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
|
Bradford BM, Walmsley-Rowe L, Reynolds J, Verity N, Mabbott NA. Cell adhesion molecule CD44 is dispensable for reactive astrocyte activation during prion disease. Sci Rep 2024; 14:13749. [PMID: 38877012 PMCID: PMC11178777 DOI: 10.1038/s41598-024-63464-3] [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: 02/22/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024] Open
Abstract
Prion diseases are fatal, infectious, neurodegenerative disorders resulting from accumulation of misfolded cellular prion protein in the brain. Early pathological changes during CNS prion disease also include reactive astrocyte activation with increased CD44 expression, microgliosis, as well as loss of dendritic spines and synapses. CD44 is a multifunctional cell surface adhesion and signalling molecule which is considered to play roles in astrocyte morphology and the maintenance of dendritic spine integrity and synaptic plasticity. However, the role of CD44 in prion disease was unknown. Here we used mice deficient in CD44 to determine the role of CD44 during prion disease. We show that CD44-deficient mice displayed no difference in their response to CNS prion infection when compared to wild type mice. Furthermore, the reactive astrocyte activation and microgliosis that accompanies CNS prion infection was unimpaired in the absence of CD44. Together, our data show that although CD44 expression is upregulated in reactive astrocytes during CNS prion disease, it is dispensable for astrocyte and microglial activation and the development of prion neuropathogenesis.
Collapse
Affiliation(s)
- Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| | - Lauryn Walmsley-Rowe
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Joe Reynolds
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
- Maurice Wohl Basic and Clinical Neuroscience Institute, King's College London, Denmark Hill, London, SE5 9NU, UK
| | - Nicholas Verity
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| |
Collapse
|
3
|
Sulimai N, Lominadze D. Fibrinogen and Neuroinflammation During Traumatic Brain Injury. Mol Neurobiol 2020; 57:4692-4703. [PMID: 32776201 DOI: 10.1007/s12035-020-02012-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Many neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis, and traumatic brain injury (TBI) are associated with systemic inflammation. Inflammation itself results in increased blood content of fibrinogen (Fg), called hyperfibrinogenemia (HFg). Fg is not only considered an acute phase protein and a marker of inflammation, but has been shown that it can cause inflammatory responses. Fibrin deposits have been associated with memory reduction in neuroinflammatory diseases such as AD and TBI. Reduction in short-term memory has been seen during the most common form of TBI, mild-to-moderate TBI. Fibrin deposits have been found in brains of patients with mild-to-moderate TBI. The vast majority of the literature emphasizes the role of fibrin-activated microglia as the mediator in the neuroinflammation pathway. However, the recent discovery that astrocytes, which constitute approximately 30% of the cells in the mammalian central nervous system, manifest different reactive states warrants further investigations in the causative role of HFg in astrocyte-mediated neuroinflammation. Our previous study showed that Fg deposited in the vasculo-astrocyte interface-activated astrocytes. However, little is known of how Fg directly affects astrocytes and neurons. In this review, we summarize studies that show the effect of Fg on different types of cells in the vasculo-neuronal unit. We will also discuss the possible mechanism of HFg-induced neuroinflammation during TBI.
Collapse
Affiliation(s)
- Nurul Sulimai
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - David Lominadze
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
- Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
4
|
Williams JL, Manivasagam S, Smith BC, Sim J, Vollmer LL, Daniels BP, Russell JH, Klein RS. Astrocyte-T cell crosstalk regulates region-specific neuroinflammation. Glia 2020; 68:1361-1374. [PMID: 31961459 PMCID: PMC7317491 DOI: 10.1002/glia.23783] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/17/2019] [Accepted: 01/05/2020] [Indexed: 12/24/2022]
Abstract
During multiple sclerosis (MS), an inflammatory and neurodegenerative disease of the central nervous system (CNS), symptoms, and outcomes are determined by the location of inflammatory lesions. While we and others have shown that T cell cytokines differentially regulate leukocyte entry into perivascular spaces and regional parenchymal localization in murine models of MS, the molecular mechanisms of this latter process are poorly understood. Here, we demonstrate that astrocytes exhibit region-specific responses to T cell cytokines that promote hindbrain versus spinal cord neuroinflammation. Analysis of cytokine receptor expression in human astrocytes showed region-specific responsiveness to Th1 and Th17 inflammatory cytokines. Consistent with this, human and murine astrocytes treated with these cytokines exhibit differential expression of the T cell localizing molecules VCAM-1 and CXCR7 that is both cytokine and CNS region-specific. Using in vivo models of spinal cord versus brain stem trafficking of myelin-specific T cells and astrocyte-specific deletion strategies, we confirmed that Th1 and Th17 cytokines differentially regulate astrocyte expression of VCAM-1 and CXCR7 in these locations. Finally, stereotaxic injection of individual cytokines into the hindbrain or spinal cord revealed region- and cytokine-specific modulation of localizing cue expression by astrocytes. These findings identify a role for inflammatory cytokines in mediating local astrocyte-dependent mechanisms of immune cell trafficking within the CNS during neuroinflammation.
Collapse
Affiliation(s)
- Jessica L. Williams
- Department of Neurosciences, Lerner Research InstituteCleveland Clinic FoundationClevelandOhio
- Department of MedicineWashington University School of MedicineSt. LouisMissouri
| | - Sindhu Manivasagam
- Department of MedicineWashington University School of MedicineSt. LouisMissouri
| | - Brandon C. Smith
- Department of Neurosciences, Lerner Research InstituteCleveland Clinic FoundationClevelandOhio
| | - Julia Sim
- Department of Developmental BiologyWashington University School of MedicineSt. LouisMissouri
| | - Lauren L. Vollmer
- Department of MedicineWashington University School of MedicineSt. LouisMissouri
| | - Brian P. Daniels
- Department of MedicineWashington University School of MedicineSt. LouisMissouri
| | - John H. Russell
- Department of Developmental BiologyWashington University School of MedicineSt. LouisMissouri
| | - Robyn S. Klein
- Department of MedicineWashington University School of MedicineSt. LouisMissouri
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMissouri
- Department of Anatomy and NeurobiologyWashington University School of MedicineSt. LouisMissouri
| |
Collapse
|
5
|
Hlavac N, VandeVord PJ. Astrocyte Mechano-Activation by High-Rate Overpressure Involves Alterations in Structural and Junctional Proteins. Front Neurol 2019; 10:99. [PMID: 30853931 PMCID: PMC6395392 DOI: 10.3389/fneur.2019.00099] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/24/2019] [Indexed: 11/13/2022] Open
Abstract
Primary blast neurotrauma represents a unique injury paradigm characterized by high-rate overpressure effects on brain tissue. One major hallmark of blast neurotrauma is glial reactivity, notably prolonged astrocyte activation. This cellular response has been mainly defined in primary blast neurotrauma by increased intermediate filament expression. Because the intermediate filament networks physically interface with transmembrane proteins for junctional support, it was hypothesized that cell junction regulation is altered in the reactive phenotype as well. This would have implications for downstream transcriptional regulation via signal transduction pathways like nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Therefore, a custom high-rate overpressure simulator was built for in vitro testing using mechanical conditions based on intracranial pressure measurements in a rat model of blast neurotrauma. Primary rat astrocytes were exposed to isolated high-rate mechanical stimulation to study cell junction dynamics in relation to their mechano-activation. First, a time course for "classical" features of reactivity was devised by evaluation of glial fibrillary acidic protein (GFAP) and proliferating cell nuclear antigen (PCNA) expression. This was followed by gene and protein expression for both gap junction (connexins) and anchoring junction proteins (integrins and cadherins). Signal transduction analysis was carried out by nuclear localization of two molecules, NF-κB p65 and mitogen-activated protein kinase (MAPK) p38. Results indicated significant increases in connexin-43 expression and PCNA first at 24 h post-overpressure (p < 0.05), followed by structural reactivity (via increased GFAP, p < 0.05) corresponding to increased anchoring junction dynamics at 48 h post-overpressure (p < 0.05). Moreover, increased phosphorylation of focal adhesion kinase (FAK) was observed in addition to increased nuclear localization of both p65 and p38 (p < 0.05) during the period of structural reactivity. To evaluate the transcriptional activity of p65 in the nucleus, electrophoretic mobility shift assay was conducted for a binding site on the promoter region for intracellular adhesion molecule-1 (ICAM-1), an antagonist of tight junctions. A significant increase in the interaction of nuclear proteins with the NF-κB site on the ICAM-1 corresponded to increased gene and protein expression of ICAM-1 (p < 0.05). Altogether, these results indicate multiple targets and corresponding signaling pathways which involve cell junction dynamics in the mechano-activation of astrocytes following high-rate overpressure.
Collapse
Affiliation(s)
- Nora Hlavac
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute, Blacksburg, VA, United States
| | - Pamela J VandeVord
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute, Blacksburg, VA, United States.,Department of Research, Salem Veterans Affairs Medical Center, Salem, VA, United States
| |
Collapse
|
6
|
Astrup LB, Skovgaard K, Rasmussen RS, Iburg TM, Agerholm JS, Aalbæk B, Jensen HE, Nielsen OL, Johansen FF, Heegaard PMH, Leifsson PS. Staphylococcus aureus infected embolic stroke upregulates Orm1 and Cxcl2 in a rat model of septic stroke pathology. Neurol Res 2019; 41:399-412. [DOI: 10.1080/01616412.2019.1573455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Lærke Boye Astrup
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Kerstin Skovgaard
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Rune Skovgaard Rasmussen
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tine Moesgaard Iburg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Jørgen Steen Agerholm
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Bent Aalbæk
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ole Lerberg Nielsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Flemming Fryd Johansen
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Mikael Helweg Heegaard
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Páll Skúli Leifsson
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
7
|
Weinberg RP, Koledova VV, Schneider K, Sambandan TG, Grayson A, Zeidman G, Artamonova A, Sambanthamurthi R, Fairus S, Sinskey AJ, Rha C. Palm Fruit Bioactives modulate human astrocyte activity in vitro altering the cytokine secretome reducing levels of TNFα, RANTES and IP-10. Sci Rep 2018; 8:16423. [PMID: 30401897 PMCID: PMC6219577 DOI: 10.1038/s41598-018-34763-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease, are becoming more prevalent and an increasing burden on society. Neurodegenerative diseases often arise in the milieu of neuro-inflammation of the brain. Reactive astrocytes are key regulators in the development of neuro-inflammation. This study describes the effects of Palm Fruit Bioactives (PFB) on the behavior of human astrocytes which have been activated by IL-1β. When activated, the astrocytes proliferate, release numerous cytokines/chemokines including TNFα, RANTES (CCL5), IP-10 (CXCL10), generate reactive oxygen species (ROS), and express specific cell surface biomarkers such as the Intercellular Adhesion Molecule (ICAM), Vascular Cellular Adhesion Molecule (VCAM) and the Neuronal Cellular Adhesion Molecule (NCAM). Interleukin 1-beta (IL-1β) causes activation of human astrocytes with marked upregulation of pro-inflammatory genes. We show significant inhibition of these pro-inflammatory processes when IL-1β-activated astrocytes are exposed to PFB. PFB causes a dose-dependent and time-dependent reduction in specific cytokines: TNFα, RANTES, and IP-10. We also show that PFB significantly reduces ROS production by IL-1β-activated astrocytes. Furthermore, PFB also reduces the expression of ICAM and VCAM, both in activated and naïve human astrocytes in vitro. Since reactive astrocytes play an essential role in the neuroinflammatory state preceding neurodegenerative diseases, this study suggests that PFB may have a potential role in their prevention and/or treatment.
