1
|
Lu W, Wen J. The relationship among H 2S, neuroinflammation and MMP-9 in BBB injury following ischemic stroke. Int Immunopharmacol 2025; 146:113902. [PMID: 39724730 DOI: 10.1016/j.intimp.2024.113902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Blood-brain barrier (BBB) is located at the interface between the central nervous system (CNS) and the circulatory system, which maintains the microenvironmental homeostasis of the CNS. BBB damage is a result of CNS diseases, including ischemic stroke, and is a cause of CNS deterioration. Cerebral ischemia unleashes a profound inflammatory response to remove the damaged tissue in the CNS and prepare the brain for repair. However, the excessive neuroinflammation following stroke onset is associated with BBB breakdown, resulting in neuronal injury and worse neurological outcomes. Additionally, matrix metalloproteinases (MMPs) are likewise responsible for the BBB injury and participate in the pathological processes of neuroinflammation following ischemic stroke. Hydrogen sulfide (H2S) is one of gaseous signaling and freely diffusing molecules. Low concentration of H2S yields the neuroprotection against BBB damage following stroke. This review discussed the current knowledge about the detrimental roles of neuroinflammation and MMPs in BBB injury following ischemic stroke. Specifically, we provided an updated overview of H2S in protecting against BBB injury following ischemic stroke via anti-neuroinflammation and inhibiting MMP-9.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Medical Branch, Hefei Technology College, Hefei, China.
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Hou J, Jeon B, Baek J, Yun Y, Kim D, Chang B, Kim S, Kim S. High fat diet-induced brain damaging effects through autophagy-mediated senescence, inflammation and apoptosis mitigated by ginsenoside F1-enhanced mixture. J Ginseng Res 2022; 46:79-90. [PMID: 35058728 PMCID: PMC8753566 DOI: 10.1016/j.jgr.2021.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/24/2021] [Accepted: 04/11/2021] [Indexed: 01/20/2023] Open
Abstract
Background Herbal medicines are popular approaches to capably prevent and treat obesity and its related diseases. Excessive exposure to dietary lipids causes oxidative stress and inflammation, which possibly induces cellular senescence and contribute the damaging effects in brain. The potential roles of selective enhanced ginsenoside in regulating high fat diet (HFD)-induced brain damage remain unknown. Methods The protection function of Ginsenoside F1-enhanced mixture (SGB121) was evaluated by in vivo and in vitro experiments. Human primary astrocytes and SH-SY5Y cells were treated with palmitic acid conjugated Bovine Serum Albumin, and the effects of SGB121 were determined by MTT and lipid uptake assays. For in vivo tests, C57BL/6J mice were fed with high fat diet for 3 months with or without SGB121 administration. Thereafter, immunohistochemistry, western blot, PCR and ELISA assays were conducted with brain tissues. Results and conclusion SGB121 selectively suppressed HFD-induced oxidative stress and cellular senescence in brain, and reduced subsequent inflammation responses manifested by abrogated secretion of IL-6, IL-1β and TNFα via NF-κB signaling pathway. Interestingly, SGB121 protects against HFD-induced damage by improving mitophagy and endoplasmic reticulum-stress associated autophagy flux and inhibiting apoptosis. In addition, SGB121 regulates lipid uptake and accumulation by FATP4 and PPARα. SGB121 significantly abates excessively phosphorylated tau protein in the cortex and GFAP activation in corpus callosum. Together, our results suggest that SGB121 is able to favor the resistance of brain to HFD-induced damage, therefore provide explicit evidence of the potential to be a functional food. High fat diet induces oxidative stress and subsequent cellular senescence in mice brain. High fat diet induces pathologies in cortex and GFAP activation in corpus callosum. Ginsenoside F1-enhanced mixture ameliorates damaging effect by modulating autophagy flux and inflammation.
