118:527-38. [PMID: 16780563 PMCID: PMC1782311 DOI: 10.1111/j.1365-2567.2006.02395.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Inflammation in the central nervous system (CNS) can be studied in experimental autoimmune encephalomyelitis (EAE). The proinflammatory cytokines interferon-gamma (IFN-gamma) and tumour necrosis factor (TNF) are implicated in EAE pathogenesis. Signals through the type 1 TNF receptor (TNFR1) are required for severe EAE to develop, whereas deficiency in IFN-gamma or its receptor result in more severe EAE. We investigated IFN-gamma expression in TNFR1-deficient (TNFR1-/-) mice. We describe here that there were more IFN-gamma-secreting T cells present in the CNS of TNFR1-/- mice during EAE compared to wild-type (WT) mice, despite that clinical symptoms were mild, with delayed onset. There was greater expression of IL-12/23p40 by antigen-presenting cells in these mice, and in vitro, TNFR1-/- antigen-presenting cells induced greater secretion of IFN-gamma but not interleukin (IL)-17 when cultured with primed T cells than did WT antigen presenting cells. TNFR1-/- mice with EAE had significantly higher expression of CXCL10 mRNA (but not CCL5 mRNA) in the CNS compared to WT mice with EAE. These data demonstrate that IFN-gamma expression is enhanced in the CNS of TNFR1-/- mice with EAE and suggest that IFN-gamma levels do not necessarily correlate with EAE severity.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/microbiology
- Gene Expression Regulation
- Interferon-gamma/analysis
- Interferon-gamma/genetics
- Macrophages/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Tumor Necrosis Factor, Type I/analysis
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Spinal Cord/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Rachel D Wheeler
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Simone P Zehntner
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Lisa M Kelly
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Lyne Bourbonnière
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Trevor Owens
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
- Medical Biotechnology Centre, University of Southern DenmarkOdense C, Denmark
| |
Collapse
|
88
|
Gimenez MA, Sim J, Archambault AS, Klein RS, Russell JH. A tumor necrosis factor receptor 1-dependent conversation between central nervous system-specific T cells and the central nervous system is required for inflammatory infiltration of the spinal cord. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1200-9. [PMID: 16565495 PMCID: PMC1606568 DOI: 10.2353/ajpath.2006.050332] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We examined the role of tumor necrosis factor receptor 1 (TNFR1) in inflammation initiated by the adoptive transfer of central nervous system (CNS)-specific Th1 cells in experimental autoimmune encephalomyelitis, a murine model of multiple sclerosis. This adoptive transfer paradigm eliminates the confounding effects of bacterial adjuvants in the analysis of inflammation. We found that although T cells could reach the meninges and perivascular space in the absence of TNFR1, recruitment of other inflammatory cells from the blood was dramatically reduced. The reduction in the recruitment of CD11b(hi) cells correlated with a dramatic reduction in the production of the chemokines CCL2 (MCP-1) and CXLC2 (MIP-2) in TNFR1-deficient hosts. Bone marrow chimera experiments demonstrated that TNF can be effectively supplied by either the hematopoietic system or the CNS, but the essential TNFR1-responsive cells reside in the CNS. Previous work has demonstrated that microglia produce CCL2, and here we demonstrate that astrocytes and endothelial cells produced CXCL2 in the early stages of inflammation. Therefore, productive inflammation results from a conversation, or mutually responding signals, between the initiating T cells and cells in the parenchyma of the spinal cord.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Astrocytes/metabolism
- Bone Marrow Cells/metabolism
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- CD11b Antigen/metabolism
- Chemokine CCL2/biosynthesis
- Chemokine CXCL2
- Chemokines/biosynthesis
- Chemokines, CXC/biosynthesis
- Chimera
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endothelial Cells/metabolism
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Myelitis/immunology
- Myelitis/metabolism
- Myelitis/pathology
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- T-Lymphocytes/immunology
- Th1 Cells/immunology
- Tumor Necrosis Factor Decoy Receptors
Collapse
Affiliation(s)
- Mary Ann Gimenez
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
89
|
