1
|
Miller MR, Landis HE, Miller RE, Tizabi Y. Intercellular Adhesion Molecule 1 (ICAM-1): An Inflammatory Regulator with Potential Implications in Ferroptosis and Parkinson's Disease. Cells 2024; 13:1554. [PMID: 39329738 PMCID: PMC11430830 DOI: 10.3390/cells13181554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Intercellular adhesion molecule 1 (ICAM-1/CD54), a transmembrane glycoprotein, has been considered as one of the most important adhesion molecules during leukocyte recruitment. It is encoded by the ICAM1 gene and plays a central role in inflammation. Its crucial role in many inflammatory diseases such as ulcerative colitis and rheumatoid arthritis are well established. Given that neuroinflammation, underscored by microglial activation, is a key element in neurodegenerative diseases such as Parkinson's disease (PD), we investigated whether ICAM-1 has a role in this progressive neurological condition and, if so, to elucidate the underpinning mechanisms. Specifically, we were interested in the potential interaction between ICAM-1, glial cells, and ferroptosis, an iron-dependent form of cell death that has recently been implicated in PD. We conclude that there exist direct and indirect (via glial cells and T cells) influences of ICAM-1 on ferroptosis and that further elucidation of these interactions can suggest novel intervention for this devastating disease.
Collapse
Affiliation(s)
| | - Harold E. Landis
- Integrative Medicine Fellow, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | | | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| |
Collapse
|
2
|
Sheikh MA, O'Connell KS, Lekva T, Szabo A, Akkouh IA, Osete JR, Agartz I, Engh JA, Andreou D, Boye B, Bøen E, Elvsåshagen T, Hope S, Frogner Werner MC, Joa I, Johnsen E, Kroken RA, Lagerberg TV, Melle I, Drange OK, Morken G, Nærland T, Sørensen K, Vaaler AE, Weibell MA, Westlye LT, Aukrust P, Djurovic S, Steen NE, Andreassen OA, Ueland T. Systemic Cell Adhesion Molecules in Severe Mental Illness: Potential Role of Intercellular CAM-1 in Linking Peripheral and Neuroinflammation. Biol Psychiatry 2023; 93:187-196. [PMID: 36182530 DOI: 10.1016/j.biopsych.2022.06.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cell adhesion molecules (CAMs) orchestrate leukocyte trafficking and could link peripheral and neuroinflammation in patients with severe mental illness (SMI), by promoting inflammatory and immune-mediated responses and mediating signals across blood-brain barrier. We hypothesized that CAMs would be dysregulated in SMI and evaluated plasma levels of different vascular and neural CAMs. Dysregulated CAMs in plasma were further evaluated in vivo in leukocytes and brain tissue and in vitro in induced pluripotent stem cells. METHODS We compared plasma soluble levels of different vascular (VCAM-1, ICAM-1, P-SEL) and neural (JAM-A, NCAD) CAMs in circulating leukocytes in a large SMI sample of schizophrenia (SCZ) spectrum disorder (n = 895) and affective disorder (n = 737) and healthy control participants (n = 1070) controlling for age, sex, body mass index, C-reactive protein, and freezer storage time. We also evaluated messenger RNA expression of ICAM1 and related genes encoding ICAM-1 receptors in leukocytes using microarray (n = 842) and in available RNA sequencing data from the CommonMind Consortium (CMC) in postmortem samples from the dorsolateral prefrontal cortex (n = 474). The regulation of soluble ICAM-1 in induced pluripotent stem cell-derived neurons and astrocytes was assessed in patients with SCZ and healthy control participants (n = 8 of each). RESULTS Our major findings were 1) increased soluble ICAM-1 in patients with SMI compared with healthy control participants; 2) increased ITGB2 messenger RNA, encoding the beta chain of the ICAM-1 receptor, in circulating leukocytes from patients with SMI and increased prefrontal cortex messenger RNA expression of ICAM1 in SCZ; and 3) enhanced soluble ICAM-1 release in induced pluripotent stem cell-derived neurons from patients with SCZ. CONCLUSIONS Our results support a systemic and cerebral dysregulation of soluble ICAM-1 expression in SMI and especially in patients with SCZ.
Collapse
Affiliation(s)
- Mashhood A Sheikh
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Kevin S O'Connell
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Attila Szabo
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ibrahim A Akkouh
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jordi Requena Osete
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ingrid Agartz
- NORMENT, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - John A Engh
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Vestfold Hospital Trust, Division of Mental Health and Addiction, Tønsberg, Norway
| | - Dimitrios Andreou
- NORMENT, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | | | | | - Torbjørn Elvsåshagen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sigrun Hope
- Department of Neuro Habilitation, Oslo University Hospital Ullevål, Oslo, Norway
| | - Maren Caroline Frogner Werner
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Inge Joa
- Network for Clinical Psychosis Research, Division of Psychiatry, Stavanger University Hospital, Stavanger, Norway; Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Erik Johnsen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway; NORMENT Centre of Excellence, Bergen, Norway
| | - Rune A Kroken
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway; NORMENT Centre of Excellence, Bergen, Norway
| | - Trine Vik Lagerberg
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Melle
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; NORMENT, University of Oslo, Oslo, Norway
| | - Ole Kristian Drange
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway; Department of Psychiatry, Sørlandet Hospital HF, Kristiansand, Norway
| | - Gunnar Morken
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Kjetil Sørensen
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Arne E Vaaler
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway
| | - Melissa Authen Weibell
- Network for Clinical Psychosis Research, Division of Psychiatry, Stavanger University Hospital, Stavanger, Norway; Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Srdjan Djurovic
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; NORMENT, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; NORMENT, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.
| |
Collapse
|
3
|
Generation of Human iPSC-Derived Astrocytes with a mature star-shaped phenotype for CNS modeling. Stem Cell Rev Rep 2022; 18:2494-2512. [PMID: 35488987 PMCID: PMC9489586 DOI: 10.1007/s12015-022-10376-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2022] [Indexed: 11/23/2022]
Abstract
The generation of astrocytes from human induced pluripotent stem cells has been hampered by either prolonged differentiation—spanning over two months—or by shorter protocols that generate immature astrocytes, devoid of salient mature astrocytic traits pivotal for central nervous system (CNS) modeling. We directed stable hiPSC-derived neuroepithelial stem cells to human iPSC-derived Astrocytes (hiAstrocytes) with a high percentage of star-shaped cells by orchestrating an astrocytic-tuned culturing environment in 28 days. We employed RT-qPCR and ICC to validate the astrocytic commitment of the neuroepithelial stem cells. To evaluate the inflammatory phenotype, we challenged the hiAstrocytes with the pro-inflammatory cytokine IL-1β (interleukin 1 beta) and quantitatively assessed the secretion profile of astrocyte-associated cytokines and the expression of intercellular adhesion molecule 1 (ICAM-1). Finally, we quantitatively assessed the capacity of hiAstrocytes to synthesize and export the antioxidant glutathione. In under 28 days, the generated cells express canonical and mature astrocytic markers, denoted by the expression of GFAP, AQP4 and ALDH1L1. In addition, the notion of a mature phenotype is reinforced by the expression of both astrocytic glutamate transporters EAAT1 and EAAT2. Thus, hiAstrocytes have a mature phenotype that encompasses traits critical in CNS modeling, including glutathione synthesis and secretion, upregulation of ICAM-1 and a cytokine secretion profile on a par with human fetal astrocytes. This protocol generates a multifaceted astrocytic model suitable for in vitro CNS disease modeling and personalized medicine.
Collapse
|
4
|
González-Alvarado MN, Aprato J, Baumeister M, Lippert M, Ekici AB, Kirchner P, Welz T, Hoffmann A, Winkler J, Wegner M, Haase S, Linker RA. Oligodendrocytes regulate the adhesion molecule ICAM-1 in neuroinflammation. Glia 2021; 70:522-535. [PMID: 34787332 DOI: 10.1002/glia.24120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/30/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023]
Abstract
Recently, oligodendrocytes (Ol) have been attributed potential immunomodulatory effects. Yet, the exact mode of interaction with pathogenic CNS infiltrating lymphocytes remains unclear. Here, we attempt to dissect mechanisms of Ol modulation during neuroinflammation and characterize the interaction of Ol with pathogenic T cells. RNA expression analysis revealed an upregulation of immune-modulatory genes and adhesion molecules (AMs), ICAM-1 and VCAM-1, in Ol when isolated from mice undergoing experimental autoimmune encephalomyelitis (EAE). To explore whether AMs are involved in the interaction of Ol with infiltrating T cells, we performed co-culture studies on mature Ol and Th1 cells. Live cell imaging analysis showed direct interaction between both cell types. Eighty percentage of Th1 cells created contacts with Ol that lasted longer than 15 min, which may be regarded as physiologically relevant. Exposure of Ol to Th1 cells or their supernatant resulted in a significant extension of Ol processes, and upregulation of AMs as well as other immunomodulatory genes. Our observations indicate that blocking of oligodendroglial ICAM-1 can reduce the number of Th1 cells initially contacting the Ol. These results suggest that AMs may play a role in the interaction between Ol and Th1 cells. We identified Ol interacting with CD4+ cells in vivo in spinal cord tissue of EAE diseased mice indicating that our in vitro findings are of interest to further scientific research in this field. Further characterization and understanding of Ol interaction with infiltrating cells may lead to new therapeutic strategies enhancing Ol protection and remyelination potential. Oligodendrocytes regulate immune modulatory genes and adhesion molecules during autoimmune neuroinflammation Oligodendrocytes interact with Th1 cells in vitro in a physiologically relevant manner Adhesion molecules may be involved in Ol-Th1 cell interaction.