Collapse
|
3
|
Dukay B, Csoboz B, Tóth ME. Heat-Shock Proteins in Neuroinflammation. Front Pharmacol 2019; 10:920. [PMID: 31507418 PMCID: PMC6718606 DOI: 10.3389/fphar.2019.00920] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/22/2019] [Indexed: 01/01/2023] Open
Abstract
The heat-shock response, one of the main pro-survival mechanisms of a living organism, has evolved as the biochemical response of cells to cope with heat stress. The most well-characterized aspect of the heat-shock response is the accumulation of a conserved set of proteins termed heat-shock proteins (HSPs). HSPs are key players in protein homeostasis acting as chaperones by aiding the folding and assembly of nascent proteins and protecting against protein aggregation. HSPs have been associated with neurological diseases in the context of their chaperone activity, as they were found to suppress the aggregation of misfolded toxic proteins. In recent times, HSPs have proven to have functions apart from the classical molecular chaperoning in that they play a role in a wider scale of neurological disorders by modulating neuronal survival, inflammation, and disease-specific signaling processes. HSPs are gaining importance based on their ability to fine-tune inflammation and act as immune modulators in various bodily fluids. However, their effect on neuroinflammation processes is not yet fully understood. In this review, we summarize the role of neuroinflammation in acute and chronic pathological conditions affecting the brain. Moreover, we seek to explore the existing literature on HSP-mediated inflammatory function within the central nervous system and compare the function of these proteins when they are localized intracellularly compared to being present in the extracellular milieu.
Collapse
Affiliation(s)
- Brigitta Dukay
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Bálint Csoboz
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| |
Collapse
|
4
|
Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol 2019; 316:C135-C153. [PMID: 30379577 PMCID: PMC6397344 DOI: 10.1152/ajpcell.00136.2018] [Citation(s) in RCA: 546] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 12/25/2022]
Abstract
As part of the neurovascular unit, the blood-brain barrier (BBB) is a unique, dynamic regulatory boundary that limits and regulates the exchange of molecules, ions, and cells between the blood and the central nervous system. Disruption of the BBB plays an important role in the development of neurological dysfunction in ischemic stroke. Blood-borne substances and cells have restricted access to the brain due to the presence of tight junctions between the endothelial cells of the BBB. Following stroke, there is loss of BBB tight junction integrity, leading to increased paracellular permeability, which results in vasogenic edema, hemorrhagic transformation, and increased mortality. Thus, understanding principal mediators and molecular mechanisms involved in BBB disruption is critical for the development of novel therapeutics to treat ischemic stroke. This review discusses the current knowledge of how neuroinflammation contributes to BBB damage in ischemic stroke. Specifically, we provide an updated overview of the role of cytokines, chemokines, oxidative and nitrosative stress, adhesion molecules, matrix metalloproteinases, and vascular endothelial growth factor as well as the role of different cell types in the regulation of BBB permeability in ischemic stroke.
Collapse
Affiliation(s)
- Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Sylvain Doré
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
- Departments of Anesthesiology, Neurology, Psychiatry, Psychology, and Pharmaceutics, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| |
Collapse
|
5
|
Overview of Traumatic Brain Injury: An Immunological Context. Brain Sci 2017; 7:brainsci7010011. [PMID: 28124982 PMCID: PMC5297300 DOI: 10.3390/brainsci7010011] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) afflicts people of all ages and genders, and the severity of injury ranges from concussion/mild TBI to severe TBI. Across all spectrums, TBI has wide-ranging, and variable symptomology and outcomes. Treatment options are lacking for the early neuropathology associated with TBIs and for the chronic neuropathological and neurobehavioral deficits. Inflammation and neuroinflammation appear to be major mediators of TBI outcomes. These systems are being intensively studies using animal models and human translational studies, in the hopes of understanding the mechanisms of TBI, and developing therapeutic strategies to improve the outcomes of the millions of people impacted by TBIs each year. This manuscript provides an overview of the epidemiology and outcomes of TBI, and presents data obtained from animal and human studies focusing on an inflammatory and immunological context. Such a context is timely, as recent studies blur the traditional understanding of an “immune-privileged” central nervous system. In presenting the evidence for specific, adaptive immune response after TBI, it is hoped that future studies will be interpreted using a broader perspective that includes the contributions of the peripheral immune system, to central nervous system disorders, notably TBI and post-traumatic syndromes.