Falsig J, Pörzgen P, Lund S, Schrattenholz A, Leist M. The inflammatory transcriptome of reactive murine astrocytes and implications for their innate immune function. J Neurochem 2006; 96:893-907. [PMID: 16405499 DOI: 10.1111/j.1471-4159.2005.03622.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Upon injury, astrocytes assume an activated state associated with the release of inflammatory mediators. To model this, we stimulated murine primary astrocytes with a complete inflammatory cytokine mix consisting of TNF-alpha, IL-1beta and IFN-gamma. We analysed the transcriptional response of 480 genes at 4 and 16 h after stimulation on a chip designed to give a representative overview over the inflammation-relevant part of the transcriptome of macrophage-like cells. The list of the 182 genes found to be significantly regulated in astrocytes revealed an intriguing co-ordinate regulation of genes linked to the biological processes of antiviral/antimicrobial defence, antigen presentation and facilitation of leucocyte invasion. The latter group was characterized by very high up-regulations of chemokine genes. We also identified regulations of a thymidylate kinase and an interferon-regulated protein with a tetratricopeptide motive, both up to now only known from macrophages. The transcriptional regulations were confirmed on the protein level by a proteomic analysis. These findings taken together suggest that activated astrocytes in brain behave similarly in many respects to inflamed macrophages in the periphery.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Astrocytes/drug effects
- Astrocytes/immunology
- Cell Cycle/genetics
- Cell Death/genetics
- Cells, Cultured
- Cerebral Cortex/cytology
- Chemokine CCL5/genetics
- Chemokine CCL5/metabolism
- Cytokines/pharmacology
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay/methods
- Female
- Glial Fibrillary Acidic Protein/metabolism
- Immunity, Innate/genetics
- Immunity, Innate/physiology
- Immunohistochemistry/methods
- Inflammation/genetics
- Inflammation/metabolism
- Mice
- Mice, Inbred C57BL
- Models, Immunological
- Nitrites/metabolism
- Oligonucleotide Array Sequence Analysis/methods
- Pregnancy
- Proteomics/methods
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
- Stress, Physiological/genetics
- Time Factors
Collapse
Affiliation(s)
- Jeppe Falsig
- Institute of Neuropathology, University of Zürich, Schmelzbergstrasse 12, CH-8032 Zürich, Switzerland.
| | | | | | | | | |
Collapse
|
90
|
Hövelmeyer N, Hao Z, Kranidioti K, Kassiotis G, Buch T, Frommer F, von Hoch L, Kramer D, Minichiello L, Kollias G, Lassmann H, Waisman A. Apoptosis of oligodendrocytes via Fas and TNF-R1 is a key event in the induction of experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2005; 175:5875-84. [PMID: 16237080 DOI: 10.4049/jimmunol.175.9.5875] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, immunization with myelin Ags leads to demyelination and paralysis. To investigate which molecules are crucial for the pathogenesis of EAE, we specifically assessed the roles of the death receptors Fas and TNF-R1. Mice lacking Fas expression in oligodendrocytes (ODCs) were generated and crossed to TNF-R1-deficient mice. To achieve specific deletion of a loxP-flanked fas allele in ODCs, we generated a new insertion transgene, expressing the Cre recombinase specifically in ODCs. Fas inactivation alone as well as the complete absence of TNF-R1 protected mice partially from EAE induced by the immunization with myelin ODC glycoprotein. The double-deficient mice, however, showed almost no clinical signs of EAE after immunization. Histological analysis revealed that demyelination was suppressed in CNS tissue and that lymphocyte infiltration was notably reduced. We conclude that the death receptors Fas and TNF-R1 are major initiators of ODC apoptosis in EAE. Although only moderate reduction of lymphocyte infiltration into CNS tissue was observed, the absence of these receptors appears to confer protection from demyelination and development of clinical disease.
Collapse
Affiliation(s)
- Nadine Hövelmeyer
- Laboratory for Molecular Immunology, Institute for Genetics, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Russell JH. Interaction between the immune and central nervous systems. Immunol Res 2005; 32:225-9. [PMID: 16106074 DOI: 10.1385/ir:32:1-3:225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Much of the understanding of tolerance has focused on the requirements for antigen-specific lymphocyte activation and function. However, there is increasing evidence for anatomic regulation of effector access to self antigens. Recently, a number of studies have provided evidence for tissue-specific "addressins" in chemokine/chemokine receptor pairs. The central nervous system (CNS) provides special anatomic barriers to the movement of cells from the vascular compartment to the parenchyma. Herein I raise the possibility that antigen, perhaps through specialized antigen-presenting cells, may play a role in regulating access of activated lymphocytes into the CNS parenchyma. The results suggest that a reexamination of the widely held dogma that all activated lymphocytes have access to the CNS parenchyma is necessary to understand the relationship between the immune and central nervous systems.