Collapse
Affiliation(s)
| | - Jessica Aprato
- Department of Biochemistry, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Melissa Baumeister
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Magdalena Lippert
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Tobias Welz
- Department of Neurology and Molecular Cell Biology, University Hospital Regensburg, Regensburg, Germany
| | - Alana Hoffmann
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Michael Wegner
- Department of Biochemistry, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Stefanie Haase
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Ralf A Linker
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany.,Department of Neurology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
5
|
Drugs Modulating CD4+ T Cells Blood-Brain Barrier Interaction in Alzheimer's Disease. Pharmaceutics 2020; 12:pharmaceutics12090880. [PMID: 32948022 PMCID: PMC7558445 DOI: 10.3390/pharmaceutics12090880] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
The effect of Alzheimer’s disease (AD) medications on CD4+ T cells homing has not been thoroughly investigated. CD4+ T cells could both exacerbate and reduce AD symptoms based on their infiltrating subpopulations. Proinflammatory subpopulations such as Th1 and Th17 constitute a major source of proinflammatory cytokines that reduce endothelial integrity and stimulate astrocytes, resulting in the production of amyloid β. Anti-inflammatory subpopulations such as Th2 and Tregs reduce inflammation and regulate the function of Th1 and Th17. Recently, pathogenic Th17 has been shown to have a superior infiltrating capacity compared to other major CD4+ T cell subpopulations. Alzheimer’s drugs such as donepezil (Aricept), rivastigmine (Exelon), galantamine (Razadyne), and memantine (Namenda) are known to play an important part in regulating the mechanisms of the neurotransmitters. However, little is known about the effect of these drugs on CD4+ T cell subpopulations’ infiltration of the brain during AD. In this review, we focus on understanding the influence of AD drugs on CD4+ T cell subpopulation interactions with the BBB in AD. While current AD therapies improve endothelial integrity and reduce astrocytes activations, they vary according to their influence on various CD4+ T cell subpopulations. Donepezil reduces the numbers of Th1 but not Th2, Rivastigmine inhibits Th1 and Th17 but not Th2, and memantine reduces Th1 but not Treg. However, none of the current AD drugs is specifically designed to target the dysregulated balance in the Th17/Treg axis. Future drug design approaches should specifically consider inhibiting CD4+ Th17 to improve AD prognosis.
Collapse
|
6
|
Herrera J, Bockhorst K, Bhattarai D, Uray K. Gastrointestinal vascular permeability changes following spinal cord injury. Neurogastroenterol Motil 2020; 32:e13834. [PMID: 32163655 DOI: 10.1111/nmo.13834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/06/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gastrointestinal (GI) dysfunction is observed clinically after spinal cord injury (SCI) and contributes to the diminished long-term quality of life. Our study examined the acute and chronic GI vascular changes that occur following SCI. We demonstrated that the GI vascular tract in SCI mice becomes compromised during the acute phase of injury and persists into the chronic phase of injury. METHODS Gastrointestinal vasculature permeability was measured using dynamic contrast-enhanced magnetic resonance imaging (DCE MRI) at 48 hours, and 2 and 4 weeks following contusion spinal cord injury. Angiopoietin-1, a vascular stabilizing protein, was administered intravenously following injury. Intestinal contractile activity assessments were performed following the last imaging session. KEY RESULTS Our results indicated that a single administration of Ang-1 reduced vascular permeability at 48 hours but the effect was only transient. However, when the treatment paradigm was changed from a single administration to multiple administrations of Ang-1 following contusion injury, our DCE MRI data indicated a significant decrease in GI vascular permeability 4 weeks after injury compared with vehicle control treated animals. This improved GI vascular permeability was associated with improved sustained intestinal contractile activity. We also demonstrated that Ang-1 reduced the expression of sICAM-1 in the ileum compared with the saline-treated group. CONCLUSIONS AND INFERENCES We show that the GI vasculature is compromised in the acute and chronic phase of injury following spinal contusion. Our results also indicate that multiple administrations of Ang-1 can attenuate GI vascular permeability, possibly reduce inflammation, and improve sustained agonist-induced contraction compared with saline treatment.
Collapse
Affiliation(s)
- Juan Herrera
- Diagnostic and Interventional Imaging, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kurt Bockhorst
- Diagnostic and Interventional Imaging, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Deepa Bhattarai
- Pediatric Surgery University of Texas Medical School at Houston, Houston, Texas, USA
| | - Karen Uray
- Pediatric Surgery University of Texas Medical School at Houston, Houston, Texas, USA.,Medicinal Chemistry, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
7
|
Son YK, Yoon SR, Bang WY, Bae CH, Yeo JH, Yeo R, An J, Song J, Kim OY. Carpinus turczaninowii extract modulates arterial inflammatory response: a potential therapeutic use for atherosclerosis. Nutr Res Pract 2019; 13:302-309. [PMID: 31388406 PMCID: PMC6669074 DOI: 10.4162/nrp.2019.13.4.302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/09/2019] [Accepted: 05/14/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND/OBJECTIVES Vascular inflammation is an important feature in the atherosclerotic process. Recent studies report that leaves and branches of Carpinus turczaninowii (C. turczaninowii) have antioxidant capacity and exert anti-inflammatory effects. However, no study has reported the regulatory effect of C. turczaninowii extract on the arterial inflammatory response. This study therefore investigated modulation of the arterial inflammatory response after exposure to C. turczaninowii extract, using human aortic vascular smooth muscle cells (HAoSMCs). MATERIALS/METHODS Scavenging activity of free radicals, total phenolic content (TPC), cell viability, mRNA expressions, and secreted levels of cytokines were measured in LPS-stimulated (10 ng/mL) HAoSMCs treated with the C. turczaninowii extract. RESULTS C. turczaninowii extract contains high amounts of TPC (225.6 ± 21.0 mg of gallic acid equivalents/g of the extract), as well as exerts time-and dose-dependent increases in strongly scavenged free radicals (average 14.8 ± 1.97 µg/mL IC50 at 40 min). Cell viabilities after exposure to the extracts (1 and 10 µg/mL) were similar to the viability of non-treated cells. Cytokine mRNA expressions were significantly suppressed by the extracts (1 and 10 µg/mL) at 6 hours (h) after exposure. Interleukin-6 secretion was dose-dependently suppressed 2 h after incubation with the extract, at 1–10 µg/mL in non-stimulated cells, and at 5 and 10 µg/mL in LPS-stimulated cells. Similar patterns were also observed at 24 h after incubation with the extract (at 1–10 µg/mL in non-stimulated cells, and at 10 µg/mL in the LPS-stimulated cells). Soluble intracellular vascular adhesion molecules (sICAM-1) secreted from non-stimulated cells and LPS-stimulated cells were similarly suppressed in a dose-dependent manner after 24 h exposure to the extracts, but not after 2 h. In addition, sICAM-1 concentration after 24 h treatment was positively related to IL-6 levels after 2 h and 24 h exposure (r = 0.418, P = 0.003, and r = 0.524, P < 0.001, respectively). CONCLUSIONS This study demonstrates that C. turczaninowii modulates the arterial inflammatory response, and indicates the potential to be applied as a therapeutic use for atherosclerosis.
Collapse
Affiliation(s)
- Youn Kyoung Son
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon 22689, Republic of Korea
| | - So Ra Yoon
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea.,Center for Silver-targeted Biomaterials, Brain Busan 21 Plus Program, Dong-A University, 37 Nakdongdae-ro, 550beon-gil Saha-gu, Busan, 49315, Republic of Korea.,Institute of Health Insurance and Clinical Research, National Health Insurance Service Ilsan Hospital, Gyeonggi, 10444, Republic of Korea
| | - Woo Young Bang
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon 22689, Republic of Korea
| | - Chang-Hwan Bae
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon 22689, Republic of Korea
| | - Joo-Hong Yeo
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon 22689, Republic of Korea
| | - Rimkyo Yeo
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea
| | - Juhyun An
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea
| | - Juhyun Song
- Departments of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea.,Center for Silver-targeted Biomaterials, Brain Busan 21 Plus Program, Dong-A University, 37 Nakdongdae-ro, 550beon-gil Saha-gu, Busan, 49315, Republic of Korea
| |
Collapse
|
8
|
Fingolimod Suppresses the Proinflammatory Status of Interferon-γ-Activated Cultured Rat Astrocytes. Mol Neurobiol 2019; 56:5971-5986. [PMID: 30701416 DOI: 10.1007/s12035-019-1481-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022]
Abstract
Astroglia, the primary homeostatic cells of the central nervous system, play an important role in neuroinflammation. They act as facultative immunocompetent antigen-presenting cells (APCs), expressing major histocompatibility complex (MHC) class II antigens upon activation with interferon (IFN)-γ and possibly other proinflammatory cytokines that are upregulated in disease states, including multiple sclerosis (MS). We characterized the anti-inflammatory effects of fingolimod (FTY720), an established drug for MS, and its phosphorylated metabolite (FTY720-P) in IFN-γ-activated cultured rat astrocytes. The expression of MHC class II compartments, β2 adrenergic receptor (ADR-β2), and nuclear factor kappa-light-chain enhancer of activated B cells subunit p65 (NF-κB p65) was quantified in immunofluorescence images acquired by laser scanning confocal microscopy. In addition, MHC class II-enriched endocytotic vesicles were labeled by fluorescent dextran and their mobility analyzed in astrocytes subjected to different treatments. FTY720 and FTY720-P treatment significantly reduced the number of IFN-γ-induced MHC class II compartments and substantially increased ADR-β2 expression, which is otherwise small or absent in astrocytes in MS. These effects could be partially attributed to the observed decrease in NF-κB p65 expression, because the NF-κB signaling cascade is activated in inflammatory processes. We also found attenuated trafficking and secretion from dextran-labeled endo-/lysosomes that may hinder efficient delivery of MHC class II molecules to the plasma membrane. Our data suggest that FTY720 and FTY720-P at submicromolar concentrations mediate anti-inflammatory effects on astrocytes by suppressing their action as APCs, which may further downregulate the inflammatory process in the brain, constituting the therapeutic effect of fingolimod in MS.