Collapse
|
6
|
Dahiya S, Irish BP, Nonnemacher MR, Wigdahl B. Genetic variation and HIV-associated neurologic disease. Adv Virus Res 2013; 87:183-240. [PMID: 23809924 DOI: 10.1016/b978-0-12-407698-3.00006-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
HIV-associated neurologic disease continues to be a significant complication in the era of highly active antiretroviral therapy. A substantial subset of the HIV-infected population shows impaired neuropsychological performance as a result of HIV-mediated neuroinflammation and eventual central nervous system (CNS) injury. CNS compartmentalization of HIV, coupled with the evolution of genetically isolated populations in the CNS, is responsible for poor prognosis in patients with AIDS, warranting further investigation and possible additions to the current therapeutic strategy. This chapter reviews key advances in the field of neuropathogenesis and studies that have highlighted how molecular diversity within the HIV genome may impact HIV-associated neurologic disease. We also discuss the possible functional implications of genetic variation within the viral promoter and possibly other regions of the viral genome, especially in the cells of monocyte-macrophage lineage, which are arguably key cellular players in HIV-associated CNS disease.
Collapse
Affiliation(s)
- Satinder Dahiya
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Bryan P Irish
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Jovanova-Nesic K, Jovicic S, Sovilj M, Spector NH. Magnetic brain stimulation upregulates adhesion and prevents Eae: MMP-2, ICAM-1, and VCAM-1 in the choroid plexus as a target. Int J Neurosci 2009; 119:1399-418. [PMID: 19922364 DOI: 10.1080/00207450802324564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Clinical signs appearance and significant increases of ICAM-1 and MMP-2 expressions with the clusters of VCAM-1(+) immunoreactivity in the choroids plexus epithelium to transferred anti-myelin oligodendroglial antibodies into the third brain ventricle, indicate important role of choroids plexus in the induction of acute experimental autoimmune encephalomyelitis (EAE). Magnetic brain stimulation with AKMA micro-magnet flux density of 60 miliTesla, 5 mm in diameter, implanted upon the pineal gland (PG), immediately after antibody injection, significantly decreases the expression of MMP-2 and ICAM-1 in the choroids plexus of the rat brain and abruptly suppresses the induction of acute EAE.
Collapse
Affiliation(s)
- K Jovanova-Nesic
- Immunology Research Center Branislav Jankovic, 45811152 Belgrade, Serbia.
| | | | | | | |
Collapse
|
8
|
Zhao L, Toriumi H, Kuang Y, Chen H, Fu ZF. The roles of chemokines in rabies virus infection: overexpression may not always be beneficial. J Virol 2009; 83:11808-18. [PMID: 19740983 PMCID: PMC2772667 DOI: 10.1128/jvi.01346-09] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 08/31/2009] [Indexed: 12/24/2022] Open
Abstract
It was found previously that induction of innate immunity, particularly chemokines, is an important mechanism of rabies virus (RABV) attenuation. To evaluate the effect of overexpression of chemokines on RABV infection, chemokines macrophage inflammatory protein 1alpha (MIP-1alpha), RANTES, and IP-10 were individually cloned into the genome of attenuated RABV strain HEP-Flury. These recombinant RABVs were characterized in vitro for growth properties and expression of chemokines. It was found that all the recombinant viruses grew as well as the parent virus, and each of the viruses expressed the intended chemokine in a dose-dependent manner. When these viruses were evaluated for pathogenicity in the mouse model, it was found that overexpression of MIP-1alpha further decreased RABV pathogenicity by inducing a transient innate immune response. In contrast, overexpression of RANTES or IP-10 increased RABV pathogenicity by causing neurological diseases, which is due to persistent and high-level expression of chemokines, excessive infiltration and accumulation of inflammatory cells in the central nervous system, and severe enhancement of blood-brain barrier permeability. These studies indicate that overexpression of chemokines, although important in controlling virus infection, may not always be beneficial to the host.