Collapse
Affiliation(s)
- Robert P Weinberg
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Vera V Koledova
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kirsten Schneider
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - T G Sambandan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Adlai Grayson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Gal Zeidman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Anastasia Artamonova
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ravigadevi Sambanthamurthi
- Advanced Biotechnology and Breeding Centre, Malaysian Palm Oil Board, 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia
| | - Syed Fairus
- Advanced Biotechnology and Breeding Centre, Malaysian Palm Oil Board, 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia
| | - Anthony J Sinskey
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - ChoKyun Rha
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
8
|
Lagos-Cabré R, Alvarez A, Kong M, Burgos-Bravo F, Cárdenas A, Rojas-Mancilla E, Pérez-Nuñez R, Herrera-Molina R, Rojas F, Schneider P, Herrera-Marschitz M, Quest AFG, van Zundert B, Leyton L. α Vβ 3 Integrin regulates astrocyte reactivity. J Neuroinflammation 2017; 14:194. [PMID: 28962574 PMCID: PMC5622429 DOI: 10.1186/s12974-017-0968-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022] Open
Abstract
Background Neuroinflammation involves cytokine release, astrocyte reactivity and migration. Neuronal Thy-1 promotes DITNC1 astrocyte migration by engaging αVβ3 Integrin and Syndecan-4. Primary astrocytes express low levels of these receptors and are unresponsive to Thy-1; thus, inflammation and astrocyte reactivity might be necessary for Thy-1-induced responses. Methods Wild-type rat astrocytes (TNF-activated) or from human SOD1G93A transgenic mice (a neurodegenerative disease model) were used to evaluate cell migration, Thy-1 receptor levels, signaling molecules, and reactivity markers. Results Thy-1 induced astrocyte migration only after TNF priming. Increased expression of αVβ3 Integrin, Syndecan-4, P2X7R, Pannexin-1, Connexin-43, GFAP, and iNOS were observed in TNF-treated astrocytes. Silencing of β3 Integrin prior to TNF treatment prevented Thy-1-induced migration, while β3 Integrin over-expression was sufficient to induce astrocyte reactivity and allow Thy-1-induced migration. Finally, hSOD1G93A astrocytes behave as TNF-treated astrocytes since they were reactive and responsive to Thy-1. Conclusions Therefore, inflammation induces expression of αVβ3 Integrin and other proteins, astrocyte reactivity, and Thy-1 responsiveness. Importantly, ectopic control of β3 Integrin levels modulates these responses regardless of inflammation. Electronic supplementary material The online version of this article (10.1186/s12974-017-0968-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raúl Lagos-Cabré
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Alvaro Alvarez
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Facultad de Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Milene Kong
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Department of Biomedicine, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Francesca Burgos-Bravo
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Areli Cárdenas
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Departamento de Ciencias Químicas y Biológicas, Universidad Bernardo O'Higgins, 837-0854, Santiago, Chile
| | - Edgardo Rojas-Mancilla
- Departamento de Ciencias Químicas y Biológicas, Universidad Bernardo O'Higgins, 837-0854, Santiago, Chile
| | - Ramón Pérez-Nuñez
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | | | - Fabiola Rojas
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland
| | - Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Andrew F G Quest
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Brigitte van Zundert
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile. .,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.
| |
Collapse
|
9
|
Liu Z, Qu Y, Wang J, Wu R. Selenium Deficiency Attenuates Chicken Duodenal Mucosal Immunity via Activation of the NF-κb Signaling Pathway. Biol Trace Elem Res 2016; 172:465-473. [PMID: 26728795 DOI: 10.1007/s12011-015-0589-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023]
Abstract
Selenium (Se) deficiency can cause intestinal mucosal inflammation, which is related to activation of nuclear transcription factor kappa-B (NF-κB) signaling pathway. However, the mechanism of inflammatory response in chicken duodenal mucosa caused by Se deficiency and its relationship with the NF-κB signaling pathway remain elusive. In this study, we firstly obtained Se-deficient chickens bred with 0.01 mg/kg Se and the normal chickens bred with 0.4 mg/kg Se for 35 days. Then, NF-κB signaling pathway, secretory immunoglobulin A (SIgA), inflammatory cytokines, oxidized glutathione, glutathione peroxidase, and glutathione activities were determined. The results showed that Se deficiency obviously enhanced p50, p65, and p65 DNA-binding activities. The phosphorylation of IκB-α and phosphorylation of kappa-B kinase subunit alpha (IKKα) and IKKα were elevated, but IκB-α was decreased (P < 0.05). Moreover, Se deficiency reduced SIgA amount in the duodenal mucosa but increased the level of interleukin-1β (IL-1β), IL-17A, tumor necrosis factor-α (TNF-α), and interferon gamma (IFN-γ). In contrast, anti-inflammatory cytokines, such as TGF-β1 and IL-10, were significantly suppressed. Additionally, Se deficiency increased oxidized glutathione activity, whereas decreased glutathione peroxidase and glutathione activities (P < 0.05), suggesting that Se deficiency affected the regulation function of redox. Taken together, our results demonstrated that Se deficiency attenuated chicken duodenal mucosal immunity via activation of NF-κB signaling pathway regulated by redox activity, which suggested that Se is a crucial host factor involved in regulating inflammation.
Collapse
Affiliation(s)
- Zhe Liu
- College of Life Sciences and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, People's Republic of China
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China
| | - Yanpeng Qu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China
| | - Rui Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, 2 Xinyang Road, Daqing, Heilongjiang, 163319, People's Republic of China.
| |
Collapse
|
10
|
Spanos JP, Hsu NJ, Jacobs M. Microglia are crucial regulators of neuro-immunity during central nervous system tuberculosis. Front Cell Neurosci 2015; 9:182. [PMID: 26041993 PMCID: PMC4435040 DOI: 10.3389/fncel.2015.00182] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/27/2015] [Indexed: 01/11/2023] Open
Abstract
Mycobacterium tuberculosis (M. tuberculosis) infection of the central nervous system (CNS) is the most devastating manifestation of tuberculosis (TB), with both high mortality and morbidity. Although research has been fueled by the potential therapeutic target microglia offer against neurodegenerative inflammation, their part in TB infection of the CNS has not been fully evaluated nor elucidated. Yet, as both the preferential targets of M. tuberculosis and the immune-effector cells of the CNS, microglia are likely to be key determinants of disease severity and clinical outcomes. Following pathogen recognition, bacilli are internalized and capable of replicating within microglia. Cellular activation ensues, utilizing signaling molecules that may be neurotoxic. Central to initiating, orchestrating and modulating the tuberculous immune response is microglial secretion of cytokines and chemokines. However, the neurological environment is unique in that inflammatory signals, which appear to be damaging in the periphery, could be beneficial by governing neuronal survival, regeneration and differentiation. Furthermore, microglia are important in the recruitment of peripheral immune cells and central to defining the pro-inflammatory milieu of which neurotoxicity may result from many of the participating local or recruited cell types. Microglia are capable of both presenting antigen to infiltrating CD4(+) T-lymphocytes and inducing their differentiation-a possible correlate of protection against M. tuberculosis infection. Clarifying the nature of the immune effector molecules secreted by microglia, and the means by which other CNS-specific cell types govern microglial activation or modulate their responses is critical if improved diagnostic and therapeutic strategies are to be attained. Therefore, this review evaluates the diverse roles microglia play in the neuro-immunity to M. tuberculosis infection of the CNS.
Collapse
Affiliation(s)
- Jonathan Paul Spanos
- Division of Immunology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town Cape Town, South Africa
| | - Nai-Jen Hsu
- Division of Immunology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town Cape Town, South Africa
| | - Muazzam Jacobs
- Division of Immunology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town Cape Town, South Africa ; National Health Laboratory Service Johannesburg, South Africa
| |
Collapse
|
11
|
Interaction of astrocytes and T cells in physiological and pathological conditions. Brain Res 2015; 1623:63-73. [PMID: 25813828 DOI: 10.1016/j.brainres.2015.03.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/24/2022]
Abstract
The central nervous system (CNS) has long been recognized as a site of 'immune privilege' because of the existence of the blood brain barrier (BBB) which presumably isolates CNS from the peripheral immunosurveillance. Different from the peripheral organs, CNS is unique in response to all forms of CNS injury and disease which is mainly mediated by resident microglia and astrocyte. There is increasing evidence that immune cells are not only involved in neuroinflammation process but also the maintenance of CNS homeostasis. T cells, an important immune cell population, are involved in the pathogenesis of some neurological diseases by inducing either innate or adaptive immune responses. Astrocytes, which are the most abundant cell type in the CNS, maintain the integrity of BBB and actively participate in the initiation and progression of neurological diseases. Surprisingly, how astrocytes and T cells interact and the consequences of their interaction are not clear. In this review we briefly summarized T cells diversity and astrocyte function. Then, we examined the evidence for the astrocytes and T cells interaction under physiological and pathological conditions including ischemic stroke, multiple sclerosis, viral infection, and Alzheimer's disease. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
|
12
|
Huber AK, Duncker PC, Irani DN. Immune responses to non-tumor antigens in the central nervous system. Front Oncol 2014; 4:328. [PMID: 25431758 PMCID: PMC4230036 DOI: 10.3389/fonc.2014.00328] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/30/2014] [Indexed: 12/16/2022] Open
Abstract
The central nervous system (CNS), once viewed as an immune-privileged site protected by the blood-brain barrier (BBB), is now known to be a dynamic immunological environment through which immune cells migrate to prevent and respond to events such as localized infection. During these responses, endogenous glial cells, including astrocytes and microglia, become highly reactive and may secrete inflammatory mediators that regulate BBB permeability and recruit additional circulating immune cells. Here, we discuss the various roles played by astrocytes, microglia, and infiltrating immune cells during host immunity to non-tumor antigens in the CNS, focusing first on bacterial and viral infections, and then turning to responses directed against self-antigens in the setting of CNS autoimmunity.