Collapse
|
9
|
The emergence of neuroepidemiology, neurovirology and neuroimmunology: the legacies of John F. Kurtzke and Richard ‘Dick’ T. Johnson. J Neurol 2016; 264:817-828. [DOI: 10.1007/s00415-016-8293-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 12/11/2022]
|
10
|
Sun J, Dou W, Zhao Y, Hu J. A comparison of the effects of topical treatment of calcipotriol, camptothecin, clobetasol and tazarotene on an imiquimod-induced psoriasis-like mouse model. Immunopharmacol Immunotoxicol 2013; 36:17-24. [PMID: 24286371 DOI: 10.3109/08923973.2013.862542] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The interleukin-23/interleukin 17A (IL-23/IL-17A) cytokine axis plays a critical role in the pathogenesis of psoriasis. In this study, we report the effects of topical calcipotriol, camptothecin, clobetasol and tazarotene on the treatment of imiquimod (IMQ)-induced psoriasis-like inflammation, the development of which is dependent on the IL-23/IL-17A axis. IMQ-induced epidermal hyperplasia and inflammation in the BALB/c mouse ear were significantly inhibited following clobetasol treatment but not calcipotriol, camptothecin or tazarotene treatments. Real-time polymerase chain reaction showed that the mRNA levels of IL-17A, IL-17F, IL-22, IL-1β, IL-6 and TNF-α in ear skin were significantly decreased by clobetasol. In addition, we observed that calcipotriol, camptothecin and tazarotene failed to show any inhibitory effects on the IL-23/IL-17A/IL-22 axis. We also found that clobetasol treatment inhibited the proliferation of γδ T cells and C-C chemokine receptor type 6 (CCR6) expression induced by IMQ. Calcipotriol, camptothecin and tazarotene not only failed to inhibit this proliferation but also enhanced retinoic acid-related orphan receptor γ (RORγ) expression in IMQ-induced psoriasis-like inflammation. In conclusion, we suggest that clobetasol induces the relief of IMQ-induced psoriasis-like inflammation in a mouse model but that calcipotriol, camptothecin and tazarotene cannot. Therefore, we suggest that more in-depth studies on pharmacological effects of tazarotene, camptothecin and calcipotriol should be carried out.
Collapse
Affiliation(s)
- Jun Sun
- Department of Pharmacy, Changhai Hospital, the Second Military Medical University , Shanghai , China and
| | | | | | | |
Collapse
|
11
|
|
12
|
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: 51] [Impact Index Per Article: 3.6] [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
|
13
|
Autoimmune Processes in the Central Nervous System. HANDBOOK OF NEUROCHEMISTRY AND MOLECULAR NEUROBIOLOGY 2008. [PMCID: PMC7121640 DOI: 10.1007/978-0-387-30398-7_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In this chapter we discuss the factors that contribute to the unique immunological environment of the central nervous system and the mechanisms that may account for the development of autoimmunity within the CNS, including infectious agents as inducers of autoimmune disease. Consideration is given to a variety of human neurological diseases of autoimmune or presumed autoimmune etiology: autism, neuromyelitis optica, neuromyotonia, schizophrenia, lethargic encephalitis and stiff‐man syndrome. Also, we discuss autoimmunity as a possible mediator of CNS repair and examples of the protective effects of bacterial and helminth infections on CNS disease. Multiple sclerosis and models of multiple sclerosis are discussed with special attention given to the Theiler's virus‐induced demyelination model.
Collapse
|
14
|
Rothlein R, Jaeger JR. Treatment of inflammatory diseases with a monoclonal antibody to intercellular adhesion molecule 1. CIBA FOUNDATION SYMPOSIUM 2007; 189:200-8; discussion 208-11. [PMID: 7587634 DOI: 10.1002/9780470514719.ch14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A mouse monoclonal antibody to intercellular adhesion molecule 1 (ICAM-1) has been evaluated in multiple animal models of inflammation as well as in the clinic. Anti-ICAM-1 has been found to protect against allograft rejection and ischaemia/reperfusion injury in non-human primates and rabbits. In open-label phase I-II studies, anti-ICAM-1 appears to have prolonged kidney allograft survival when used as induction therapy in conjunction with traditional triple immunosuppressive therapy. Anti-ICAM-1 has shown beneficial effects in the treatment of rheumatoid arthritis when given for five days. Most patients receiving anti-ICAM-1 made antibodies to the mouse immunoglobulin.
Collapse
Affiliation(s)
- R Rothlein
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT 06877, USA
| | | |
Collapse
|
15
|
Mrass P, Weninger W. Immune cell migration as a means to control immune privilege: lessons from the CNS and tumors. Immunol Rev 2006; 213:195-212. [PMID: 16972905 DOI: 10.1111/j.1600-065x.2006.00433.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Certain organs, such as the brain, eye, and gonads, are particularly sensitive to damage by inflammation. Therefore, these tissues have developed unique immunological properties that curtail inflammatory responses, a phenomenon termed immune privilege. In addition, by co-opting some of the regulatory cues operant in immune privilege in normal organs, tumors can evade immunosurveillance. While many different mechanisms contribute to immune privilege, there is evidence that leukocyte migration is an important checkpoint in its control. This hypothesis is based on the fact that leukocyte entry into these organs is restricted by physical barriers and that the collapse of these obstacles marks a critical step in the development of inflammatory/autoimmune disease at these sites. Numerous studies in a variety of experimental systems have characterized the molecular and cellular mechanisms involved in leukocyte homing to immune-privileged organs. Recently, two-photon microscopy has revealed critical insights into the events occurring in the extravascular space of immune-privileged organs, including locomotion patterns and interactive behavior of leukocytes in the interstitial space. Here, we review our current understanding of immune cell migration to and within immune-privileged organs and highlight how this knowledge may be exploited for immunotherapeutic purposes.
Collapse
Affiliation(s)
- Paulus Mrass
- Immunology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | |
Collapse
|
16
|
Rentzos M, Paraskevas GP, Kapaki E, Nikolaou C, Zoga M, Rombos A, Tsoutsou A, Vassilopoulos D D. Interleukin-12 is reduced in cerebrospinal fluid of patients with Alzheimer's disease and frontotemporal dementia. J Neurol Sci 2006; 249:110-4. [PMID: 16843497 DOI: 10.1016/j.jns.2006.05.063] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 04/14/2006] [Accepted: 05/26/2006] [Indexed: 11/19/2022]
Abstract
UNLABELLED Interleukin-12 is a heterodimeric cytokine produced by activated blood monocytes, macrophages and glial cells. It enhances differentiation and proliferation of T cells and increases production of proinflammatory cytokines, such as Interferon-gamma and Tumor Necrosis Factor-alpha. There is little information about the involvement of IL-12 in the pathophysiology of Alzheimer's disease (AD) and other tauopathies. OBJECTIVES The objective of our study was to assess the role of IL-12 as a potential marker of immune reactions in patients with AD and frontotemporal dementia (FTD). PATIENTS AND METHODS We measured by immunoassay cerebrospinal fluid (CSF) IL-12 levels in 19 patients with AD and 7 patients with FTD in comparison with CSF IL-12 levels in 30 patients with non-inflammatory neurological diseases served as neurological control patients (NCTRL). IL-12 levels were correlated with age, age of disease onset, disease duration, MMSE score, and rate of dementia progression. Abeta42 and Total tau (tau(T)) levels in CSF were also measured. RESULTS Patients with AD had significantly lower CSF IL-12 levels compared with NCTRL patients (p<0.001). Patients with FTD had also lower CSF IL-12 levels compared with NCTRL patients (p<0.05). Age, sex, disease duration and MMSE score did not affect IL-12 levels in any of the groups. In AD a significant positive correlation was noted between IL-12 levels and tau(T) levels (Rs=0.46, p=0.048). CONCLUSIONS Our findings may suggest a reduced inflammatory reaction during the course of AD and FTD. A neurotrophic role of IL-12 and other proinflammatory cytokines cannot be excluded.
Collapse
Affiliation(s)
- Michael Rentzos
- Department of Neurology, Aeginition Hospital, Athens National University, School of Medicine, 72-74 Vas. Sophias Ave, Athens 11528, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Kepplinger B, Baran H, Kainz A, Ferraz-Leite H, Newcombe J, Kalina P. Age-Related Increase of Kynurenic Acid in Human Cerebrospinal Fluid – IgG and β 2-Microglobulin Changes. Neurosignals 2005; 14:126-35. [PMID: 16088227 DOI: 10.1159/000086295] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2004] [Accepted: 03/10/2005] [Indexed: 11/19/2022] Open
Abstract
Kynurenic acid (KYNA) is an endogenous metabolite in the kynurenine pathway of tryptophan degradation and is an antagonist at the glycine site of the N-methyl-D-aspartate as well as at the alpha 7 nicotinic cholinergic receptors. In the brain tissue KYNA is synthesised from L-kynurenine by kynurenine aminotransferases (KAT) I and II. A host of immune mediators influence tryptophan degradation. In the present study, the levels of KYNA in cerebrospinal fluid (CSF) and serum in a group of human subjects aged between 25 and 74 years were determined by using a high performance liquid chromatography method. In CSF and serum KAT I and II activities were investigated by radioenzymatic assay, and the levels of beta(2)-microglobulin, a marker for cellular immune activation, were determined by ELISA. The correlations between neurochemical and biological parameters were evaluated. Two subject groups with significantly different ages, i.e. <50 years and >50 years, p < 0.001, showed statistically significantly different CSF KYNA levels, i.e. 2.84 +/- 0.16 fmol/microl vs. 4.09 +/- 0.14 fmol/microl, p < 0.001, respectively; but this difference was not seen in serum samples. Interestingly, KYNA is synthesised in CSF principally by KAT I and not KAT II, however no relationship was found between enzyme activity and ageing. A positive relationship between CSF KYNA levels and age of subjects indicates a 95% probability of elevated CSF KYNA with ageing (R = 0.6639, p = 0.0001). KYNA levels significantly correlated with IgG and beta(2)-microglobulin levels (R = 0.5244, p = 0.0049; R = 0.4253, p = 0.043, respectively). No correlation was found between other biological parameters in CSF or serum. In summary, a positive relationship between the CSF KYNA level and ageing was found, and the data would suggest age-dependent increase of kynurenine metabolism in the CNS. An enhancement of CSF IgG and beta(2)-microglobulin levels would suggest an activation of the immune system during ageing. Increased KYNA metabolism may be involved in the hypofunction of the glutamatergic and/or nicotinic cholinergic neurotransmission in the ageing CNS.