Collapse
Affiliation(s)
- Ling Zhao
- Departments of Pathology, Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602, State-Key Laboratory of Agricultural Microbiology, Department of Preventive Veterinary, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Harufusa Toriumi
- Departments of Pathology, Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602, State-Key Laboratory of Agricultural Microbiology, Department of Preventive Veterinary, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yi Kuang
- Departments of Pathology, Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602, State-Key Laboratory of Agricultural Microbiology, Department of Preventive Veterinary, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huanchun Chen
- Departments of Pathology, Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602, State-Key Laboratory of Agricultural Microbiology, Department of Preventive Veterinary, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhen F. Fu
- Departments of Pathology, Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602, State-Key Laboratory of Agricultural Microbiology, Department of Preventive Veterinary, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
9
|
Tumor necrosis factor alpha is not a pathogenic determinant in acute lethal encephalitis induced by a highly neurovirulent strain of mouse hepatitis virus. Arch Virol 2007; 153:549-53. [PMID: 18074094 DOI: 10.1007/s00705-007-0006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2007] [Accepted: 11/26/2007] [Indexed: 10/22/2022]
Abstract
To investigate the role of tumor necrosis factor alpha (TNFalpha) in the pathogenesis of acute viral encephalitis, TNFalpha-deficient mice were infected with a highly neurovirulent strain of mouse hepatitis virus, JHM, and compared with JHM-infected C57BL/6 mice as controls. All the JHM-infected mice had succumbed to infection by 6 days postinfection. The virus replication kinetics, histopathological changes and mRNA expression levels of proinflammatory cytokines in the brain did not differ between TNFalpha-deficient and control C57BL/6 mice. These results suggest that TNFalpha is not a pathogenic determinant in JHM-induced acute lethal encephalitis.
Collapse
|
10
|
Kinoshita K, Tanjoh K, Noda A, Sakurai A, Yamaguchi J, Azuhata T, Utagawa A, Moriya T. Interleukin-8 production from human umbilical vein endothelial cells during brief hyperglycemia: the effect of tumor necrotic factor-alpha. J Surg Res 2007; 144:127-31. [PMID: 17707405 DOI: 10.1016/j.jss.2007.03.020] [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] [Received: 12/15/2006] [Revised: 02/20/2007] [Accepted: 03/04/2007] [Indexed: 01/08/2023]
Abstract
BACKGROUND This study evaluated the changes in chemokine interleukin (IL)-8 production from endothelial cells under various hyperglycemic conditions and investigated whether the hyperglycemia associated with the acute inflammatory response could enhance the IL-8 production from the endothelial cells. MATERIALS AND METHODS Human umbilical endothelial cells (HUVECs) were seeded at a concentration of 1 x 10(5) cells/well and cultured. The culture medium was replaced with Medium 199 containing various concentrations of glucose (final glucose concentration of culture medium was 100, 200, 300, 400, 500 mg/dL; n = 7 each) with or without 100 ng of tumor necrosis factor-alpha (TNF-alpha). After 12 or 24 h at 37 degrees C, the supernatants were collected from the cultures and stored at -80 degrees C until cytokine assay. IL-8 levels of the samples from the supernatants were quantified using a commercially available enzyme-linked immunosorbent assay kit. RESULTS The IL-8 production by the HUVECs was significantly higher in the high glucose culture than in the control culture (glucose concentration of 100 mg/dL) (P < 0.05). Moreover, the hyperglycemia associated with elevated TNF-alpha was found to enhance the level of IL-8 production by the HUVECs cultured at all glucose concentrations and over both time courses, compared to the control (P < 0.05). CONCLUSIONS In this study we observed a significant augmentation of IL-8 production by endothelial cells during short-term hyperglycemia, and a similar but significantly stronger augmentation was obtained through TNF treatment. These findings suggest that the hyperglycemia associated with acute inflammatory response after trauma may put the patients at high risk for secondary tissue damage.