Collapse
Affiliation(s)
- Amanda K Huber
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - Patrick C Duncker
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - David N Irani
- Department of Neurology, University of Michigan Medical School , Ann Arbor, MI , USA
| |
Collapse
|
13
|
Garcia JA, Pino PA, Mizutani M, Cardona SM, Charo IF, Ransohoff RM, Forsthuber TG, Cardona AE. Regulation of adaptive immunity by the fractalkine receptor during autoimmune inflammation. THE JOURNAL OF IMMUNOLOGY 2013; 191:1063-72. [PMID: 23817416 DOI: 10.4049/jimmunol.1300040] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fractalkine, a chemokine anchored to neurons or peripheral endothelial cells, serves as an adhesion molecule or as a soluble chemoattractant. Fractalkine binds CX3CR1 on microglia and circulating monocytes, dendritic cells, and NK cells. The aim of this study is to determine the role of CX3CR1 in the trafficking and function of myeloid cells to the CNS during experimental autoimmune encephalomyelitis (EAE). Our results show that, in models of active EAE, Cx3cr1(-/-) mice exhibited more severe neurologic deficiencies. Bone marrow chimeric mice confirmed that CX3CR1 deficiency in bone marrow enhanced EAE severity. Notably, CX3CR1 deficiency was associated with an increased accumulation of CD115(+)Ly6C(-)CD11c(+) dendritic cells into EAE-affected brains that correlated with enhanced demyelination and neuronal damage. Furthermore, higher IFN-γ and IL-17 levels were detected in cerebellar and spinal cord tissues of CX3CR1-deficient mice. Analyses of peripheral responses during disease initiation revealed a higher frequency of IFN-γ- and IL-17-producing T cells in lymphoid tissues of CX3CR1-deficient as well as enhanced T cell proliferation induced by CX3CR1-deficient dendritic cells. In addition, adoptive transfer of myelin oligodendrocyte glycoprotein35-55-reactive wild-type T cells induced substantially more severe EAE in CX3CR1-deficient recipients when compared with wild-type recipients. Collectively, the data demonstrate that besides its role in chemoattraction, CX3CR1 is a key regulator of myeloid cell activation contributing to the establishment of adaptive immune responses.
Collapse
Affiliation(s)
- Jenny A Garcia
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Lam RA, Chwee JY, Le Bert N, Sauer M, Pogge von Strandmann E, Gasser S. Regulation of self-ligands for activating natural killer cell receptors. Ann Med 2013; 45:384-94. [PMID: 23701136 DOI: 10.3109/07853890.2013.792495] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are able to lyse infected and tumor cells while sparing healthy cells. Recognition of diseased cells by NK cells is governed by several activating and inhibitory receptors. We review numerous pathways that have been implicated in the regulation of self-ligands for activating receptors, including NKG2D, DNAM-1, LFA-1, NKp30, NKp44, NKp46, NKp65, and NKp80 found on NK cells and some T cells. Understanding how the regulation of self-encoded ligand expression is regulated may provide novel avenues for future therapeutic approaches to infections and cancer.
Collapse
Affiliation(s)
- Runyi A Lam
- Immunology Programme, Centre for Life Sciences, Department of Microbiology, National University of Singapore 117456, Singapore
| | | | | | | | | | | |
Collapse
|
15
|
Oh DY, Cui P, Hosseini H, Mosse J, Toh BH, Chan J. Potently immunosuppressive 5-fluorouracil-resistant mesenchymal stromal cells completely remit an experimental autoimmune disease. THE JOURNAL OF IMMUNOLOGY 2012; 188:2207-17. [PMID: 22291191 DOI: 10.4049/jimmunol.1101040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We treated mice with 5-fluorouracil (5-FU) to isolate a quiescent and undifferentiated mesenchymal stromal cell (MSC) population from the bone marrow. We examined these 5-FU-resistant MSCs (5-FU-MSCs) free from hematopoietic components for CFU fibroblasts (CFU-Fs) and assessed their immunosuppressive potential in vitro and in vivo. We differentiated fibroblastic CFU-Fs (Fibro-CFU-Fs) from mixed CFU-Fs, based on the absence of in situ expression of CD11b and CD45 hematopoietic markers, as well as on their differentiation capacity. Fibro-CFU-Fs were associated with increased numbers of large-sized Fibro-CFU-Fs (≥9 mm(2)) that displayed enhanced capacity for differentiation into adipogenic and osteogenic mesenchymal lineages. Administration of these 5-FU-resistant CD11b(-)CD45(-) MSCs 6 d after myelin oligodendrocyte glycoprotein (MOG) immunization completely remitted MOG-induced experimental autoimmune encephalomyelitis after initial development of mild disease. The remission was accompanied by reduced CNS cellular infiltration and demyelination, as well as a significant reduction in anti-MOG Ab and splenocyte proliferation to MOG. MOG-stimulated splenocytes from these mice showed elevated levels of Th2 cytokines (IL-4, IL-5, and IL-6) and decreased IL-17. Compared with untreated MSCs, 5-FU-MSCs demonstrated potent immunosuppression of Con A-stimulated splenocytes in vitro, even at a 1:320 MSC/splenocyte ratio. Immunosuppression was accompanied by elevated IL-1ra, IL-10, and PGE(2). Blocking IL-1ra, IL-10, and PGE(2), but not IL-6, heme oxygenase-1, and NO, attenuated 5-FU-MSC-induced immunosuppression. Together, our findings suggested that immunosuppression by 5-FU-MSC is mediated by a combination of elevated IL-1ra, IL-10, and PGE(2), anti-inflammatory Th2 cytokines, and decreased IL-17. Our findings suggested that 5-FU treatment identifies a population of potently immunosuppressive 5-FU-MSCs that have the potential to be exploited to remit autoimmune diseases.
Collapse
Affiliation(s)
- Ding Yuan Oh
- Centre for Inflammatory Disease, Monash University, Melbourne, Victoria 3163, Australia
| | | | | | | | | | | |
Collapse
|
16
|
Watari K, Nakamura M, Fukunaga Y, Furuno A, Shibata T, Kawahara A, Hosoi F, Kuwano T, Kuwano M, Ono M. The antitumor effect of a novel angiogenesis inhibitor (an octahydronaphthalene derivative) targeting both VEGF receptor and NF-κB pathway. Int J Cancer 2011; 131:310-21. [PMID: 21826646 DOI: 10.1002/ijc.26356] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 07/27/2011] [Indexed: 01/14/2023]
Abstract
Development of a novel type of angiogenesis inhibitor will be essential for further improvement of therapeutics against cancer patients. We examined whether an octahydronaphthalene derivative, AMF-26, which was screened as an inhibitor of intercellular adhesion molecule-1 (ICAM-1) production stimulated by inflammatory stimuli in vascular endothelial cells, could block angiogenesis in response to vascular endothelial growth factor (VEGF) and/or inflammatory cytokines. Low dose AMF-26 effectively inhibited the tumor necrosis factor-α (TNF-α)- or the interleukin-1β (IL-1β)-induced production of ICAM-1 in human umbilical vascular endothelial cells (HUVECs). We found that the TNF-α-induced phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) and nuclear translocation of p65 were impaired by AMF-26 in both endothelial cells and cancer cells. AMF-26 was found to inhibit the phosphorylation of VEGF receptor 1 (VEGFR1), VEGFR2 and the downstream signaling molecules Akt, extracellular signal-regulated kinase (ERK)1/2 stimulated by VEGF in HUVECs. Therefore, the VEGF-induced proliferation, migration and tube formation of vascular endothelial cells was highly susceptible to inhibition by AMF-26. Oral administration of AMF-26 significantly blocked VEGF- or IL-1β-induced angiogenesis in the mouse cornea, and also tumor angiogenesis and growth. Together, our results indicate that AMF-26 inhibits angiogenesis through suppression of both VEGFR1/2 and nuclear factor-κB (NF-κB) signaling pathways when stimulated by VEGF or inflammatory cytokines. AMF-26 could be a promising novel candidate drug for cancer treatments.
Collapse
Affiliation(s)
- Kosuke Watari
- Department of Pharmaceutical Oncology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Jaal J, Brüchner K, Hoinkis C, Dörr W. Radiation‐induced variations in urothelial expression of intercellular adhesion molecule 1 (ICAM‐1): association with changes in urinary bladder function. Int J Radiat Biol 2009; 80:65-72. [PMID: 14761851 DOI: 10.1080/09553000310001632921] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE To assess the effect of single-dose irradiation on intercellular adhesion molecule 1 (ICAM-1) expression in the urothelium of mouse urinary bladder and to correlate ICAM-1 variations with fluctuations in storage capacity during the early and late radiation response. MATERIALS AND METHODS Groups of female C3H mice were subjected to irradiation with either 20 or 0 Gy. The intensity of immunohistochemical ICAM-1 staining in the urothelium was assessed in a semiquantitative way applying an arbitrary score (0-5). Changes in bladder storage function were assessed by transurethral cystometry. RESULTS For the early radiation response phase, a reduction in bladder capacity by >50%, i.e. a positive functional radiation response, was seen in 40% of the irradiated animals between days 0 and 15, and in 64% of animals during days 16-30. During the late response phase, 71% of the animals sacrificed after day 180 developed a positive functional response. Urothelial cells were found to express ICAM-1 constitutively. Irradiation resulted in an early rise in staining signal by day 2, with a maximum on day 4 and a return to control values on day 13. A permanent increase in ICAM-1 staining signal was observed in the late phase, from day 90 to 360 after irradiation. The expression of ICAM-1 in animals with a positive late response was 4.2+/-1.2 (mean+/-standard deviation), compared with 2.6+/-1.0 in non-responders (p=0.0009). CONCLUSION Irradiation induces significant acute and chronic changes in urothelial ICAM-1 expression indicating that the urothelium contributes to the pathogenesis of both acute and late radiation effects in the urinary bladder.