Collapse
Affiliation(s)
- Berthold Kepplinger
- Department of Neurology, Klinikum Mostviertel Amstetten, General Hospital, Amstetten, Austria
| | | | | | | | | | | |
Collapse
|
18
|
Choi WH, Ji KA, Jeon SB, Yang MS, Kim H, Min KJ, Shong M, Jou I, Joe EH. Anti-inflammatory roles of retinoic acid in rat brain astrocytes: Suppression of interferon-gamma-induced JAK/STAT phosphorylation. Biochem Biophys Res Commun 2005; 329:125-31. [PMID: 15721283 DOI: 10.1016/j.bbrc.2005.01.110] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Indexed: 10/25/2022]
Abstract
The anti-inflammatory effect of retinoic acid (RA) has been investigated for several decades. However, the underlying mechanisms responsible for this effect are largely unknown. In this study, we demonstrate that 9-cis-RA (cRA) and all-trans-RA (tRA) inhibit interferon-gamma (IFN-gamma)-induced inflammatory responses in astrocytes. In primary cultured rat brain astrocytes and C6 astroglioma cells, both cRA and tRA decreased IFN-gamma-induced expression of interferon regulatory factor-1. Both RA isoforms also reduced IFN-gamma-induced activation of signal transducers and activators of transcription (STAT)1, STAT3, Janus kinase (JAK)1, and JAK2. This inhibitory effect was significant when cells were pre-treated with RA prior to IFN-gamma. Furthermore, the effect of pre-treated RA was abolished in the presence of cycloheximide, indicating a requirement for de novo protein synthesis. Suppressors of cytokine signaling (SOCS), which are negative regulators of the JAK/STAT pathway, may be candidate mediators of the anti-inflammatory function of RA. Both cRA and tRA induced SOCS3 mRNA expression. These results suggest that RA induces an anti-inflammatory effect by suppressing the activation of the JAK/STAT pathway in IFN-gamma-treated astrocytes. SOCS3 may be at least one of the mechanisms that mediate the anti-inflammatory roles of RA.
Collapse
Affiliation(s)
- Woo-Hyuck Choi
- Neuroscience Graduate Program, Ajou University School of Medicine, Suwon 442-721, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Jeon SB, Ji KA, You HJ, Kim JH, Jou I, Joe EH. Nordihydroguaiaretic acid inhibits IFN-γ-induced STAT tyrosine phosphorylation in rat brain astrocytes. Biochem Biophys Res Commun 2005; 328:595-600. [PMID: 15694390 DOI: 10.1016/j.bbrc.2005.01.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Indexed: 01/05/2023]
Abstract
The Janus kinase (JAK) and signal transducers and activators of transcription (STAT) signal cascades are major pathways that mediate the inflammatory functions of interferon-gamma (IFN-gamma), an important pro-inflammatory cytokine. Therefore, regulation of JAK/STAT signaling should modulate IFN-gamma-mediated inflammation. In this study, we found that nordihydroguaiaretic acid (NDGA), a well-known lipoxygenase (LO) inhibitor, suppressed IFN-gamma-induced inflammatory responses in brain astrocytes. In the presence of NDGA, interferon regulatory factor-1 expression was significantly reduced. Expression of monocyte chemotactic protein-1 and interferon-gamma inducible protein-10 mRNA in response to IFN-gamma was significantly suppressed in the presence of NDGA, as was tyrosine-phosphorylation of JAK and STAT. However, the 5-LO products, leukotriene B(4) (LTB(4)) and leukotriene C(4), were not detected in cells treated with IFN-gamma, indicating that the effect of NDGA seemed to be independent of 5-LO inhibition. In addition, two other 5-LO inhibitors (Rev5901 and AA861) did not mimic the effect of NDGA, and the 5-LO metabolites, 5-hydroxyeicosatetraenoic acid and LTB(4), were unable to reverse NDGA-driven suppression of STAT activation or affect basal STAT phosphorylation. Taken together, these results suggest that NDGA regulates IFN-gamma-mediated inflammation through mechanisms unrelated to the inhibition of 5-LO.
Collapse
Affiliation(s)
- Sae-Bom Jeon
- Interdisciplinary Course for Neuroscience and Technology, Ajou University School of Medicine, Suwon 442-721, South Korea
| | | | | | | | | | | |
Collapse
|
20
|
Rentzos M, Michalopoulou M, Nikolaou C, Cambouri C, Rombos A, Dimitrakopoulos A, Vassilopoulos D. The role of soluble intercellular adhesion molecules in neurodegenerative disorders. J Neurol Sci 2004; 228:129-35. [PMID: 15694193 DOI: 10.1016/j.jns.2004.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2004] [Revised: 10/08/2004] [Accepted: 11/02/2004] [Indexed: 12/11/2022]
Abstract
UNLABELLED Immunological disturbances have been implicated in the pathogenesis of some neurodegenerative disorders like Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). Adhesion molecules are markers of activated endothelial cells upregulated by action of cytokines. MATERIALS AND METHODS To investigate the activation or not of the vascular cells in AD and ALS, serum soluble intercellular adhesion molecule-1 (ICAM-1) and soluble E-selectin were evaluated (enzyme-like immunosorbent assay, ELISA) in 22 patients with Alzheimer's disease (AD), 20 patients with amyotrophic lateral sclerosis (ALS), 34 patients with non-inflammatory neurological diseases (NIND) and 15 control subjects. RESULTS Patients with AD had higher s-ICAM-1 levels compared to NIND patients and control subjects (p<0.0027 and p<0.04, respectively). Patients with ALS had not higher s-ICAM-1 levels compared to NIND patients and control subjects (p<0.21 and p<0.31, respectively). Soluble-E-selectin levels in AD and ALS patients were not statistically different compared to NIND patients and controls (p<0.4, p<0.9 and p<0.3, p<0.19, respectively). CONCLUSIONS The presence of high s-ICAM values may be related to immunological processes involved in pathogenetic mechanisms of AD. The not statistically significant values of s-E selectin, a glycoprotein considered an exclusive marker of endothelial activation, seem to suggest the neural rather than the endothelial s-ICAM origin in patients with AD. The low values of s-ICAM-1 and sE-selectin in the serum of ALS patients do not exclude the presence of an unconventional immunological abnormality in this disorder.
Collapse
Affiliation(s)
- M Rentzos
- Department of Neurology, Aeginition Hospital, Athens Medical School, 72-74 Vas.Sophias Av, Greece.
| | | | | | | | | | | | | |
Collapse
|
21
|
Rentzos M, Michalopoulou M, Nikolaou C, Cambouri C, Rombos A, Dimitrakopoulos A, Kapaki E, Vassilopoulos D. Serum levels of soluble intercellular adhesion molecule-1 and soluble endothelial leukocyte adhesion molecule-1 in Alzheimer's disease. J Geriatr Psychiatry Neurol 2004; 17:225-31. [PMID: 15533994 DOI: 10.1177/0891988704269822] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Serum soluble intercellular adhesion molecule-1 (s-ICAM-1) and soluble E-selectin (s-ELAM-1) were evaluated in 25 patients with Alzheimer's disease (AD), 54 patients with noninflammatory neurological diseases (NIND), and 15 control subjects. Patients with AD had a higher s-ICAM-1 level compared with the NIND patients and the control subjects (P< .001 and P< .04, respectively). The presence of high s-ICAM-1 values may be related to immunological processes involved in pathogenetic mechanisms of AD. The not statistically significant values of (s-ELAM-1), a glycoprotein considered an exclusive marker of endothelial activation, compared with the NIND patients and healthy subjects (P< .47 and P< .17, respectively), seem to suggest the neural rather than the endothelial s-ICAM origin in patients with AD.
Collapse
Affiliation(s)
- M Rentzos
- Department of Neurology, Aeginition Hospital, Athens Medical School, Vass. Sophias av. 72-74, 11528 Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hang CH, Shi JX, Tian J, Li JS, Wu W, Yin HX. Effect of systemic LPS injection on cortical NF-κB activity and inflammatory response following traumatic brain injury in rats. Brain Res 2004; 1026:23-32. [PMID: 15476694 DOI: 10.1016/j.brainres.2004.07.090] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2004] [Indexed: 11/18/2022]
Abstract
The aim of current study is to investigate the effect of systemic administration of lipopolysaccharide (LPS) on the temporal pattern of cortical nuclear factor kappa B (NF-kappaB) binding activity, inflammatory response and secondary damage in the injured brain following traumatic brain injury (TBI). Right parietal cortical contusion in rats was made by using weight-dropping method. The rats were randomly divided into sham, LPS, TBI and TBI-LPS groups, with LPS injected intraperitoneally. NF-kappaB binding activity, cytokines, intercellular adhesion molecule-1 (ICAM-1) and brain damage were detected by electrophoretic mobility shift assay (EMSA), enzyme-linked immunosorbent assay (ELISA), immunohistochemistry and terminal deoxynucleotidyl-transferase-mediated biotin-dUTP nick end labeling (TUNEL) apoptosis, respectively. The results showed that systemic administration of LPS following TBI could induce an immediate, strong and persistent upregulation of NF-kappaB, tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6) and ICAM-1 in the area surrounding the injured brain. As compared with rats of sham, LPS and TBI groups, NF-kappaB binding activity, TNF-alpha and IL-6 were significantly upregulated in the surrounding cortex of injured site as early as 3 h postinjury when challenged with LPS, kept at high level up to 7-days postinjury. ICAM-1-positive vessels and apoptotic TUNEL-positive cells in the injured brain were also significantly increased in TBI-LPS rats. It was concluded that inflammatory response and secondary brain damage occurred in the injured brain could be highly exacerbated by endotoxemia.