Collapse
Affiliation(s)
- Kosaku Kinoshita
- Department of Emergency and Critical Care Medicine, Nihon University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Jang MH, Kim CJ, Kim EH, Kim MG, Leem KH, Kim J. Effects of Platycodon grandiflorum on Lipopolysaccharide-Stimulated Production of Prostaglandin E2, Nitric Oxide, and Interleukin-8 in Mouse Microglial BV2 Cells. J Med Food 2006; 9:169-74. [PMID: 16822201 DOI: 10.1089/jmf.2006.9.169] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The roots of Platycodon grandiflorum, which belongs to the Campanulaceae family, have been used as a food material and as a traditional Oriental medicine. The effect of P. grandiflorum against lipopolysaccharide (LPS)-stimulated inflammation was investigated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, reverse transcription-polymerase chain reaction, prostaglandin E(2 )(PGE(2)) immunoassay, nitric oxide (NO) detection assay, and interleurkin-8 (IL- 8) immunoassay on BV2 microglial cells. The aqueous extract of P. grandiflorum was shown to suppress PGE(2 )synthesis and NO production by inhibiting LPS-stimulated cyclooxygenase (COX)-2 activity and expression of inducible NO synthase (iNOS) mRNAs. In addition, the treatment with P. grandiflorum reduced the LPS-induced IL-8 release. These results suggest that P. grandiflorum inhibits PGE(2) and NO production through its suppression of LPS-induced COX-2 and iNOS expression, and also reduces IL-8 secretion by microglial cells.
Collapse
Affiliation(s)
- Mi-Hyeon Jang
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
12
|
Singh KK, Hughes MD, Chen J, Spector SA. Impact of MCP-1-2518-G allele on the HIV-1 disease of children in the United States. AIDS 2006; 20:475-8. [PMID: 16439891 DOI: 10.1097/01.aids.0000200540.09856.58] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Guyon A, Banisadr G, Rovère C, Cervantes A, Kitabgi P, Melik-Parsadaniantz S, Nahon JL. Complex effects of stromal cell-derived factor-1 alpha on melanin-concentrating hormone neuron excitability. Eur J Neurosci 2005; 21:701-10. [PMID: 15733088 DOI: 10.1111/j.1460-9568.2005.03890.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Stromal cell-derived factor 1alpha (SDF-1alpha), a chemoattractant for leucocytes and neurons, and its receptor, CXCR4 are expressed in subsets of neurons of specific brain areas. In rat lateral hypothalamic area (LHA) we show, using immunocytochemistry, that CXCR4 is localized within melanin-concentrating hormone (MCH)-expressing neurons, mainly involved in feeding behaviour regulation. We investigated whether SDF-1alpha may control MCH neuronal activity. Patch-clamp recordings in rat LHA slices revealed multiple effects of SDF-1alpha on the membrane potential of MCH neurons, indirect through glutamate/GABA release and direct through GIRK current activation. Moreover, SDF-1alpha at 0.1-1 nM decreased peak and discharge frequency of action potential evoked by current pulses. These effects were further confirmed in voltage-clamp experiments, SDF-1alpha depressing both potassium and sodium currents. At 10 nM, however, SDF-1alpha increased peak and discharge frequency of action potential evoked by current pulses. Using a specific CXCR4 antagonist, we demonstrated that only the depressing effect on AP discharge was mediated through CXCR4 while the opposite effect was indirect. Together, our studies reveal for the first time a direct effect of SDF-1alpha on voltage-dependent membrane currents of neurons in brain slices and suggest that this chemokine may regulate MCH neuron activity.