Collapse
Affiliation(s)
- J Jaal
- Klinik und Poliklinik für Strahlentherapie und Radioonkologie, Medizinische Fakultät Carl Gustav Carus der Technischen Universität, D-01307 Dresden, Germany.
| | | | | | | |
Collapse
|
18
|
Decker Y, McBean G, Godson C. Lipoxin A4 inhibits IL-1beta-induced IL-8 and ICAM-1 expression in 1321N1 human astrocytoma cells. Am J Physiol Cell Physiol 2009; 296:C1420-7. [PMID: 19357230 DOI: 10.1152/ajpcell.00380.2008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is a growing appreciation that endogenously produced mediators may actively promote the resolution of inflammation. Lipoxins (LX) are a group of recently discovered lipid mediators that have been shown to exert anti-inflammatory and proresolution effects on cells of myeloid and nonmyeloid origin. LXs mediate a number of processes, including regression of pro-inflammatory cytokine production, inhibition of cell proliferation, and stimulation of phagocytosis of apoptotic leukocytes by macrophages. Lipoxin A(4) (LXA(4)) is one of the principal LXs formed by mammalian cells. Recently, a G protein-coupled receptor that binds LXA(4,) the lipoxin A(4) receptor, was identified in astrocytes and microglia, suggesting that these cells may be a target for LX action in the brain. In this study, we have investigated the potential of LXA(4) to modify inflammatory responses of astrocytes, using the 1321N1 human astrocytoma cell line as a model system. As shown by quantitative RT-PCR, LXA(4) (10 nM) significantly inhibited (P < 0.05) the IL-1beta-induced stimulation of IL-8 and ICAM-1 expression in these cells. Furthermore, LXA(4) (10 nM) decreased the expression of IL-1beta-induced IL-8 protein levels (P < 0.05). LXA(4) (10 nM) was found to inhibit IL-1beta-induced degradation of IkappaBalpha (P < 0.05), and the activation of an NFkappaB regulated reporter gene construct (P < 0.05). Overall, these data suggest that LXA(4) exerts anti-inflammatory effects in 1321N1 astrocytoma cells at least in part via an NFkappaB-dependent mechanism. It is concluded that LXA(4) may represent a potentially novel therapeutic approach to acute or chronic inflammation in the brain.
Collapse
Affiliation(s)
- Yann Decker
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | |
Collapse
|
19
|
Shen A, Yang J, Gu Y, Zhou D, Sun L, Qin Y, Chen J, Wang P, Xiao F, Zhang L, Cheng C. Lipopolysaccharide-evoked activation of p38 and JNK leads to an increase in ICAM-1 expression in Schwann cells of sciatic nerves. FEBS J 2008; 275:4343-53. [DOI: 10.1111/j.1742-4658.2008.06577.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
20
|
Rodriguez S, Kunde YA, McCleskey TM, Hong-Geller E. Upregulation of I-CAM1 in response to beryllium exposure in small airway epithelial cells. Toxicol Lett 2008; 179:140-7. [DOI: 10.1016/j.toxlet.2008.04.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2008] [Revised: 04/28/2008] [Accepted: 04/28/2008] [Indexed: 01/10/2023]
|
21
|
Sriram K, O'Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol 2007; 2:140-53. [PMID: 18040839 DOI: 10.1007/s11481-007-9070-6] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 03/01/2007] [Indexed: 01/05/2023]
Abstract
Proinflammatory cytokines and chemokines have been implicated in the pathogenesis of several neurological and neurodegenerative disorders. Prominent among such factors is the pleiotropic cytokine, tumor necrosis factor (TNF)-alpha. Under normal physiological conditions, TNF-alpha orchestrates a diverse array of functions involved in immune surveillance and defense, cellular homeostasis, and protection against certain neurological insults. However, paradoxical effects of this cytokine have been observed. TNF-alpha is elicited in the brain following injury (ischemia, trauma), infection (HIV, meningitis), neurodegeneration (Alzheimer's, Parkinson's), and chemically induced neurotoxicity. The multifarious identity for this cytokine appears to be influenced by several mechanisms. Among the most prominent are the regulation of TNFalpha-induced NF-kappaB activation by adapter proteins such as TRADD and TRAF, and second, the heterogeneity of microglia and their distribution pattern across brain regions. Here, we review the differential role of TNF-alpha in response to brain injury, with emphasis on neurodegeneration, and discuss the possible mechanisms for such diverse and region-specific effects.
Collapse
Affiliation(s)
- Krishnan Sriram
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, CDC-NIOSH, 1095 Willowdale Road, Morgantown, WV 26505, USA
| | | |
Collapse
|
22
|
Chen JT, Chen CH, Horng CT, Chien MW, Lu DW, Liang JB, Tai MC, Chang YH, Chen PL, Chen YH. Glucosamine sulfate inhibits proinflammatory cytokine-induced icam-1 production in human conjunctival cells in vitro. J Ocul Pharmacol Ther 2007; 22:402-16. [PMID: 17238806 DOI: 10.1089/jop.2006.22.402] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE We investigated whether glucosamine sulfate modulates the production of ICAM-1 induced by proinflammatory cytokines and whether glucosamine sulfate inhibits leukocyte adhesion to a monolayer of human conjunctival epithelial cells stimulated with proinflammatory cytokines. METHODS We used flow cytometry and either primary cultured human conjunctival cells or the Chang conjunctival cell model to determine the effects of glucosamine sulfate on the production of ICAM-1 in response to tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma, interleukin (IL)-1beta, IL-6, TNF-alpha plus IFN-gamma, or TNF-alpha plus IL-1beta. The effects of glucosamine sulfate on the expression of the ICAM-1 gene, upregulated by various cytokines, were determined by semiquantitative reverse transcription-polymerase chain reaction. The activation and nuclear translocation of the nuclear factors NF-kappaB and STAT1 were evaluated by the transient transfection of reporter gene systems and immunocytochemistry. The influence of glucosamine-sulfate-modulated ICAM-1 on neutrophil adhesion was demonstrated in a model that measures the adherence of conjunctival cells and neutrophils. RESULTS TNF-alpha, IFN-gamma, and IL-1beta significantly increased the production of ICAM-1 by both primary cultured human conjunctival cells and Chang conjunctival cells. Glucosamine sulfate effectively downregulated the production of ICAM-1 induced by TNF-alpha, IFN-gamma, IL-1beta, TNF-alpha plus IFN-gamma, or TNF-alpha plus IL-1beta. This downregulation occurred through the interferon-stimulated response element, IFN-gamma activation sequence, and binding sequence of NF-kappaB at the mRNA and protein levels. Glucosamine sulfate further inhibited the nuclear translocation of p65 protein in TNF-alpha- and IL-1beta-stimulated Chang conjunctival cells and phosphorylated STAT1 in IFN-gamma-stimulated Chang conjunctival cells. Glucosamine sulfate also significantly reduced the number of neutrophils adhering to a conjunctival monolayer in response to TNF-alpha, IFN-gamma, or IL-1beta. CONCLUSIONS Our results suggest that glucosamine sulfate inhibits ICAM-1 production in conjunctival epithelial cells in vitro. Therefore, glucosamine sulfate might be valuable in the treatment of inflammatory ocular-surface conditions.
Collapse
Affiliation(s)
- Jiann-Torng Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
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: 3.2] [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
|
24
|
Zhu YN, Zhong XG, Feng JQ, Yang YF, Fu YF, Ni J, Liu QF, Tang W, Zhao WM, Zuo JP. Periplocoside E inhibits experimental allergic encephalomyelitis by suppressing interleukin 12-dependent CCR5 expression and interferon-gamma-dependent CXCR3 expression in T lymphocytes. J Pharmacol Exp Ther 2006; 318:1153-62. [PMID: 16751252 DOI: 10.1124/jpet.106.105445] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Periplocoside E (PSE) was found to inhibit primary T-cell activation in our previous study. Now we examined the effect and mechanisms of PSE on the central nervous system (CNS) demyelination in experimental allergic encephalomyelitis (EAE). C57BL/6 mice immunized with myelin oligodendrocyte glyco-protein (MOG) were treated with PSE following immunization and continued throughout the study. The effect on the progression of EAE and other relevant parameters were assessed. PSE reduced the incidence and severity of EAE. Spinal cord histopathology analysis showed that the therapeutic effect of PSE was associated with reduced mononuclear cell infiltration and CNS inflammation. As reverse transcription-polymerase chain reaction analysis showed, PSE decreased the CD4(+), CD8(+), and CD11b(+) cell infiltration. T cells from lymph nodes of MOG-immunized mice expressed enhanced levels of CCR5 and CXCR3 mRNA compared with T cells from normal mice. However, CCR5 and CXCR3 expressions were suppressed in T cells from PSE-treated mice. In vitro study also showed PSE inhibited interferon (IFN)-gamma-dependent CXCR3 expression in T cells through suppressing T-cell receptor (TCR) ligation-induced IFN-gamma production, whereas it inhibited interleukin (IL)-12-dependent CCR5 expression through suppressing IL-12 reactivity in TCR-triggered T cells. As a result, the initial influx of T cells into CNS was inhibited in PSE-treated mice. The consequent activation of macrophages/microglia cells was inhibited in spinal cord from PSE-treated mice as determination of chemokine expressions (CCL2, CCL3, CCL4, CCL5, CXCL9, and CXCL10). Consistently, the secondary influx of CD4(+), CD8(+), and CD11b(+) cells was decreased in spinal cords from PSE-treated mice. These findings suggest the potential therapeutic effect of PSE on multiple sclerosis.
Collapse
Affiliation(s)
- Yi-Na Zhu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Moynagh PN. The interleukin-1 signalling pathway in astrocytes: a key contributor to inflammation in the brain. J Anat 2006; 207:265-9. [PMID: 16185251 PMCID: PMC1571539 DOI: 10.1111/j.1469-7580.2005.00445.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A dysregulated inflammatory response in the central nervous system (CNS) lies at the heart of many neuropathological conditions such as multiple sclerosis and Alzheimer's disease. A key component of these inflammatory conditions is the accumulation of leukocytes in the CNS. The infiltration of leukocytes into the brain is dependent on the induction of leukocyte adhesion molecules and chemoattractant chemokines. Recent studies have suggested the astrocyte to be a key cell in mediating the inflammatory process in the brain and in expressing adhesion molecules and chemokines. Here I overview work in my laboratory and others that demonstrates interleukin-1 (IL-1) to be a key inducer of the expression of these molecules in astrocytes. The temporal expression is sustained in nature and this is due to prolonged activation of the transcription factor NFkappaB. The molecular basis to the sustained activation of NFkappaB is also discussed. The IL-1 signalling pathway thus emerges as a valuable therapeutic target in the treatment of presently incurable neuropathological conditions.
Collapse
Affiliation(s)
- Paul N Moynagh
- Department of Pharmacology, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland.
| |
Collapse
|
26
|
Lucchi NW, Moore JM. LPS induces secretion of chemokines by human syncytiotrophoblast cells in a MAPK-dependent manner. J Reprod Immunol 2006; 73:20-7. [PMID: 16870263 DOI: 10.1016/j.jri.2006.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Revised: 05/10/2006] [Accepted: 05/25/2006] [Indexed: 10/24/2022]
Abstract
The maintenance of pregnancy depends on the nature and magnitude of the immune responses induced within the placenta. An elevated proinflammatory response in the intervillous space (IVS) is associated with adverse pregnancy outcomes. It is becoming more apparent that the syncytiotrophoblast (ST) cells, which are in direct contact with maternal blood, are capable of contributing to the local immune environment in response to maternal hematogenous infections or exposure to proinflammatory stimuli. In this study, we investigated mechanisms by which ST might recruit maternal immune effectors to the IVS in response to bacterial infections. To assess this, primary trophoblasts were isolated from fresh term placentas and stimulated with lipopolysaccharide (LPS). LPS induced time-dependent expression and secretion of IL-8, macrophage inflammatory protein (MIP)-1alpha and MIP-1beta from ST cells and an upregulation of ICAM-1. The stimulation also resulted in the activation of ERK1/2 mitogen-activated protein kinase (MAPK) but not p38 or JNK1/2. Inhibition of ERK1/2 lead to a reduction in the secretion of MIP-1beta and IL-8 suggesting that their production is at least partly dependent on ERK1/2 activation. Results from this study reveal a potential mechanism by which differentiated ST cells modulate the local maternal immune responses during an intrauterine bacterial infection. Such responses could contribute to the clearance of the infection but also pathological features observed in intrauterine infections of the placenta.