Collapse
Affiliation(s)
- Chun-hua Hang
- Department of Neurosurgery, Jinling Hospital, Clinical School of Medicine, Nanjing University, Nanjing 210002, PR China.
| | | | | | | | | | | |
Collapse
|
23
|
Stüve O, Prod'homme T, Youssef S, Dunn S, Neuhaus O, Weber M, Hartung HP, Steinman L, Zamvil SS. Statins as potential therapeutic agents in multiple sclerosis. Curr Neurol Neurosci Rep 2004; 4:237-44. [PMID: 15102350 DOI: 10.1007/s11910-004-0044-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase inhibitors (ie, statins) are oral cholesterol-lowering drugs. Statins are well tolerated and have an excellent safety record. These agents competitively inhibit HMG CoA reductase, which is the enzyme that catalyzes the conversion of HMG CoA to L-mevalonate. Although L-mevalonate is a key intermediate in cholesterol synthesis, several of its metabolites are involved in post-translational modification of specific proteins involved in cell proliferation and differentiation. Thus, independent of their cholesterol-reducing properties, statins have important pleiotropic biologic effects. Recent reports indicate that statins have anti-inflammatory and neuroprotective properties. Whether statins will be of clinical benefit for patients with multiple sclerosis and other neurodegenerative diseases of the central nervous system will only be known after they are evaluated in prospective randomized clinical trials.
Collapse
Affiliation(s)
- Olaf Stüve
- Department of Neurology, University of California-San Francisco, 521 Parnassus Avenue, C-440, San Francisco, CA 94143-0114, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Somersalo K, Anikeeva N, Sims TN, Thomas VK, Strong RK, Spies T, Lebedeva T, Sykulev Y, Dustin ML. Cytotoxic T lymphocytes form an antigen-independent ring junction. J Clin Invest 2004. [PMID: 14702108 DOI: 10.1172/jci200419337] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Immunological synapses are organized cell-cell junctions between T lymphocytes and APCs composed of an adhesion ring, the peripheral supramolecular activation cluster (pSMAC), and a central T cell receptor cluster, the central supramolecular activation cluster (cSMAC). In CD8(+) cytotoxic T lymphocytes, the immunological synapse is thought to facilitate specific killing by confining cytotoxic agents to the synaptic cleft. We have investigated the interaction of human CTLs and helper T cells with supported planar bilayers containing ICAM-1. This artificial substrate provides identical ligands to CD4(+) and CD8(+) T cells, allowing a quantitative comparison. We found that cytotoxic T lymphocytes form a ring junction similar to a pSMAC in response to high surface densities of ICAM-1 in the planar bilayer. MICA, a ligand for NKG2D, facilitated the ring junction formation at lower surface densities of ICAM-1. ICAM-1 and MICA are upregulated in tissues by inflammation- and stress-associated signaling, respectively. Activated CD8(+) T cells formed fivefold more ring junctions than did activated CD4(+) T cells. The ring junction contained lymphocyte function associated antigen-1 and talin, but did not trigger polarization and granule translocation to the interface. This result has specific implications for the mechanism of effective CTL hunting for antigen in tissues. Abnormalities in this process may alter CTL reactivity.
Collapse
Affiliation(s)
- Kristina Somersalo
- Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York, New York 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Pathogenic mechanisms of central nervous system (CNS) involvement in systemic lupus erythematosus (SLE) remain unknown. We recently reported the presence of autoantibodies in the brain tissue ex vivo of autoimmune MRL/lpr mice. We postulated that at least some of these autoantibodies are produced in situ because of B-cell entry into the brain. The blood-brain barrier (BBB) blocks the entry of most large molecules and cells into the brain. In certain CNS pathologies, however, immune cells gain entry due to elevated expression of adhesion molecules. This study looked at adhesion molecule expression, ICAM-1 and VCAM-1, in the brains of MRL/lpr mice. Using immunofluorescent antibody binding assays and confocal laser imaging, we show that expression of ICAM-1 and VCAM-1 is elevated in MRL/lpr mice brains at 4 months of age as compared to age-matched controls. These results suggest a possible mechanism for leukocyte entry into the brains of autoimmune mice that in turn suggest immune-mediated pathology in CNS-lupus.
Collapse
Affiliation(s)
- Andleeb Zameer
- Department of Microbiology, Arizona State University,Tempe, AZ 85287-2701, USA
| | | |
Collapse
|
26
|
Somersalo K, Anikeeva N, Sims TN, Thomas VK, Strong RK, Spies T, Lebedeva T, Sykulev Y, Dustin ML. Cytotoxic T lymphocytes form an antigen-independent ring junction. J Clin Invest 2004; 113:49-57. [PMID: 14702108 PMCID: PMC300769 DOI: 10.1172/jci19337] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2003] [Accepted: 10/28/2003] [Indexed: 11/17/2022] Open
Abstract
Immunological synapses are organized cell-cell junctions between T lymphocytes and APCs composed of an adhesion ring, the peripheral supramolecular activation cluster (pSMAC), and a central T cell receptor cluster, the central supramolecular activation cluster (cSMAC). In CD8(+) cytotoxic T lymphocytes, the immunological synapse is thought to facilitate specific killing by confining cytotoxic agents to the synaptic cleft. We have investigated the interaction of human CTLs and helper T cells with supported planar bilayers containing ICAM-1. This artificial substrate provides identical ligands to CD4(+) and CD8(+) T cells, allowing a quantitative comparison. We found that cytotoxic T lymphocytes form a ring junction similar to a pSMAC in response to high surface densities of ICAM-1 in the planar bilayer. MICA, a ligand for NKG2D, facilitated the ring junction formation at lower surface densities of ICAM-1. ICAM-1 and MICA are upregulated in tissues by inflammation- and stress-associated signaling, respectively. Activated CD8(+) T cells formed fivefold more ring junctions than did activated CD4(+) T cells. The ring junction contained lymphocyte function associated antigen-1 and talin, but did not trigger polarization and granule translocation to the interface. This result has specific implications for the mechanism of effective CTL hunting for antigen in tissues. Abnormalities in this process may alter CTL reactivity.
Collapse
MESH Headings
- Antigen-Presenting Cells/cytology
- Antigen-Presenting Cells/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cell Adhesion/immunology
- Clone Cells
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Microscopy, Confocal
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell/analysis
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/physiology
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/immunology
Collapse
Affiliation(s)
- Kristina Somersalo
- Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York, New York 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Studies in animal models have suggested a role for stem cells in repair and regeneration of the nervous system. Human equivalents of stem and precursor cells have been isolated and their efficacy is being evaluated in rodent and primate models. Difficulties exist in translating results of these preclinical models to therapy in humans. Evolutionary differences among rodents, primates, and humans; fundamental differences in the anatomy and physiology; differences in immune responses in xenotransplant models; the paucity of good transplant models of chronic disease; and allelic variability in the cells themselves make any study evaluating the efficacy of cells in transplant models difficult to interpret. As no better alternatives to testing in animals exist, we suggest that at this early stage a considered step-by-step approach to testing and comparison of different transplant strategies in isolation will prepare us better for clinical trials than simple evaluation of functional outcomes in various models of disease. We emphasize that we do not recommend delaying or abandoning clinical trials; rather, we suggest that one anticipate failures and design experiments and data collection such that we learn from these failures to ensure future success in as rapid a time frame as possible.
Collapse
Affiliation(s)
- Irene Ginis
- Gerontology Research Center, Stem Cell Biology Unit/Laboratory of Neuroscience, National Institute on Aging, National Institutes of Health, 5600 Nathan Shock Drive, Room 4E02, Baltimore, MD 21224, USA
| | | |
Collapse
|
28
|
Abstract
1. Neural stem cells can be cultured from the CNS of different mammalian species at many stages of development. They have an extensive capacity for self-renewal and will proliferate ex vivo in response to mitogenic growth factors or following genetic modification with immortalising oncogenes. Neural stem cells are multipotent since their differentiating progeny will give rise to the principal cellular phenotypes comprising the mature CNS: neurons, astrocytes and oligodendrocytes. 2. Neural stem cells can also be derived from more primitive embryonic stem (ES) cells cultured from the blastocyst. ES cells are considered to be pluripotent since they can give rise to the full cellular spectrum and will, therefore, contribute to all three of the embryonic germ layers: endoderm, mesoderm and ectoderm. However, pluripotent cells have also been derived from germ cells and teratocarcinomas (embryonal carcinomas) and their progeny may also give rise to the multiple cellular phenotypes contributing to the CNS. In a recent development, ES cells have also been isolated and grown from human blastocysts, thus raising the possibility of growing autologous stem cells when combined with nuclear transfer technology. 3. There is now an emerging recognition that the adult mammalian brain, including that of primates and humans, harbours stem cell populations suggesting the existence of a previously unrecognised neural plasticity to the mature CNS, and thereby raising the possibility of promoting endogenous neural reconstruction. 4. Such reports have fuelled expectations for the clinical exploitation of neural stem cells in cell replacement or recruitment strategies for the treatment of a variety of human neurological conditions including Parkinson's disease (PD), Huntington's disease, multiple sclerosis and ischaemic brain injury. Owing to their migratory capacity within the CNS, neural stem cells may also find potential clinical application as cellular vectors for widespread gene delivery and the expression of therapeutic proteins. In this regard, they may be eminently suitable for the correction of genetically-determined CNS disorders and in the management of certain tumors responsive to cytokines. Since large numbers of stem cells can be generated efficiently in culture, they may obviate some of the technical and ethical limitations associated with the use of fresh (primary) embryonic neural tissue in current transplantation strategies. 5. While considerable recent progress has been made in terms of developing new techniques allowing for the long-term culture of human stem cells, the successful clinical application of these cells is presently limited by our understanding of both (i) the intrinsic and extrinsic regulators of stem cell proliferation and (ii) those factors controlling cell lineage determination and differentiation. Although such cells may also provide accessible model systems for studying neural development, progress in the field has been further limited by the lack of suitable markers needed for the identification and selection of cells within proliferating heterogeneous populations of precursor cells. There is a further need to distinguish between the committed fate (defined during normal development) and the potential specification (implying flexibility of fate through manipulation of its environment) of stem cells undergoing differentiation. 6. With these challenges lying ahead, it is the opinion of the authors that stem-cell therapy is likely to remain within the experimental arena for the foreseeable future. In this regard, few (if any) of the in vivo studies employing neural stem cell grafts have shown convincingly that behavioural recovery can be achieved in the various model paradigms. Moreover, issues relating to the quality control of cultured cells and their safety following transplantation have only begun to be addressed. 7. While on the one hand cell biotechnologists have been quick to realise the potential commercial value, human stem cell research and its clinical applications has been the subject of intense ethical and legislative considerations. The present chapter aims to review some recent aspects of stem cell research applicable to developmental neurobiology and the potential applications in clinical neuroscience.