Collapse
Affiliation(s)
- A Guyon
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC)-UMR 6097 CNRS, 660 Route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| | | | | | | | | | | | | |
Collapse
|
14
|
Singh KK, Ellis RJ, Marquie-Beck J, Letendre S, Heaton RK, Grant I, Spector SA. CCR2 polymorphisms affect neuropsychological impairment in HIV-1-infected adults. J Neuroimmunol 2005; 157:185-92. [PMID: 15579296 DOI: 10.1016/j.jneuroim.2004.08.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2004] [Indexed: 11/21/2022]
Abstract
CCR2 is a minor coreceptor for human immune deficiency virus-1 (HIV-1) and its impact on HIV-1-related neuropsychological impairment (NPI) remains unknown. We studied the impact of CCR2-V64I polymorphisms on the development of NPI in 121 HIV-1 patients. The CCR2-64-I allele was associated with rate of progression to NPI when measured from the first study visit (Log Rank p=0.01) or from the estimated time of seroconversion (p=0.02). CCR2-V64I was not associated with plasma or CSF HIV-1 RNA load, suggesting that the impact of CCR2 on neuropathogenesis may involve alterations in inflammatory responses within the CNS rather than a direct impact on viral entry or replication.
Collapse
Affiliation(s)
- Kumud K Singh
- Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0672, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Hashimoto M, Nitta A, Fukumitsu H, Nomoto H, Shen L, Furukawa S. Involvement of glial cell line-derived neurotrophic factor in activation processes of rodent macrophages. J Neurosci Res 2005; 79:476-87. [PMID: 15635609 DOI: 10.1002/jnr.20368] [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: 12/23/2022]
Abstract
The physiological roles of glial cell line-derived neurotrophic factor (GDNF) expressed in the microglia/macrophages of the injured spinal cord have not yet been clarified. mRNA expression of chemokines, including monocyte chemoattractant protein (MCP)-1, was evoked within 1 hr after transection of the spinal cord, and GDNF mRNA expression was similarly up-regulated. Immunohistochemical analysis showed that GDNF was coexpressed with MCP-1 in the CD11b-positive cells. Therefore, we examined further the effects of GDNF on cultured rat peritoneal macrophages. GDNF enhanced the phagocytic activity of the macrophages via GFRalpha-1, glycosylphosphatidylinositol-anchored specific binding site of GDNF, in a c-Ret-independent manner. The influence of autocrine and/or paracrine GDNF synthesis was evaluated by performing activation experiments using macrophages cultured from heterozygous (+/-) GDNF gene-deficient mice or wild-type (+/+) mice. There were no morphological differences dependent on genetic types or stimulators. However, the GDNF mRNA level, but not the MCP-1 or GFRalpha-1 mRNA level, was substantially lower in the mutant macrophages than in the +/+ cells irrespective of stimulation with MCP-1 or lipopolysaccharide (LPS). The phagocytic activity enhanced by MCP-1 or LPS was significantly lower in the mutant cells (+/-) than in the +/+ ones, demonstrating the involvement of endogenous GDNF in the activation processes of macrophages in vitro and suggesting that not only neuroprotective function but also activation of macrophages is effected by the GDNF produced after a spinal cord injury.
Collapse
Affiliation(s)
- Manabu Hashimoto
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Song L, Pachter JS. Monocyte chemoattractant protein-1 alters expression of tight junction-associated proteins in brain microvascular endothelial cells. Microvasc Res 2004; 67:78-89. [PMID: 14709405 DOI: 10.1016/j.mvr.2003.07.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The chemokine monocyte chemoattractant protein (MCP-1) is recognized to mediate extravasation of mononuclear leukocytes into the brain during a variety of neuroinflammatory conditions. In large part produced by parenchymal neural cells during these disease states, it is unclear how this chemokine can stimulate the migration of circulating leukocytes that lie behind the highly impermeant blood-brain barrier (BBB). Based on the premise that disruption of tight junctions (TJs) could foster leukocyte extravasation, experiments were conducted to test the hypothesis that MCP-1 alters the expression and/or distribution of the TJ-associated proteins zonulae occludens-1 (ZO-1) and occludin in brain microvascular endothelial cells (BMEC) comprising the BBB. Exposure to MCP-1 caused a loss in immunostaining of ZO-1 at inter-endothelial junctional regions in both cultured BMEC and isolated brain microvessels, as well as a similar effect on occludin in cultured BMEC, but did not alter occludin staining in microvessels. In cellular fractionation experiments, ZO-1 associated predominantly with the detergent-resistant cytoskeletal framework (CSK) in both cultured BMEC and brain microvessels, while a slimmer majority of occludin partitioned with the CSK. Following MCP-1 exposure, ZO-1 was reduced in the CSK fraction of cultured BMEC and microvessels, with a shift of ZO-1 to the detergent-soluble fraction in both cases. Occludin exhibited a similar pattern of MCP-1-induced loss and shift from the CSK in cultured BMEC, but remained nearly constant in microvessels. Lastly, expression of caveolin-1, a major structural component of membrane microdomains thought to be functionally complexed with TJs, was additionally altered by MCP-1 treatment of both cultured BMEC and microvessels. These results indicate that, in addition to its chemotactic activity, MCP-1 might alter BBB integrity during CNS inflammation.