Collapse
Affiliation(s)
- Naomi W Lucchi
- Department of Infectious Diseases, College of Veterinary Medicine and Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr. Athens, GA, USA
| | | |
Collapse
|
27
|
Müller K, Köhn FM, Port M, Abend M, Molls M, Ring J, Meineke V. Intercellular adhesion molecule-1: a consistent inflammatory marker of the cutaneous radiation reaction both in vitro
and in vivo. Br J Dermatol 2006; 155:670-9. [PMID: 16965414 DOI: 10.1111/j.1365-2133.2006.07407.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Radiation damage to skin is a key diagnostic and prognostic parameter for patients accidentally exposed to radiation. Moreover, skin is a target organ for crucial side-effects of routine radiotherapy. The pathophysiology of the cutaneous radiation reaction is in many respects still unknown. The acute inflammatory radiation reaction of skin has been shown to involve alterations in cell-cell and cell-matrix interactions, which are mediated by cellular adhesion molecules. OBJECTIVES To evaluate the effect of ionizing radiation on intercellular adhesion molecule-1 (ICAM-1) expression in human skin cells. METHODS Dermal monolayer cells, a three-dimensional skin model and skin biopsies were investigated for ICAM-1 expression after ionizing radiation using flow cytometry, quantitative reverse transcription-polymerase chain reaction and immunohistochemistry. ICAM-1 expression in monolayer cells pretreated with protein kinase inhibitors and dexamethasone prior to irradiation was analysed by flow cytometry. RESULTS Using different sources of skin cells, we demonstrated a consistent upregulation of both ICAM-1 mRNA and protein expression by ionizing radiation. Blocking experiments revealed that tumour necrosis factor-alpha, another ICAM-1 inducer, does not account for the effect of radiation. Radiation-induced upregulation of ICAM-1 expression was significantly attenuated by inhibitors to protein kinase C, mitogen-activated protein (MAP) ERK kinase, p38 MAP kinase and phosphatidylinositol 3-kinase. The anti-inflammatory agent dexamethasone suppressed the effect of radiation on ICAM-1 expression, suggesting its usefulness to treat the cutaneous radiation reaction. CONCLUSIONS Our data suggest that ICAM-1 is a consistent inflammatory parameter of the cutaneous radiation reaction both in vitro and in vivo that might provide new therapeutic options for diagnosis and treatment of effects of radiation.
Collapse
Affiliation(s)
- K Müller
- Bundeswehr Institute of Radiobiology, Neuherbergstr. 11, D-80937 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
28
|
Tometten M, Blois S, Kuhlmei A, Stretz A, Klapp BF, Arck PC. Nerve Growth Factor Translates Stress Response and Subsequent Murine Abortion via Adhesion Molecule-Dependent Pathways1. Biol Reprod 2006; 74:674-83. [PMID: 16371592 DOI: 10.1095/biolreprod.105.044651] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Spontaneous abortion is a frequent threat affecting 10%-25% of human pregnancies. Psychosocial stress has been suggested to be attributable for pregnancy losses by challenging the equilibrium of systems mandatory for pregnancy maintenance, including the nervous, endocrine, and immune system. Strong evidence indicates that stress-triggered abortion is mediated by adhesion molecules, i.e., intercellular adhesion molecule 1 (ICAM1) and leukocyte function associated molecule 1, now being referred to as integrin alpha L (ITGAL), which facilitate recruitment of inflammatory cells to the feto-maternal interface. The neurotrophin beta-nerve growth factor (NGFB), which has been shown to be upregulated in response to stress in multiple experimental settings including in the uterine lining (decidua) during pregnancy, increases ICAM1 expression on endothelial cells. Here, we investigated whether and how NGFB neutralization has a preventive effect on stress-triggered abortion in the murine CBA/J x DBA/2J model. We provide experimental evidence that stress exposure upregulates the frequency of abortion and the expression of uterine NGFB. Further, adhesion molecules ICAM1 and selectin platelet (SELP, formerly P-Selectin) and their ligands ITGAL and SELP ligand (SELPL, formerly P selectin glycoprotein ligand 1) respectively increase in murine deciduas in response to stress. Subsequently, decidual cytokines are biased toward a proinflammatory and abortogenic cytokine profile. Additionally, a decrease of pregnancy protective CD8alpha(+) decidual cells is present. Strikingly, all such uterine stress responses are abrogated by NGFB neutralization. Hence, NGFB acts as a proximal mediator in the hierarchical network of immune rejection by mediating an abortogenic environment comprised of classical signs of neurogenic inflammation.
Collapse
Affiliation(s)
- Mareike Tometten
- Charité, Universitätsmedizin Berlin, Joint Institution of the Freie Universität and Humboldt Universität, Biomedical Research Center, Campus Virchow, 13353 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Briscoe T, Duncan J, Cock M, Choo J, Rice G, Harding R, Scheerlinck JPY, Rees S. Activation of NF-κB transcription factor in the preterm ovine brain and placenta after acute LPS exposure. J Neurosci Res 2006; 83:567-74. [PMID: 16435393 DOI: 10.1002/jnr.20757] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Intrauterine infection may be causally related to inflammation and injury of the fetal brain, however the mechanisms by which this occurs are unclear. We have investigated whether nuclear factor (NF)-kappaB, a transcription factor for proinflammatory cytokines, is activated in the fetal brain after acute LPS-exposure. At 95 days of gestation (term = approximately 147 days), 5 fetuses received a single intravenous bolus dose of LPS (1 microg/kg); 6 fetuses served as controls. Fetal blood samples were taken hourly for 6 hr post LPS-exposure to assess physiological status. Ewes and fetuses were then euthanased, placental and brain tissue examined histologically, and NF-kappaB activation assessed in several regions of the fetal brain using an electromobility shift assay (EMSA). Oxidative stress was measured using lipid peroxidation and 8-isoprostane biochemical assays and brain cytokine concentrations analysed by enzyme linked immunosorbent assay (ELISA). LPS-exposed fetuses (relative to controls) were hypoxemic and the haematocrit and lactate levels had increased. In the brains of LPS-exposed fetuses compared to controls, NF-kappaB binding activity was elevated in the hippocampus and the thalamus/basal ganglia; 8-isoprostane levels were elevated overall (P < 0.05) in the parietal/occipital/temporal lobes and thalamus/basal ganglia. TNF-alpha and IL-6 concentrations were not elevated, however, there was a tendency for an elevation of IFN-gamma concentrations in the thalamus/basal ganglia. IFN-gamma concentration was elevated (P < 0.05) in the plasma 4 hr after LPS-exposure. In the placenta, NF-kappaB binding activity was increased (P < 0.05). We conclude that acute systemic administration of LPS leads to increased binding activity of NF-kappaB subunits in specific regions of the fetal brain and in the placenta, but that there is no clear-cut relationship between this elevation and vulnerability to endotoxic damage.
Collapse
Affiliation(s)
- T Briscoe
- Department of Anatomy and Cell Biology, University of Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Waters CE, Shi-Wen X, Denton CP, Abraham DJ, Pearson JD. Signaling pathways regulating intercellular adhesion molecule 1 expression by endothelin 1: Comparison with interleukin-1β in normal and scleroderma dermal fibroblasts. ACTA ACUST UNITED AC 2006; 54:649-60. [PMID: 16447227 DOI: 10.1002/art.21572] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Endothelin 1 (ET-1) has been implicated in the pathogenesis of fibrotic and inflammatory diseases, including scleroderma. In addition to modulating vascular tone and extracellular matrix turnover, ET-1 up-regulates cell surface adhesion molecules including intercellular adhesion molecule 1 (ICAM-1), which is key to cell-cell and cell-matrix adhesion and leukocyte infiltration. This study was undertaken to delineate the signal transduction pathways utilized by ET-1 and compare them with those adopted by proinflammatory cytokine interleukin-1beta (IL-1beta) in normal and scleroderma dermal fibroblasts. METHODS Protein expression induced by ET-1 and IL-1beta on normal dermal fibroblasts, with or without signaling inhibitors, was detected by enzyme-linked immunosorbent assay, while messenger RNA (mRNA) levels were analyzed by LightCycler polymerase chain reaction. Expression of protein kinase Cdelta (PKCdelta) and PKCepsilon protein in normal dermal fibroblasts and scleroderma dermal fibroblasts was determined by Western blotting, and PKCepsilon involvement in ET-1 signaling was confirmed through transfection of an ICAM-1 promoter construct into murine PKCepsilon-/- fibroblasts. NF-kappaB activation was confirmed via electrophoretic mobility supershift assay, and analysis of the ICAM-1 promoter region was achieved via transfection of deletion constructs into human dermal fibroblasts. RESULTS In normal dermal fibroblasts, ET-1 induced ICAM-1 mRNA and surface protein expression in a dose- and time-dependent manner via both receptor subtypes, ET(A) and ET(B); antagonism of both abolished the ET-1 response. MEK was involved in the signaling cascade, but phosphatidylinositol 3-kinase and p38 MAPK were not. Key to the cascade was activation of NF-kappaB, achieved by ligation of either receptor subtype. PKCepsilon activation led to downstream activation of MEK and, in part, NF-kappaB. IL-1beta signaling required NF-kappaB and MEK activation, along with activation of PKCdelta. ET-1 and IL-1beta each utilized the same ICAM-1 promoter region and the same NF-kappaB site at -157 bp. Responses to ET-1 and IL-1beta differed in scleroderma dermal fibroblasts, with ET-1 sensitivity decreasing and IL-1beta responses remaining intact. Expression of PKCepsilon and PKCdelta in scleroderma dermal fibroblasts was also altered. CONCLUSION The findings of this study indicate that differences in sensitivity to ET-1 and IL-1beta in scleroderma dermal fibroblasts may be explained by altered expression of the PKC isoforms and cytokine receptors.