Collapse
Affiliation(s)
- John H Russell
- Program in Immunology, Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
92
|
Archambault AS, Sim J, Gimenez MAT, Russell JH. Defining antigen-dependent stages of T cell migration from the blood to the central nervous system parenchyma. Eur J Immunol 2005; 35:1076-85. [PMID: 15761850 DOI: 10.1002/eji.200425864] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In experimental autoimmune encephalomyelitis (EAE), intravenous transfer of activated CD4(+) myelin-specific T cells is sufficient to induce disease. Transferred T cells access the CNS parenchyma by trafficking across the blood brain barrier (BBB) vascular endothelium into the perivascular space, and then across the glial limitans that is made up of astrocytes and microglia. Flow cytometry analysis of cells isolated from CNS tissue does not distinguish between T cell populations at the various stages of migration. In this study, we have used GK1.5 (anti-CD4) treatment along with immunohistochemistry to distinguish between populations of T cells that are associated with the vasculature, T cells that have migrated into the perivascular space, and T cells in the parenchyma. We have also re-evaluated antigen specificity requirements of T cells as they are recruited to the CNS parenchyma. Activated myelin-specific T cells are restricted to the CNS vasculature for at least 24 h post transfer. MHC class II expression on the recipient is required for cells to traffic across the CNS vascular endothelium. Further, Con A-stimulated or non-CNS-specific (ovalbumin-specific) T cells fail to migrate into the perivascular space, and only enter the CNS parenchyma when co-transferred with myelin-specific T cells. Our results indicate that Th1 populations cannot accumulate in the perivascular (subarachnoid, Virchow-Robbins) space without a CNS antigen-specific signal.
Collapse
Affiliation(s)
- Angela S Archambault
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
93
|
Keshavan P, Deem TL, Schwemberger SJ, Babcock GF, Cook-Mills JM, Zucker SD. Unconjugated bilirubin inhibits VCAM-1-mediated transendothelial leukocyte migration. THE JOURNAL OF IMMUNOLOGY 2005; 174:3709-18. [PMID: 15749910 DOI: 10.4049/jimmunol.174.6.3709] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
UNLABELLED During lymphocyte migration, engagement of VCAM-1 stimulates the generation of endothelial cell-derived reactive oxygen species (ROS) and activation of matrix metalloproteinases, facilitating endothelial retraction. Because bilirubin is a potent antioxidant, we examined the hypothesis that this bile pigment inhibits VCAM-1-dependent cellular events. The migration of isolated murine splenic lymphocytes across monolayers of murine endothelial cell lines (which constitutively express VCAM-1) is significantly inhibited by physiological concentrations of bilirubin, in the absence of an effect on lymphocyte adhesion. Bilirubin administration also suppresses VCAM-1-stimulated ROS generation and reduces endothelial cell matrix metalloproteinase activity. In a murine asthma model characterized by VCAM-1-dependent airway inflammation, treatment of C57BL6/J mice with i.p. bilirubin decreases the total leukocyte count in the lung parenchyma and lavage fluid, through specific inhibition of eosinophil and lymphocyte infiltration. Blood eosinophil counts were increased in bilirubin-treated animals, while VCAM-1 expression in the capillary endothelium and cytokine levels in both lung lavage and supernatants from cultured lymph node lymphocytes were unchanged, suggesting that bilirubin inhibits leukocyte migration. CONCLUSION bilirubin blocks VCAM-1-dependent lymphocyte migration in vitro and ameliorates VCAM-1-mediated airway inflammation in vivo, apparently through the suppression of cellular ROS production. These findings support a potential role for bilirubin as an endogenous immunomodulatory agent.
Collapse
Affiliation(s)
- Pavitra Keshavan
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH 45267, USA.
| | | | | | | | | | | |
Collapse
|
94
|
Carpentier PA, Begolka WS, Olson JK, Elhofy A, Karpus WJ, Miller SD. Differential activation of astrocytes by innate and adaptive immune stimuli. Glia 2005; 49:360-74. [PMID: 15538753 DOI: 10.1002/glia.20117] [Citation(s) in RCA: 289] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immunologic privilege of the central nervous system (CNS) makes it crucial that CNS resident cells be capable of responding rapidly to infection. Astrocytes have been reported to express Toll-like receptors (TLRs), hallmark pattern recognition receptors of the innate immune system, and respond to their ligation with cytokine production. Astrocytes have also been reported to respond to cytokines of the adaptive immune system with the induction of antigen presentation functions. Here we have compared the ability of TLR stimuli and the adaptive immune cytokines interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha) to induce a variety of immunologic functions of astrocytes. We show that innate signals LPS- and poly I:C lead to stronger upregulation of TLRs and production of the cytokines IL-6 and TNF-alpha as well as innate immune effector molecules IFN-alpha4, IFN-beta, and iNOS compared with cytokine-stimulated astrocytes. Both innate stimulation and adaptive stimulation induce similar expression of the chemokines CCL2, CCL3, and CCL5, as well as similar enhancement of adhesion molecule ICAM-1 and VCAM-1 expression by astrocytes. Stimulation with adaptive immune cytokines, however, was unique in its ability to induce upregulation of MHC II and the functional ability of astrocytes to activate CD4(+) T cells. These results indicate potentially important and changing roles for astrocytes during the progression of CNS infection.
Collapse
Affiliation(s)
- Pamela A Carpentier
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|