Collapse
Affiliation(s)
- Li Song
- Blood-Brain Barrier Laboratory, Department of Pharmacology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | |
Collapse
|
17
|
Cardona AE, Gonzalez PA, Teale JM. CC chemokines mediate leukocyte trafficking into the central nervous system during murine neurocysticercosis: role of gamma delta T cells in amplification of the host immune response. Infect Immun 2003; 71:2634-42. [PMID: 12704138 PMCID: PMC153218 DOI: 10.1128/iai.71.5.2634-2642.2003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
According to a previous report, the degree of the host immune response highly correlates with severity of the disease in the murine model for neurocysticercosis. In wild-type mice, Mesocestoides corti infection induced a rapid and extensive accumulation of gamma delta T cells and macrophages in the brain. NK cells, dendritic cells, alpha beta T cells, and B cells were also recruited to the brain but at lower levels. In contrast, gamma delta T-cell-deficient mice exhibited decreased cellular infiltration and reduced central nervous system (CNS) pathology. To understand the mechanisms of leukocyte recruitment into the CNS, chemokine expression was analyzed in infected brains in the present study. MCP-1 (CCL2), MIP-1 alpha (CCL3), and MIP-1 beta (CCL4) were up-regulated within 2 days after M. corti infection. Protein expression of RANTES (CCL5), eotaxin (CCL11), and MIP-2 was detected later, at 1 week postinfection. Correlating with the decreased cellular infiltration, delta chain T-cell receptor-deficient (TCR delta(-/-)) mice exhibited substantially reduced levels of most of the chemokines analyzed (with the exception of eotaxin). The results suggest that gamma delta T cells play an important role in the CNS immune response by producing chemokines such as MCP-1 and MIP-1 alpha, enhancing leukocyte trafficking into the brain during murine neurocysticercosis.
Collapse
Affiliation(s)
- Astrid E Cardona
- Department of Microbiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | | | | |
Collapse
|
18
|
de Freitas MS, Spohr TCLS, Benedito AB, Caetano MS, Margulis B, Lopes UG, Moura-Neto V. Neurite outgrowth is impaired on HSP70-positive astrocytes through a mechanism that requires NF-kappaB activation. Brain Res 2002; 958:359-70. [PMID: 12470872 DOI: 10.1016/s0006-8993(02)03682-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the adult central nervous system (CNS), prominent reactive astrocytosis is seen in acute traumatic brain injury, neurodegenerative diseases and a variety of viral infections. Reactive astrocytes synthesize a number of factors that could play different roles in neuronal regeneration. In this study, the effects of thermal stress were evaluated on nuclear factor-kappaB (NF-kappaB) activation and proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) secretion in primary astrocytic cultures. The ability of HSP70-positive astrocytes to support or inhibit neurite outgrowth was investigated in neuron-astrocyte cocultures. Cultured astrocytes from cerebral cortex of rats were exposed to transient hyperthermia (42 degrees C/30 min) and incubated at 37 degrees C for different periods of recovery. During HSP70 accumulation, astrocytes extended large and thick processes associated to rearrangement of glial fibrillary acidic protein (GFAP) filaments and an increase in protein synthesis and GFAP, suggesting an astrogliosis event. A delay of NF-kappaB activation appeared closely related to TNF-alpha secretion by HSP70-positive astrocytes. These cells demonstrated a functional shift from neurite growth-promoting to non-permissive substrate. We also found that gliotoxin, a specific NF-kappaB inhibitor, partially abrogated the inhibitory ability of reactive astrocytes. These findings may suggest a involvement of NF-kappaB and TNF-alpha in modulating the failure of HSP70-positive astrocytes to provide functional support to neuritic outgrowth.