Collapse
|
31
|
Curran NM, Griffin BD, O'Toole D, Brady KJ, Fitzgerald SN, Moynagh PN. The Synthetic Cannabinoid R(+)WIN 55,212-2 Inhibits the Interleukin-1 Signaling Pathway in Human Astrocytes in a Cannabinoid Receptor-independent Manner. J Biol Chem 2005; 280:35797-806. [PMID: 16105834 DOI: 10.1074/jbc.m507959200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
R(+)WIN 55,212-2 is a synthetic cannabinoid that controls disease progression in models of multiple sclerosis. This is associated with its ability to reduce migration of leukocytes into the central nervous system. Because leukocyte migration is dependent on induction of adhesion molecules and chemokines by pro-inflammatory cytokines, we examined the effects of R(+)WIN 55,212-2 on their expression. Using 1321N1 astrocytoma and A-172 glioblastoma as cell models we show that R(+)WIN 55,212-2, but not its inactive chiral form S(-)WIN 55,212-2, strongly inhibits the interleukin-1 (IL-1) induction of the adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) and the chemokine IL-8. This inhibition is not mediated via the CB1 or CB2 cannabinoid receptors, because their selective antagonists and pertussis toxin failed to affect the inhibitory effects of R(+)WIN 55,212-2. Furthermore reverse transcription-PCR analysis did not detect the expression of either receptor in 1321N1 cells. R(+)WIN 55,212-2 was shown to inhibit adhesion molecule and chemokine expression at the level of transcription, because it strongly inhibited the IL-1 induction of ICAM-1, VCAM-1, and IL-8 mRNAs and blocked the IL-1 activation of their promoters. The NFkappaB pathway was then assessed as a lead target for R(+)WIN 55,212-2. NFkappaB was measured by expression of a transfected NFkappaB-regulated reporter gene. Using this assay, R(+)WIN 55,212-2 strongly inhibited IL-1 activation of NFkappaB. Furthermore R(+)WIN 55,212-2 inhibited the ability of overexpressed Myd88, Tak-1, and IKK-2 to induce the reporter gene suggesting that R(+)WIN 55,212-2 acts at or downstream of IKK-2 in the IL-1 pathway. However R(+)WIN 55,212-2 failed to inhibit IL-1-induced degradation of IkappaBalpha, excluding IKK-2 as a direct target. In addition electrophoretic mobility shift and chromatin immunoprecipitation assays showed that R(+)WIN 55,212-2 does not regulate the IL-1-induced nuclear translocation of NFkappaB or the ability of the latter to bind to promoters regulating expression of ICAM-1 and IL-8. These data suggest that R(+)WIN 55,212-2 blocks IL-1 signaling by inhibiting the transactivation potential of NFkappaB.
Collapse
Affiliation(s)
- Niamh M Curran
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
32
|
Lavigne P, Benderdour M, Lajeunesse D, Shi Q, Fernandes JC. Expression of ICAM-1 by osteoblasts in healthy individuals and in patients suffering from osteoarthritis and osteoporosis. Bone 2004; 35:463-70. [PMID: 15268898 DOI: 10.1016/j.bone.2003.12.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2003] [Revised: 11/17/2003] [Accepted: 12/24/2003] [Indexed: 10/26/2022]
Abstract
OBJECTIVES To describe the pattern of expression of intercellular adhesion molecule-1 (ICAM-1) at the surface of human osteoblasts (Ob) recovered from normal (control), osteoporotic (OP), and osteoarthritic (OA) bone. To relate ICAM-1 expression in OA Ob with interleukin-6 (IL-6) and prostaglandin E2 (PGE2) production. MATERIALS AND METHODS Trabecular bone specimens were taken from patients suffering from OA of the hip (n = 19) or knee (n = 19) or from hip fracture caused by osteoporosis (n = 10). Control bone specimens came from the posterosuperior iliac crest (n = 5) and from the femoral condyle of organ donors (n = 6). Bone explants were digested with collagenase and cultured. Ob were obtained after 6 weeks. ICAM-1 expression was studied by immunocytology. IL-6 and PGE2 were evaluated by standard ELISA. RESULTS Average ICAM-1 expression was different between control and OP bone (P < 0.02). Separation of specimens into high and low ICAM-1 expression showed a significant difference between high and low ICAM-1 expressors. The distribution of specimens after subclassification into high or low ICAM-1 expression groups revealed only 18.2% of patients in the high expression group for the controls, compared to 70% for OP bone (P < 0.03), 52.6% for hip OA and 47.4% for knee OA. IL-6 and PGE2 levels in OA Ob from both groups were found to be significantly elevated with high ICAM-1 expression compared to low ICAM-1 expression. CONCLUSION The results show that ICAM-1 expression in human bone seems to be pathology-dependent and correlates with IL-6 and PGE2 production, at least in OA individuals. This implies that ICAM-1 could discriminate functionally different populations of Ob and possibly alter the clinical evolution of the disease.
Collapse
Affiliation(s)
- Patrick Lavigne
- Orthopaedics Research Laboratory, Department of Orthopaedics, Centre hospitalier Sacré-Coeur, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
33
|
Abou-Bacar A, Pfaff AW, Georges S, Letscher-Bru V, Filisetti D, Villard O, Antoni E, Klein JP, Candolfi E. Role of NK cells and gamma interferon in transplacental passage of Toxoplasma gondii in a mouse model of primary infection. Infect Immun 2004; 72:1397-401. [PMID: 14977944 PMCID: PMC356035 DOI: 10.1128/iai.72.3.1397-1401.2004] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protective immunity in mice infected with Toxoplasma gondii is mainly mediated by NK cells, CD4 and CD8 T cells, and type 1 cytokines, such as gamma interferon (IFN-gamma). To clarify the roles of NK cells and IFN-gamma in protection against primary congenital toxoplasmosis, we used recombination activating gene 2 knockout (RAG-2(-/-)) mice, which lack T and B lymphocytes, in comparison with the wild-type BALB/c model. RAG-2(-/-) mice had a significantly lower risk of fetal toxoplasmosis than BALB/c mice (25 versus 63.9%; P = 0.003). This protection was associated with an increased number of maternal NK cells, IFN-gamma secretion by spleen cells, and decreased parasitemia. In the RAG-2(-/-) mice, NK cell depletion increased both the rate of fetal infection, to 56.5% (P = 0.02), and the blood parasite burden. Conversely, in the BALB/c mice, this treatment did not modify maternofetal transmission or the blood parasite burden. Neutralization of IFN-gamma in both infected RAG-2(-/-) and BALB/c mice decreased congenital Toxoplasma transmission, contrasting with an exacerbation of maternal infection. These data suggest that a partially protective immunity against congenital toxoplasmosis is achieved due to the increased number of NK cells in RAG-2(-/-) mice. However, it seems that IFN-gamma enhances, directly or indirectly, the transplacental transmission.
Collapse
MESH Headings
- Animals
- Cell Count
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- Female
- Infectious Disease Transmission, Vertical
- Interferon-gamma/biosynthesis
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neutralization Tests
- Pregnancy
- Pregnancy Complications, Parasitic/immunology
- Toxoplasmosis, Animal/complications
- Toxoplasmosis, Animal/immunology
- Toxoplasmosis, Animal/transmission
- Toxoplasmosis, Congenital/etiology
- Toxoplasmosis, Congenital/immunology
Collapse
Affiliation(s)
- Ahmed Abou-Bacar
- Institut de Parasitologie et de Pathologie Tropicale, INSERM U392, 67000 Strasbourg, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mégarbane B, Marchal P, Marfaing-Koka A, Belliard O, Jacobs F, Chary I, Brivet FG. Increased diffusion of soluble adhesion molecules in meningitis, severe sepsis and systemic inflammatory response without neurological infection is associated with intrathecal shedding in cases of meningitis. Intensive Care Med 2004; 30:867-74. [PMID: 15067502 DOI: 10.1007/s00134-004-2253-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2003] [Accepted: 02/26/2004] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Sepsis and systemic inflammatory response syndrome (SIRS) result in the release in plasma of inflammatory cytokines and soluble forms of adhesion molecules in relation to endothelial activation. This study was designed to compare cerebrospinal fluid (CSF) concentrations of adhesion molecules in meningitis and SIRS without neurological infection and to evaluate in meningitis whether they originate from passive diffusion through damaged blood-CSF barrier or from local production. DESIGN Prospective observational study. SETTING University hospital medical intensive care unit. PATIENTS Nineteen patients with meningitis and 41 patients with sepsis or SIRS without cerebrospinal infection consecutively admitted to the critical care unit over an 18-month period. INTERVENTIONS Soluble forms of adhesion molecules (ICAM-1, VCAM-1, E-selectin) and cytokines (interleukin (IL)-1beta and TNF-alpha) were measured in paired CSF and blood samples. RESULTS Serum concentrations of soluble adhesion molecules and cytokines were increased in the two groups, without significant differences. The CSF concentrations were elevated in both cases, whereas patients with meningitis demonstrated significantly higher CSF concentrations of soluble ICAM-1, VCAM-1, E-selectin, and TNF-alpha ( p<0.001), with higher corresponding CSF/serum ratios. Correlations between CSF and serum concentrations were found only in meningitis. These correlations were strong for soluble ICAM-1 (r(2)=0.7, p<0.001) and E-selectin (r(2)=0.9, p<0.001), but weaker for VCAM-1. VCAM-1 CSF/serum ratios were increased, in comparison with ICAM-1 and E-selectin CSF/serum ratios, despite similar molecular weights. Serum and CSF levels of cytokines and adhesion molecules were not predictive of death for the whole population, except concentrations of ICAM-1 significantly increased in non-surviving patients ( p<0.05). CONCLUSIONS The CSF soluble adhesion molecules are increased in sepsis, SIRS and meningitis. In meningitis, the correlation between CSF and serum concentrations of adhesion molecules and the presence of a discrepancy of CSF/serum ratios for molecules of the same molecular weight may suggest intrathecal shedding in addition to diffusion through blood-CSF barrier.