Collapse
Affiliation(s)
- T Ostenfeld
- MRC Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
29
|
Dietrich PY, Walker PR, Saas P. Death receptors on reactive astrocytes: a key role in the fine tuning of brain inflammation? Neurology 2003; 60:548-54. [PMID: 12607528 DOI: 10.1212/01.wnl.0000042049.74547.7f] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Immune responses protect the CNS against pathogens. However, the fact that there is little dispensable tissue in the brain makes regulation necessary to avoid disastrous immune-mediated damage. Astrocytes respond vigorously to any brain injury (e.g., tumor, stroke, AD, MS, HIV) and are postulated to play an important role in the fine tuning of brain inflammation. The authors propose that astrocytes use death receptors to modulate pro- and anti-inflammatory effects.
Collapse
Affiliation(s)
- Pierre-Yves Dietrich
- Hôpital Universitaire, Division d'Oncologie, Laboratoire d'Immunologie des Tumeurs, Switzerland.
| | | | | |
Collapse
|
30
|
Weaver LC, Marsh DR, Gris D, Meakin SO, Dekaban GA. Central mechanisms for autonomic dysreflexia after spinal cord injury. PROGRESS IN BRAIN RESEARCH 2002; 137:83-95. [PMID: 12440361 DOI: 10.1016/s0079-6123(02)37009-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Lynne C Weaver
- Spinal Cord Injury Laboratory, BioTherapeutics Research Group, John P. Robarts Research Institute, 100 Perth Drive, P.O. Box 5015, London, ON N6A 5K8, Canada.
| | | | | | | | | |
Collapse
|
31
|
Feinstein DL, Heneka MT, Gavrilyuk V, Dello Russo C, Weinberg G, Galea E. Noradrenergic regulation of inflammatory gene expression in brain. Neurochem Int 2002; 41:357-65. [PMID: 12176079 DOI: 10.1016/s0197-0186(02)00049-9] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It is now well accepted that inflammatory events contribute to the pathogenesis of numerous neurological disorders, including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease, and AID's dementia. Whereas inflammation in the periphery is subject to rapid down regulation by increases in anti-inflammatory molecules and the presence of scavenging soluble cytokine receptors, the presence of an intact blood-brain barrier may limit a similar autoregulation from occurring in brain. Mechanisms intrinsic to the brain may provide additional immunomodulatory functions, and whose dysregulation could contribute to increased inflammation in disease. The findings that noradrenaline (NA) reduces cytokine expression in microglial, astroglial, and brain endothelial cells in vitro, and that modification of the noradrenergic signaling system occurs in some brain diseases having an inflammatory component, suggests that NA could act as an endogenous immunomodulator in brain. Furthermore, accumulating studies indicate that modification of the noradrenergic signaling system occurs in some neurodiseases. In this article, we will briefly review the evidence that NA can modulate inflammatory gene expression in vitro, summarize data supporting a similar immunomodulatory role in brain, and present recent data implicating a role for NA in attenuating the cortical inflammatory response to beta amyloid protein.
Collapse
|
32
|
Nam SY, Cho KS, Heo YM, Ha JC, Kim YH, Keun Yi H, Han Hwang P, Kim HM, Podack ER. Regulation of lymphocyte clustering by CD30-mediated ICAM-1 up-regulation. Cell Immunol 2002; 219:38-47. [PMID: 12473266 DOI: 10.1016/s0008-8749(02)00583-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
CD30 is expressed transiently on activated B and T lymphocytes and constitutively on several B- and T cell lymphomas. CD30 functions include participation in negative selection of thymocytes, costimulation of activated T cells, isotype switching of B cells, and regulation of the effector activity of cytotoxic lymphocytes. Although CD30 is not a marker for T helper 2 (TH2) cells, it may participate in the polarization of TH1 and TH2 cells. The pleiotropic functions of CD30 are initiated by interaction of CD30-expressing cells with other immune competent cells expressing CD30-L and providing the signals for modulation of effector cell activity. Here, we report that CD30 signals generated by anti-CD30 on activated, normal murine T cells strongly up-regulate the expression of intercellular adhesion molecule 1 (ICAM-1, CD54), and to a lesser extent, ICAM-2 (CD102). CD30 signals moreover delay the subsequent decline of ICAM expression. CD30 cross-linking did not alter the expression of CD11a/CD18 (LFA-1), the counter receptor for ICAM abundant on T cells. CD30-mediated ICAM-1 up-regulation is independent of cytokine secretion and appears to be transmitted directly through NF-kappaB activation. CD30-mediated up-regulation of ICAM-1 expression led to a significant increase in cluster formation of lymph node cells. Increased lymphocyte self-aggregation mediated by CD30 may set the stage for fraternal signaling to modulate lymphocyte function.
Collapse
Affiliation(s)
- Sang Yun Nam
- Department of Microbiology, School of Natural Science, Jeonju University, 560-759, Jeonju, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Stüve O, Cree BC, von Büdingen HC, Yousef S, Bowen JD, Genain CP, Hauser SL, Steinman L, Zamvil SS. Approved and future pharmacotherapy for multiple sclerosis. Neurologist 2002; 8:290-301. [PMID: 12803676 DOI: 10.1097/00127893-200209000-00002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Pharmacotherapy for relapsing-remitting multiple sclerosis (MS) advanced with the demonstration that interferon beta and glatiramer acetate improve the clinical course of this disease. Mitoxantrone is the first drug approved by the Food and Drug Administration for treatment of secondary progressive MS. Despite this progress, the agents presently available are only partially effective, are difficult to administer, and may have significant side effects. Several orally administered immunomodulatory agents are presently being evaluated for treatment of MS. One class of drugs, HMG CoA inhibitors (statins), is safe and well-tolerated and could become another mainstay of MS therapy. REVIEW SUMMARY This article reviews the clinical evidence for approved MS therapies and discusses their mechanisms of action. Furthermore, the clinical and laboratory data suggesting a potential role for statins in MS therapy are discussed. CONCLUSIONS Although treatment with interferon beta, glatiramer acetate, and mitoxantrone, the approved therapies, provide important treatment options for patients with relapsing-remitting MS and secondary progressive MS, the potential benefits of other medications, including statins, should be explored in controlled clinical trials.
Collapse
Affiliation(s)
- Olaf Stüve
- Department of Neurology, University of California, San Francisco 94143-0114, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hetier E, Ayala J, Bousseau A, Denèfle P, Prochiantz A. Amoeboid Microglial Cells and not Astrocytes Synthesize TNF-alpha in Swiss Mouse Brain Cell Cultures. Eur J Neurosci 2002; 2:762-768. [PMID: 12106276 DOI: 10.1111/j.1460-9568.1990.tb00466.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The role of tumour necrosis factor (TNF-alpha) in brain physiology and pathology has been the focus of several studies. However, the source of this lymphokine in the central nervous system and the regulation of its synthesis is still poorly understood. We have therefore used purified astrocytes and brain macrophages in culture to compare the abilities of these two cell types to synthesize TNF-alpha and its mRNA. We find that, in the Swiss mouse, no significant TNF activity or TNF-alpha mRNA are produced by astrocytes, even following activation with lipopolysaccharides (LPS). On the other hand, purified microglial cells express a cytotoxic activity able to kill TNF-sensitive LM cells. Part of this activity is released into the culture medium and part remains bound to the membrane after mild paraformaldehyde treatment, demonstrating the existence in the culture of the soluble and membrane-bound forms of TNF activity. The fact that amoeboid microglial cells, and not astrocytes, are the actual source of TNF in brain cultures was further demonstrated by Northern blot analysis and in situ hybridization using a TNF-alpha specific oligonucleotide probe. The definition of the cell type which, in the CNS, is responsible for TNF synthesis will allow the regulation of this lymphokine to be analysed and opens the way for a better understanding of the interactions between amoeboid microglial cells and the other cell types which make up the nervous system.