Collapse
Affiliation(s)
- Bruno Mégarbane
- Department of Medical Intensive Care Unit and Medical Emergency, AP-HP Antoine Béclère Hospital, Clamart, France.
| | | | | | | | | | | | | |
Collapse
|
35
|
Maldonado M, Baybis M, Newman D, Kolson DL, Chen W, McKhann G, Gutmann DH, Crino PB. Expression of ICAM-1, TNF-alpha, NF kappa B, and MAP kinase in tubers of the tuberous sclerosis complex. Neurobiol Dis 2004; 14:279-90. [PMID: 14572449 DOI: 10.1016/s0969-9961(03)00127-x] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Individuals affected with tuberous sclerosis complex (TSC) develop cortical tubers characterized by disorganized cytoarchitecture and morphologically abnormal cell types, such as dysplastic neurons (DNs) and giant cells (GCs). As part of ongoing cDNA array analysis to study the molecular pathogenesis of tuber formation, we detected increased expression of intercellular adhesion molecule-1 (ICAM-1) mRNA, a cell adhesion molecule (CAM) that functions in cytokine signaling, in tubers. Western and immunohistochemical analyses revealed that ICAM-1 protein was selectively expressed in tubers, but was only minimally expressed in control cortex, adjacent nontuberal cortex, or in non-TSC focal cortical dysplasia. Increased expression of ICAM-1 was found in mice in which the Tsc1 gene was conditionally inactivated in astrocytes. Expression of molecules involved in ICAM-1 activation and cytokine signaling were increased in tubers, including tumor necrosis factor alpha (TNF-alpha), mitogen activated protein kinase (MAPK), and nuclear factor kappa B (NF-kappaB). Numerous CD68-immunoreactive macrophages were observed clustered around GCs further supporting an inflammatory response in tubers. Expression of caspase 8 and Fas support cytokine activation and detection of TUNEL reactivity suggests ongoing cell death in tubers. Specific alterations in ICAM-1, TNF-alpha, NF-kappaB1, and MAPK expression coupled with the detection of numerous CD68-immunoreactive macrophages suggests activation of proinflammatory cytokine signaling pathways in tubers that may culminate in cell death.
Collapse
Affiliation(s)
- Michelle Maldonado
- PENN Epilepsy Center and Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Raivich G, Bohatschek M, Werner A, Jones LL, Galiano M, Kloss CUA, Zhu XZ, Pfeffer K, Liu ZQ. Lymphocyte infiltration in the injured brain: role of proinflammatory cytokines. J Neurosci Res 2003; 72:726-33. [PMID: 12774313 DOI: 10.1002/jnr.10621] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Studies using mouse axotomised facial motoneuron model show a strong and highly selective entry of CD3+ lymphocytes into the affected nucleus, with a maximum at Day 14, which coincides with the peak of neuronal cell death, microglial phagocytosis, and increased synthesis of interleukin-1 beta (IL1beta), tumour necrosis factor-alpha (TNFalpha) and interferon-gamma (IFNgamma). We explored the possible involvement of these cytokines during the main phase of lymphocyte recruitment into the axotomised facial motor nucleus 7-21 days after nerve cut using mice homozygously deficient for IL1 receptor type 1 (IL1R1-/-), TNF receptor type 1 (TNFR1-/-), type 2 (TNFR2-/-) and type 1 and 2 (TNFR1&2-/-), IFNgamma receptor type 1 (IFNgammaR1-/-), and the appropriate controls for the genetic background. Transgenic deletion of IL1R1 led to a 54% decrease and that of TNFR2 to a 44% reduction in the number of CD3+ T-cells in the axotomised facial motor nucleus, with a similar relative decrease at Day 7, 14, and 21. Deletion of TNFR1 or IFNgammaR1 had no significant effect. Deletion of both TNFR1 and 2 (TNFR1&2-/-) caused a somewhat stronger, 63% decrease than did TNFR2 deletion alone, but this could be due to an almost complete inhibition of neuronal cell death. No mutations seemed to inhibit aggregation of CD3+ T-cells around glial nodules consisting of Ca-ion binding adaptor protein-1 (IBA1)+ phagocytotic microglia and neuronal debris. Altogether, the current data show the importance of IL1R1 and TNFR2 as the key players during the main phase of lymphocyte recruitment to the damaged part of the central nervous system.
Collapse
Affiliation(s)
- Gennadij Raivich
- Department of Neuromorphology, Max-Planck Institute for Neurobiology, Martinsried, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ledeboer A, Wierinckx A, Bol JGJM, Floris S, Renardel de Lavalette C, De Vries HE, van den Berg TK, Dijkstra CD, Tilders FJH, van dam AM. Regional and temporal expression patterns of interleukin-10, interleukin-10 receptor and adhesion molecules in the rat spinal cord during chronic relapsing EAE. J Neuroimmunol 2003; 136:94-103. [PMID: 12620647 DOI: 10.1016/s0165-5728(03)00031-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) mediate leukocyte infiltration into the CNS, in experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS). Because exogenous interleukin-10 (IL-10) inhibits ICAM-1 and VCAM-1 expression and clinical EAE, we hypothesize that endogenous IL-10 signaling may suppress expression of adhesion molecules. In a rat model of chronic relapsing EAE, expression levels of IL-10 and its receptor (IL-10R1), ICAM-1 and VCAM-1 mRNA in the spinal cord are markedly increased, whereas levels of IL-10 mRNA remain relatively low. The temporal pattern of mRNA and protein expression showed marked differences between spinal cord levels. During relapse, IL-10, IL-10R1, ICAM-1, VCAM-1 mRNA levels and neurological scores show positive correlations. We conclude that endogenous IL-10 is not a crucial factor inhibiting adhesion molecule expression in this model.
Collapse
MESH Headings
- Animals
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/metabolism
- Chemotaxis, Leukocyte/immunology
- Chronic Disease
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Regulation/genetics
- Gene Expression Regulation/immunology
- Immunohistochemistry
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/immunology
- Intercellular Adhesion Molecule-1/metabolism
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Male
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/metabolism
- Multiple Sclerosis, Relapsing-Remitting/pathology
- RNA, Messenger/immunology
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred Strains
- Reaction Time/immunology
- Receptors, Interleukin/genetics
- Receptors, Interleukin/immunology
- Receptors, Interleukin/metabolism
- Receptors, Interleukin-10
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Time Factors
- Vascular Cell Adhesion Molecule-1/genetics
- Vascular Cell Adhesion Molecule-1/immunology
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- Annemarie Ledeboer
- Department of Medical Pharmacology, Research Institute Neurosciences, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kesson AM, Cheng Y, King NJC. Regulation of immune recognition molecules by flavivirus, West Nile. Viral Immunol 2003; 15:273-83. [PMID: 12081012 DOI: 10.1089/08828240260066224] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have shown the flaviviruses can up-regulate the cell surface expression of the immune recognition molecules, major histocompatability complex class-I and class-II (MHC-I, MHC-II), ICAM-1, VCAM, and E-selectin, in an interferon-independent and tumor necrosis factor-independent manner. This up-regulation is associated with an increased transcription of the relevant genes and is due to activation of the transcription factor, nuclear factor-kappa B. The level of up-regulation is determined in part by the cell cycle position of the cell when infected with the flavivirus, as quiescent cells show a greater increase in the level of expression of the immune recognition molecules, MHC-I and ICAM-1, than cells in other phases of the cell cycle. The resultant increased cell surface expression is functional with the increased expression resulting in increased recognition by flavivirus-specific and allo-specific cytotoxic T cells.
Collapse
Affiliation(s)
- Alison M Kesson
- Department of Virology and Microbiology, The Children's Hospital at Westmead, NSW, Australia.
| | | | | |
Collapse
|
39
|
Drescher KM, Zoecklein LJ, Rodriguez M. ICAM-1 is crucial for protection from TMEV-induced neuronal damage but not demyelination. J Neurovirol 2002; 8:452-8. [PMID: 12402172 DOI: 10.1080/13550280260422767] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Previous work has suggested that the factors protecting mice from Theiler's murine encephalomyelitis virus (TMEV)-induced spinal cord demyelination are distinct from those involved in protection of the brain during the acute encephalitic phase. In this study, we examined the requirement for intercellular adhesion molecule-1 (ICAM-1) in both of these processes. During the acute phase of infection (days 7 to 10 after intracerebral infection with TMEV), no differences in brain or spinal cord pathology or virus burdens were observed between ICAM-1-knockout mice and the infected immunocompetent control mice of a similar background. Examination of brain pathology later in infection (that is, day 45 post infection [p.i.]) revealed that ICAM-1-deficient mice experienced increased levels of pathology in gray matter regions of the brain. We observed an increase in striatal damage and meningeal inflammation in the brains of TMEV-infected ICAM-1-knockout mice compared to C57BL/6J mice. Despite the increase in brain pathology, no immunoreactivity to viral antigens was detected, suggesting that the virus had been cleared by this time. Resistance to demyelination was similar in both groups, indicating that the resulting immune response was sufficient for protection of the spinal cord white matter.
Collapse
Affiliation(s)
- Kristen M Drescher
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, Nebraska, USA
| | | | | |
Collapse
|
40
|
Solà C, Casal C, Tusell JM, Serratosa J. Astrocytes enhance lipopolysaccharide-induced nitric oxide production by microglial cells. Eur J Neurosci 2002; 16:1275-83. [PMID: 12405988 DOI: 10.1046/j.1460-9568.2002.02199.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several stimuli result in glial activation and induce nitric oxide (NO) production in microglial and astroglial cells. The bacterial endotoxin lipopolysaccharide (LPS) has been widely used to achieve glial activation in vitro, and several studies show that both microglial and, to a lesser extent, astroglial cell cultures produce NO after LPS treatment. However, NO production in endotoxin-treated astrocyte cultures is controversial. We characterized NO production in microglial, astroglial and mixed glial cell cultures treated with lipopolysaccharide, measured as nitrite accumulation in the culture media. We also identified the NO-producing cells by immunocytochemistry, using specific markers for the inducible NO synthase (iNOS) isoform, microglial and astroglial cells. Only microglial cells showed iNOS immunoreactivity. Thus, contaminating microglial cells were responsible for NO production in the secondary astrocyte cultures. We then analysed the effect of astrocytes on NO production by microglial cells using microglial-astroglial cocultures, and we observed that this production was clearly enhanced in the presence of astroglial cells. Soluble factors released by astrocytes did not appear to be directly responsible for such an effect, whereas nonsoluble factors present in the cell membrane of LPS-treated astrocytes could account, at least in part, for this enhancement.
Collapse
Affiliation(s)
- Carme Solà
- Department of Pharmacology and Toxicology, Institut d'Investigacions Biomèdiques de Barcelona-CSIC, IDIBAPS, Barcelona, Spain.
| | | | | | | |
Collapse
|
41
|
Massengale JL, Gasche Y, Chan PH. Carbohydrate source influences gelatinase production by mouse astrocytes in vitro. Glia 2002; 38:240-5. [PMID: 11968061 DOI: 10.1002/glia.10070] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Molecular mediators of ischemic brain injury include intercellular adhesion molecule-1 (ICAM-1) and matrix metalloproteinase-9 (MMP-9), involved in the alteration of blood-brain barrier permeability and induced in astroglial cultures by tumor necrosis factor-alpha (TNF-alpha). Hyperglycemia is known to aggravate in vivo ischemic brain damage, while treatment with sorbitol shows benefit in reducing vasogenic brain edema. This study investigated whether a culture medium carbohydrate source could alter the astrocyte production of MMP-9 and ICAM-1 in vitro. The growth of astrocytes in 12.5 mM glucose, 25 mM glucose, or 25 mM sorbitol for 14 days did not alter cellular release of lactate dehydrogenase, uptake of Trypan blue, or surface expression of glial fibrillary acidic protein (GFAP). ICAM-1 levels were similar in astrocytes grown in glucose or sorbitol both under basal conditions and after TNF-alpha stimulation for 48 h. In contrast, levels of proMMP-9 released from astrocytes cultured for 14 days in 25 mM sorbitol reached only 55-28% of those obtained from cultures in 25 mM glucose after stimulation with 1,000 U/ml (P = 0.05) or 5,000 U/ml (P < 0.025) TNF-alpha, respectively. Limiting the duration of pre-stimulation sorbitol exposure to 48 h resulted in lower proMMP-9 levels than in glucose cultures after 5,000, but not 1,000, U/ml TNF-alpha, and differences were not significant when sorbitol exposure was further reduced to 24 h. Incubation in mixed glucose/sorbitol media did not affect the release of proMMP-9. These findings suggest that MMP-9 production may be increased in astrocytes as a consequence of glucose metabolism, which can be avoided by growth in sorbitol alone.