Collapse
Affiliation(s)
- Emmanuelle Hetier
- INSERM U114, and CNRS URA 1414, Chaire de Neuropharmacologie, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
35
|
Tompkins SM, Padilla J, Dal Canto MC, Ting JPY, Van Kaer L, Miller SD. De novo central nervous system processing of myelin antigen is required for the initiation of experimental autoimmune encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4173-83. [PMID: 11937578 DOI: 10.4049/jimmunol.168.8.4173] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We demonstrate the absolute requirement for a functioning class II-restricted Ag processing pathway in the CNS for the initiation of experimental autoimmune encephalomyelitis (EAE). C57BL/6 (B6) mice deficient for the class II transactivator, which have defects in MHC class II, invariant chain (Ii), and H-2M (DM) expression, are resistant to initiation of myelin oligodendrocyte protein (MOG) peptide, MOG(35-55)-specific EAE by both priming and adoptive transfer of encephalitogenic T cells. However, class II transactivator-deficient mice can prime a suboptimal myelin-specific CD4(+) Th1 response. Further, B6 mice individually deficient for Ii and DM are also resistant to initiation of both active and adoptive EAE. Although both Ii-deficient and DM-deficient APCs can present MOG peptide to CD4(+) T cells, neither is capable of processing and presenting the encephalitogenic peptide of intact MOG protein. This phenotype is not Ag-specific, as DM- and Ii-deficient mice are also resistant to initiation of EAE by proteolipid protein peptide PLP(178-191). Remarkably, DM-deficient mice can prime a potent peripheral Th1 response to MOG(35-55), comparable to the response seen in wild-type mice, yet maintain resistance to EAE initiation. Most striking is the demonstration that T cells from MOG(35-55)-primed DM knockout mice can adoptively transfer EAE to wild-type, but not DM-deficient, mice. Together, these data demonstrate that the inability to process antigenic peptide from intact myelin protein results in resistance to EAE and that de novo processing and presentation of myelin Ags in the CNS is absolutely required for the initiation of autoimmune demyelinating disease.
Collapse
MESH Headings
- Adoptive Transfer
- Amino Acid Sequence
- Animals
- Antigen Presentation/genetics
- Antigens, Differentiation, B-Lymphocyte/genetics
- Cell Movement/genetics
- Cell Movement/immunology
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Female
- Glycoproteins/administration & dosage
- Glycoproteins/immunology
- Glycoproteins/metabolism
- Histocompatibility Antigens Class II/genetics
- Immunity, Innate/genetics
- Injections, Subcutaneous
- Lymphocyte Activation/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Myelin Proteolipid Protein/administration & dosage
- Myelin Proteolipid Protein/immunology
- Myelin Proteolipid Protein/metabolism
- Myelin-Oligodendrocyte Glycoprotein
- Nuclear Proteins
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Trans-Activators/deficiency
- Trans-Activators/genetics
Collapse
Affiliation(s)
- Stephen Mark Tompkins
- Department of Microbiology-Immunology and Pathology, Northwestern University Medical School, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- Maria Luisa Cotrina
- Department of Anatomy and Cell Biology, New York Medical College, Valhalla, NY 10595, USA
| | | |
Collapse
|
37
|
DeVito WJ, Stone S. Prenatal exposure to ethanol alters the neuroimmune response to a central nervous system wound in the adult rat. Alcohol 2001; 25:39-47. [PMID: 11668016 DOI: 10.1016/s0741-8329(01)00161-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined the long-term effects of in utero ethanol exposure on the expression of tumor necrosis factor-alpha (TNF-alpha), glial fibrillary acidic protein (GFAP), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and ED1 in the tissue at the site of a central nervous system (CNS) wound. Adult rats obtained from dams fed control diets or an ethanol diet were fed either control diets or an ethanol diet 5 days before and after infliction of a CNS wound. In pair-fed controls, the expression of TNF-alpha, GFAP, ICAM-1, VCAM-1, and ED1 immunoreactive proteins was increased in the tissue at the wound site when compared with that in nonlesioned tissues. In adult rats previously exposed to ethanol in utero and then fed a liquid diet before and after infliction of a CNS wound, however, expression of TNF-alpha, GFAP, and ICAM-1 was markedly decreased when compared with findings in pair-fed controls. In contrast, VCAM-1 levels and ED1 immunoreactive proteins were markedly increased when compared with findings for pair-fed controls. Furthermore, in adult rats exposed to ethanol in utero, re-exposure to ethanol before and after sustaining a CNS wound resulted in further decreases in TNF-alpha, GFAP, and ICAM-1 levels and marked increases in VCAM-1 levels and ED1 immunoreactive proteins. Results of these studies suggest to us that prenatal exposure to ethanol has a long-term immunoteratogenic effect in the CNS, resulting in altered responses of key components of the neuroimmune response, which could leave the animal immunocompromised as an adult.
Collapse
Affiliation(s)
- W J DeVito
- Division of Endocrinology, University of Massachusetts Medical Center, 55 Lake Avenue North, Worcester, MA 01655, USA.
| | | |
Collapse
|
38
|
Pleines UE, Morganti-Kossmann MC, Rancan M, Joller H, Trentz O, Kossmann T. S-100 beta reflects the extent of injury and outcome, whereas neuronal specific enolase is a better indicator of neuroinflammation in patients with severe traumatic brain injury. J Neurotrauma 2001; 18:491-8. [PMID: 11393252 DOI: 10.1089/089771501300227297] [Citation(s) in RCA: 153] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
It has been hypothesized that immunoactivation may contribute to brain damage and affect outcome after traumatic brain injury (TBI). In order to determine the role of inflammation after TBI, we studied the interrelationship of the immune mediators sICAM-1 and IL-6 with the levels of S-100beta and neuronal specific enolase (NSE), both recognized markers of brain damage. In addition, the extent and type of cerebral injury and the neurological outcome were related to these measured markers of injury. An evident elevation of S-100beta (range of means: 2.7-81.4 ng/mL) and NSE (range of means: 2.0-81.3 ng/mL) was observed in CSF of all 13 patients during the first 3 posttraumatic days and decreased over 2 weeks. In parallel, the production of sICAM-1 (range of means: 0.7-11.9 ng/mL) and IL-6 (range of means: 0.1-8.2 ng/mL) was also markedly enhanced in CSF. The CSF means of S-100beta and NSE per patient correlated with IL-6 (r = 0.60, p < 0.05; and r = 0.64, p < 0.05, respectively), whereas the corresponding means in serum showed a significant correlation only between NSE and IL-6 (r = 0.56, p < 0.05). Maximal CSF values of NSE and sICAM-1 correlated with each other (r = 0.57, p < 0.05). The contusion sizes assessed on the CT scans correlated with the means of S-100beta (r = 0.63, p < 0.05) and NSE (r = 0.71, p < 0.05) in CSF and with the mean of S-100beta in serum, although not statistically significant (r = 0.52, p = 0.06), but not with serum NSE. Interestingly, linear regression analysis demonstrated that means of S-100beta in CSF (r = 0.78, p = 0.002) and serum (r = 0.82, p < 0.001) correlated with the GOS. These results indicate that the elevation of these parameters in CSF depends on the extent of injury and that S-100beta may be a predictor of outcome after TBI, whereas NSE reflects better the inflammatory response.
Collapse
Affiliation(s)
- U E Pleines
- Division of Research, University Hospital Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
39
|
Gabryel B, Trzeciak HI. Role of astrocytes in pathogenesis of ischemic brain injury. Neurotox Res 2001; 3:205-21. [PMID: 14715474 DOI: 10.1007/bf03033192] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Astrocytes play an important role in the homeostasis of the CNS both in normal conditions and after ischemic injury. The swelling of astrocytes is observed during and several seconds after brain ischemia. Then ischemia stimulates sequential morphological and biochemical changes in glia and induces its proliferation. Reactive astrocytes demonstrate stellate morphology, increased glial fibrillary acidic protein (GFAP) immunoreactivity, increased number of mitochondria as well as elevated enzymatic and non-enzymatic antioxidant activities. Astrocytes can re-uptake and metabolize glutamate and in this way they control its extracellular concentration. The ability of astrocytes to protect neurons against the toxic action of free radicals depends on their specific energy metabolism, high glutathione level, increased antioxidant enzyme activity (catalase, superoxide dismutase, glutathione peroxidase) and overexpression of antiapoptotic bcl-2 gene. Astrocytes produce cytokines (TNF-alpha, IL-1, IL-6) involved in the initiation and maintaining of immunological response in the CNS. In astrocytes, like in neurones, ischemia induces the expression of immediate early genes: c-fos, c-jun, fos B, jun B, jun D, Krox-24, NGFI-B and others. The protein products of these genes modulate the expression of different proteins, both destructive ones and those involved in the neuroprotective processes.
Collapse
Affiliation(s)
- B Gabryel
- Department of Pharmacology, Silesian Medical University, Medyków 18 St., 40-752 Katowice, Poland.
| | | |
Collapse
|
40
|
Frohman EM, Monson NL, Lovett-Racke AE, Racke MK. Autonomic regulation of neuroimmunological responses: implications for multiple sclerosis. J Clin Immunol 2001; 21:61-73. [PMID: 11332655 DOI: 10.1023/a:1011016124524] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The expression of neural regulatory molecules by immune cells that infiltrate the nervous system upon injury may be a mechanism for cross regulation between the nervous system and the immune system. Several lines of evidence implicate nerve growth factor signaling through its receptors as a potential source of communication between the two systems. The expression of beta-adrenergic receptors and sympathetic innervation of lymphoid organs represents another example of communication between the immune and the nervous system. In this review, we discuss mechanisms of how factors in common between the nervous system and the immune system may result in regulatory circuits which are important in both healthy and diseased states. These studies may have relevance for a number of inflammatory conditions in humans, including multiple sclerosis.
Collapse
Affiliation(s)
- E M Frohman
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, 75235, USA.
| | | | | | | |
Collapse
|
41
|
Lau LT, Yu AC. Astrocytes produce and release interleukin-1, interleukin-6, tumor necrosis factor alpha and interferon-gamma following traumatic and metabolic injury. J Neurotrauma 2001; 18:351-9. [PMID: 11284554 DOI: 10.1089/08977150151071035] [Citation(s) in RCA: 239] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The brain is no longer considered immune-privileged due to its capability of producing cytokines in response to neurotrauma; however, the cellular sources of cytokines have not been defined. This study focused on the production of four inflammatory cytokines, interleukin-1 (IL-1alpha), interleukin-6 (IL-6), tumor necrosis factor alpha (TNFalpha), and interferon gamma (IFN-gamma) in primary culture of astrocytes under two different injury models which simulated in vivo mechanical trauma (scratch injury) and ischemia. Results demonstrated that astrocytes after scratch injury were positively immunostained with IL-1alpha, IL-6, and TNFalpha. A slot-blot study of culture media showed that the release of IL-1alpha, IL-6, TNFalpha, and IFN-gamma by astrocytes subsequent to scratch and ischemic injury reached approximately twice the control values. The temporal expression of these cytokines was different for the two models. All four cytokines began to increase 1 h postscratch and remained at high levels throughout the experiment. In the ischemic model, however, the increase of cytokine expression was delayed until 4-8 h of ischemia, when sharp increases were seen in all four cytokines. In this culture system, the exogenous influence of blood-borne factors and leukocytes, which occur with in vivo trauma and ischemia, was eliminated. Accordingly, the cytokines detected in the culture media were derived from astrocytes. This study provides the first evidence that astrocytes, without the influence from other cell types, can produce and release cytokines following mechanical and ischemic injury.