Collapse
Affiliation(s)
- Justin L Massengale
- Department of Neurosurgery, Program in Neurosciences, Stanford University School of Medicine, California 94305-5487, USA
| | | | | |
Collapse
|
42
|
Ren Z, Yao Q, Chen C. HIV-1 envelope glycoprotein 120 increases intercellular adhesion molecule-1 expression by human endothelial cells. J Transl Med 2002; 82:245-55. [PMID: 11896203 DOI: 10.1038/labinvest.3780418] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection is often associated with central nervous system damage and vascular complications. However, the mechanisms of this association are largely unknown. We examined the effect of HIV-1 envelope glycoprotein 120 (gp120) on cell adhesion molecule expression by endothelial cells. We found, for the first time, that both soluble and membrane-bound gp120 could significantly increase the expression of human endothelial intercellular adhesion molecule-1 (ICAM-1) at both mRNA and protein levels, but not vascular cell adhesion molecule-1 and E-selectin. The specificity of gp120-mediated response was demonstrated by blocking experiments using a specific monoclonal antibody against gp120, which successfully abolished the gp120-mediated increase of ICAM-1 expression. Furthermore, there was a significant increase of human monocytic cell line THP-1 adherence onto the gp120-treated endothelial monolayers. This increased cell adhesion was effectively blocked by either anti-gp120 or anti-ICAM antibodies. These findings suggest that HIV-1 gp120-mediated endothelial ICAM-1 expression could be one of the important mechanisms of HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Zeguang Ren
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | |
Collapse
|
43
|
Jiang J, McCool BA, Parrish AR. Cadmium- and mercury-induced intercellular adhesion molecule-1 expression in immortalized proximal tubule cells: evidence for a role of decreased transforming growth factor-beta1. Toxicol Appl Pharmacol 2002; 179:13-20. [PMID: 11884233 DOI: 10.1006/taap.2001.9345] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A definitive association between the aberrant expression of cytokines and adhesion molecules in renal failure has been established. In this regard a relationship between cytokine and adhesion molecule expression is suggested but has not been shown in models of proximal tubular cell injury. To investigate the impact of acute injury on the relationship between transforming growth factor-beta 1 (TGF-beta1) and intercellular adhesion molecule-1 (ICAM-1) expression, two immortalized mouse proximal tubular epithelial cell lines were exposed to cadmium chloride or mercuric chloride (0-50 microM) for 0-8 h. ELISA and Western blot measured expression of secreted and intercellular TGF-beta1, respectively. Direct cellular ELISA or Western blot was used to assess ICAM-1 expression. Challenge with cadmium caused a greater loss of cell viability than did mercury. Interestingly, cadmium significantly decreased the amount of TGF-beta1 in the conditioned media. Although a similar trend was seen in mercury-challenged cells, no significant differences were observed. The decrease in TGF-beta1 in the culture media was not due to decreased expression of this cytokine, as intercellular levels were not affected by metal-induced injury. Significant increases in ICAM-1 protein expression were observed following cadmium and mercury challenge. The increase in ICAM-1 appears to be due to increased mRNA, as Northern blot analysis demonstrated increased message expression following a 4-h cadmium or mercury challenge. Supplementation of the culture media with exogenous TGF-beta1 decreased basal ICAM-1 expression and attenuated the cadmium-induced increase. These data suggest that metal-induced injury is associated with increased ICAM-1 expression. The mechanism of this induction may involve the decreased TGF-beta1 in the conditioned media following metal challenge. Taken together, these studies suggest a link between cytokine and adhesion molecule expression in renal injury.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Medical Pharmacology and Toxicology, Texas A&M University System Health Science Center, College Station, Texas 77843-1114, USA
| | | | | |
Collapse
|
44
|
Abstract
Astrocytes are the major glial cell within the central nervous system (CNS) and have a number of important physiological properties related to CNS homeostasis. The aspect of astrocyte biology addressed in this review article is the astrocyte as an immunocompetent cell within the brain. The capacity of astrocytes to express class II major histocompatibility complex (MHC) antigens and costimulatory molecules (B7 and CD40) that are critical for antigen presentation and T-cell activation are discussed. The functional role of astrocytes as immune effector cells and how this may influence aspects of inflammation and immune reactivity within the brain follows, emphasizing the involvement of astrocytes in promoting Th2 responses. The ability of astrocytes to produce a wide array of chemokines and cytokines is discussed, with an emphasis on the immunological properties of these mediators. The significance of astrocytic antigen presentation and chemokine/cytokine production to neurological diseases with an immunological component is described.
Collapse
Affiliation(s)
- Y Dong
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0005, USA
| | | |
Collapse
|
45
|
Acarin L, González B, Castellano B. Glial activation in the immature rat brain: implication of inflammatory transcription factors and cytokine expression. PROGRESS IN BRAIN RESEARCH 2001; 132:375-89. [PMID: 11545004 DOI: 10.1016/s0079-6123(01)32089-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- L Acarin
- Department of Cell Biology, Physiology and Immunology, Unit of Histology, School of Medicine, Universitat Autònoma de Barcelona, Campus de Bellaterra, 08193 Bellaterra, Spain.
| | | | | |
Collapse
|
46
|
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
|
47
|
Abstract
CXC chemokine receptor 4 (CXCR4) is a co-receptor for human immunodeficiency virus (HIV) infection and is believed to be involved in the pathogenesis of AIDS-associated neurologic disorders and brain tumors. The physiological roles of CXCR4 in developmental patterning of the nervous and hematopoietic system; gastrointestinal angiogenesis; and cardiac organogenesis were established by studies in gene-targeted mice. Studies on CXCR4 expression and regulation in neuroepithelial cells are fundamental for understanding its physiopathologic roles in the central nervous system (CNS). We show here that CXCR4 expression by primary mouse astrocytes is suppressed by exposure to tumor necrosis factor-alpha (TNF-alpha). TNF-alpha caused a pronounced down-regulation of CXCR4 mRNA in a dose- and time-dependent manner. TNF-alpha-mediated decrease of CXCR4 mRNA accumulation resulted in decreased CXCR4 protein expression. As a result, the ability of stromal cell-derived factor-1alpha (SDF-1alpha) to induce activation of MAP kinases, Erk1/2 was impaired. The half life of CXCR4 mRNA in the presence and absence of TNF-alpha stimulation was comparable, suggesting that TNF-alpha down-regulated CXCR4 mRNA at the transcriptional level. These results suggest that TNF-alpha could modulate HIV and brain tumor pathogenesis and immune-mediated inflammation in the central nervous system (CNS) by regulation of CXCR4 expression.
Collapse
Affiliation(s)
- Y Han
- Department of Neurosciences, The Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
48
|
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
|
49
|
Böttner M, Krieglstein K, Unsicker K. The transforming growth factor-betas: structure, signaling, and roles in nervous system development and functions. J Neurochem 2000; 75:2227-40. [PMID: 11080174 DOI: 10.1046/j.1471-4159.2000.0752227.x] [Citation(s) in RCA: 221] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transforming growth factor-betas (TGF-betas) are among the most widespread and versatile cytokines. Here, we first provide a brief overview of their molecular biology, biochemistry, and signaling. We then review distribution and functions of the three mammalian TGF-beta isoforms, beta1, beta2, and beta3, and their receptors in the developing and adult nervous system. Roles of TGF-betas in the regulation of radial glia, astroglia, oligodendroglia, and microglia are addressed. Finally, we review the current state of knowledge concerning the roles of TGF-betas in controlling neuronal performances, including the regulation of proliferation of neuronal precursors, survival/death decisions, and neuronal differentiation.
Collapse
Affiliation(s)
- M Böttner
- Neuroanatomy and Center for Neuroscience, University of Heidelberg, Heidelberg, Germany
| | | | | |
Collapse
|
50
|
Lee SJ, Drabik K, Van Wagoner NJ, Lee S, Choi C, Dong Y, Benveniste EN. ICAM-1-induced expression of proinflammatory cytokines in astrocytes: involvement of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4658-66. [PMID: 11035109 DOI: 10.4049/jimmunol.165.8.4658] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
ICAM-1 is a transmembrane glycoprotein of the Ig superfamily involved in cell adhesion. ICAM-1 is aberrantly expressed by astrocytes in CNS pathologies such as multiple sclerosis, experimental allergic encephalomyelitis, and Alzheimer's disease, suggesting a possible role for ICAM-1 in these disorders. ICAM-1 has been shown to be important for leukocyte diapedesis through brain microvessels and subsequent binding to astrocytes. However, other functional roles for ICAM-1 expression on astrocytes have not been well elucidated. Therefore, we investigated the intracellular signals generated upon ICAM-1 engagement on astrocytes. ICAM-1 ligation by a mAb to rat ICAM-1 induced mRNA expression of proinflammatory cytokines such as IL-1alpha, IL-1beta, IL-6, and TNF-alpha. Examination of cytokine protein production revealed that ICAM-1 ligation results in IL-6 secretion by astrocytes, whereas IL-1beta and IL-1alpha protein is expressed intracellularly in astrocytes. The involvement of mitogen-activated protein kinases (MAPKs) in ICAM-1-mediated cytokine expression in astrocytes was tested, as the MAPK extracellular signal-regulated kinase (ERK) was previously shown to be activated upon ICAM-1 engagement. Our results indicate that ERK1/ERK2, as well as p38 MAPK, are activated upon ligation of ICAM-1. Studies using pharmacological inhibitors demonstrate that both p38 MAPK and ERK1/2 are involved in ICAM-1-induced IL-6 expression, whereas only ERK1/2 is important for IL-1alpha and IL-1beta expression. Our data support the role of ICAM-1 on astrocytes as an inflammatory mediator in the CNS and also uncover a novel signal transduction pathway through p38 MAPK upon ICAM-1 ligation.
Collapse
Affiliation(s)
- S J Lee
- Department of Cell Biology, University of Alabama, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|