Collapse
Affiliation(s)
- L T Lau
- Department of Biology, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, China
| | | |
Collapse
|
42
|
Clark WM, Lutsep HL. Potential of anticytokine therapies in central nervous system ischaemia. Expert Opin Biol Ther 2001; 1:227-37. [PMID: 11727532 DOI: 10.1517/14712598.1.2.227] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Central nervous system (CNS) ischaemia is associated with an acute inflammatory response which appears to potentiate CNS injury, especially following reperfusion. This response includes the release of inflammatory mediators called cytokines including IL-1 and TNF-alpha, which triggers the production of additional cytokines including IL-6 and activates leukocytes which infiltrate into the CNS. Increased expression of cytokines has been demonstrated to occur in the first few hours after CNS ischaemia. Preliminary clinical studies suggest that plasma levels of IL-6 are correlated with functional recovery while brain levels of cytokines have been demonstrated to increase following experimental ischaemia. Although there are no current clinical 'anti-cytokine' treatment studies for stroke, experimental studies modulating IL-1 and TNF-alpha have shown neuroprotection.
Collapse
Affiliation(s)
- W M Clark
- Oregon Stroke Center, Dept. of Neurology UHS44, Oregon Health Sciences University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97201, USA.
| | | |
Collapse
|
43
|
McLaren FH, Svendsen CN, Van der Meide P, Joly E. Analysis of neural stem cells by flow cytometry: cellular differentiation modifies patterns of MHC expression. J Neuroimmunol 2001; 112:35-46. [PMID: 11108931 DOI: 10.1016/s0165-5728(00)00410-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neural stem cells are currently considered very hopeful candidates for cell replacement therapy in neurodegenerative pathologies such as Parkinson's disease. Here we show that different cell types derived from neurospheres amplified in vitro can be identified by FACS analysis relying solely on physical parameters (FSC/SSC) or autofluorescence. Additionally, after treatment with a panel of inflammatory cytokines, neurospheres and their differentiated progeny were shown to express MHC antigens which could potentially cause transplant rejection. Astrocytes expressed the highest levels of MHC. Hence removing such cells prior to transplantation could potentially optimise transplant survival.
Collapse
Affiliation(s)
- F H McLaren
- Laboratory of Functional Immunogenetics, Molecular Immunology Programme, Babraham Institute, Babraham, CB2 4AT, Cambridge, UK
| | | | | | | |
Collapse
|
44
|
Harry GJ, Lefebvre d'Hellencourt C, Bruccoleri A, Schmechel D. Age-dependent cytokine responses: trimethyltin hippocampal injury in wild-type, APOE knockout, and APOE4 mice. Brain Behav Immun 2000; 14:288-304. [PMID: 11120597 DOI: 10.1006/brbi.2000.0606] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this study, the hippocampal neurotoxicant trimethyltin (TMT) was used to examine possible differential susceptibility associated with the apolipoprotein E genotype. Mice-wild type (C57BL6J), APOE knockout, and APOE4 transgenic-received either saline or TMT (2 mg/kg, ip) at either 21 days or 8 months of age. At both ages, similar mRNA levels were seen in the hippocampus across genotypes for ICAM-1, A20, and MAC-1. GFAP mRNA was higher in the APOE knockouts and APOE4 as compared to wild-type mice. Within 24 h, TMT produced cell death of hippocampal dentate granule neurons and mild astrogliosis in all animals. In 21-day-old mice, TMT exposure significantly increased mRNA levels for ICAM-1 and MIP-1alpha in all genotypes. EB-22, GFAP, TNFalpha, and TGF-beta1 levels were significantly elevated in both wild-type and APOE knockout mice following TMT. At 8 months of age, genotype specific differences were observed. mRNA levels for GFAP, TNFbeta, TNFalpha, and MIP-1alpha were increased in both APOE knockout and APOE4 mice compared to wild-type mice. TMT exposure significantly increased mRNA levels for GFAP and MIP-1alpha in all animals. TNFalpha mRNA levels were increased in wild-type and APOE4 mice while EB22 mRNA levels were increased in both the APOE knockout and APOE4 mice but not wild-type mice. These data suggest an age-dependent effect on both microglia early inflammatory responses to injury associated with the APOE genotype.
Collapse
Affiliation(s)
- G J Harry
- Neurotoxicology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | | | |
Collapse
|
45
|
Cornet A, Bettelli E, Oukka M, Cambouris C, Avellana-Adalid V, Kosmatopoulos K, Liblau RS. Role of astrocytes in antigen presentation and naive T-cell activation. J Neuroimmunol 2000; 106:69-77. [PMID: 10814784 DOI: 10.1016/s0165-5728(99)00215-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Astrocytes may have a role in antigen presentation in inflammatory diseases of the central nervous system (CNS) such as MS and EAE. In this study, we have assessed whether purified astrocyte cultures could stimulate naive CD4(+) or CD8(+) T-cells from TCR transgenic mice. As previously described, astrocytes sustained antigen-specific CD4(+) T-cell proliferation only in the presence of IFN-gamma, which promotes expression of both MHC class II and B7 molecules on astrocytes. In addition, we show that astrocytes also have the capacity to present antigens to naive CD8(+) T-cell and promote their proliferation. In one system, this CD8(+) T-cell proliferation was dependent on IFN-gamma-induced upregulation of B7 molecules on astrocytes. However, in a second TCR transgenic system, astrocytes could induce naive CD8(+) T-cell proliferation even in the absence of IFN-gamma. The possible implications of these findings for the pathophysiology of CNS inflammatory diseases are discussed.
Collapse
Affiliation(s)
- A Cornet
- INSERM U 267, Paul Brousse Hospital, 14 avenue Paul Vaillant-Couturier, 94807, Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
46
|
DeVito WJ, Stone S, Mori K, Shamgochian M. Ethanol inhibits prolactin- and tumor necrosis factor-?-, but not gamma interferon-induced expression of intercellular adhesion molecule-1 in human astrocytoma cells. J Cell Biochem 2000. [DOI: 10.1002/(sici)1097-4644(20000601)77:3<455::aid-jcb10>3.0.co;2-s] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
47
|
Balyasnikova IV, Pelligrino DA, Greenwood J, Adamson P, Dragon S, Raza H, Galea E. Cyclic adenosine monophosphate regulates the expression of the intercellular adhesion molecule and the inducible nitric oxide synthase in brain endothelial cells. J Cereb Blood Flow Metab 2000; 20:688-99. [PMID: 10779013 DOI: 10.1097/00004647-200004000-00006] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The authors studied whether cyclic AMP (cAMP), a widespread regulator of inflammation, modulates the cytokine-mediated expression of the intercellular adhesion molecule, intercellular adhesion molecule-1 (ICAM-1), and the inflammatory nitric oxide synthase 2 (NOS-2), in primary and immortalized brain endothelial cell cultures (GP8.3 cell line). When measured by enzyme-linked immunosorbent assay (ELISA), ICAM-1 was constitutively expressed and was up-regulated twofold by interleukin-1beta, with no effect of interferon-gamma. The NOS-2 activity, assessed by nitrite accumulation, was absent from untreated cultures but was induced by interleukin-1beta and interferon-gamma acting synergistically. Stimulation of cAMP-dependent pathways with forskolin or dibutyryl cAMP decreased ICAM-1 protein expression, whereas it increased NOS-2 protein expression. For both ICAM-1 and NOS-2, mRNA expression correlated with protein expression. Blockade of NOS activity with L-N-monomethylargiuine (L-NMMA) did not alter ICAM-1 expression, indicating that the nitric oxide released by NOS-2 did not cause the down-regulation of ICAM-1. Analysis of NFKB activation indicated that cAMP acted through a mechanism other than inhibition of nuclear translocation of NFKB. The authors conclude that cAMP modulates the expression of proinflammatory molecules in brain endothelium. This suggests that inflammatory processes at the blood-brain barrier in vivo may be regulated by perivascular neurotransmitters via cAMP.
Collapse
Affiliation(s)
- I V Balyasnikova
- Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Yuan L, Neufeld AH. Tumor necrosis factor-?: A potentially neurodestructive cytokine produced by glia in the human glaucomatous optic nerve head. Glia 2000. [DOI: 10.1002/1098-1136(200010)32:1<42::aid-glia40>3.0.co;2-3] [Citation(s) in RCA: 193] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
49
|
Abstract
Multiple sclerosis (MS) is best understood as an inflammatory disease of the central nervous system (CNS) white matter characterized by demyelination, focal T cell and macrophage infiltrates, axonal injury and loss of neurological function. Our current understanding invokes proinflammatory cells and mediators that may be triggered by environmental factors to mediate disease in a genetically susceptible host. Five major themes which have been associated with the pathogenesis of MS lesions will be discussed: (1) The differential activation states of myelin-reactive T cells from MS patients vs. normal individuals, (2) the selective expression of chemokines, adhesion molecules and matrix metalloproteinases, (3) the proposed roles of the B7 costimulatory pathway, (4) the proinflammatory cytokines and (5) the role of molecular mimicry.
Collapse
Affiliation(s)
- A Bar-Or
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115-5187, USA
| | | | | | | |
Collapse
|
50
|
Affiliation(s)
- J F Collawn
- Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, AL 35294-0005, USA
| | | |
Collapse
|