Collapse
Affiliation(s)
- Marta S de Freitas
- Departamento de Farmacologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, RJ, Brazil
| | | | | | | | | | | | | |
Collapse
|
19
|
Ma M, Wei T, Boring L, Charo IF, Ransohoff RM, Jakeman LB. Monocyte recruitment and myelin removal are delayed following spinal cord injury in mice with CCR2 chemokine receptor deletion. J Neurosci Res 2002; 68:691-702. [PMID: 12111830 DOI: 10.1002/jnr.10269] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The inflammatory response initiated after spinal cord injury (SCI) is characterized by the accumulation of macrophages at the impact site. Monocyte chemoattractant protein-1 (MCP-1) is a strong candidate for mediating chemotaxis of monocytes to the injured nervous system. To help in defining the role of MCP-1 in inflammation after SCI, we evaluated the time course of macrophage accumulation for 2 weeks following a midthoracic spinal cord contusion injury in mice lacking CCR2, a principal receptor for MCP-1. Mice with a deletion of CCR2 resulted in significantly reduced Mac-1 immunoreactivity restricted to the lesion epicenter at 7 days postinjury. The regions devoid of Mac-1 immunoreactivity corresponded to areas of reduced myelin degradation at this time. By 14 days postinjury, however, there were no differences in Mac-1 staining between CCR2 (+/+) and CCR2 (-/-) mice. Analyses of mRNA levels by RNase protection assay (RPA) revealed increases in MCP-1 as well as MCP-3 and MIP-2 mRNA at 1 day postinjury compared with 7 day postinjury. There were no differences in chemokine expression between CCR2-deficient mice and wild-type littermate controls. The CCR2-deficient mice also exhibited reduced expression of mRNA for chemokine receptors CCR1 and CCR5. Together, these results indicate that chemokines acting through CCR2 contribute to the early recruitment of monocytes to the lesion epicenter following SCI.
Collapse
Affiliation(s)
- Manhong Ma
- Department of Physiology and Cell Biology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | |
Collapse
|
20
|
Kinoshita K, Kraydieh S, Alonso O, Hayashi N, Dietrich WD. Effect of posttraumatic hyperglycemia on contusion volume and neutrophil accumulation after moderate fluid-percussion brain injury in rats. J Neurotrauma 2002; 19:681-92. [PMID: 12165130 DOI: 10.1089/08977150260139075] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The purpose of this study was to evaluate the effects of posttraumatic hyperglycemia on contusion volume and neutrophil accumulation following moderate traumatic brain injury (TBI) in rats. A parasagittal fluid-percussion (F-P) brain injury (1.8-2.1 atm) was induced in male Sprague-Dawley rats. Rats were then randomized into four trauma groups (n = 7/group) by the timing of dextrose injection (2.0 gm/kg/ip), which included (1) early (E) group: 5 min after TBI; (2) delayed (D) group: 4 h after TBI; (3) 24-h group: 24 h after TBI; or (4) control (C) group: no dextrose injection. A sham operated control group also received dextrose to document physiological parameters (n = 4). Rats were perfusion fixed 3 days following TBI, and the brains were processed for routine histopathological and immunocytochemical analysis. Contusion areas and volumes, as well as the frequency of myeloperoxidase immunoreactive polymorphonuclear leukocytes (PMNLs) were determined. Dextrose injections significantly increased blood glucose levels (p < 0.005) in all treated groups. Although acute hyperglycemia following TBI did not significantly affect total contusion volume, contusion area was significantly elevated in the early treatment group. In addition, early posttraumatic hyperglycemia enhanced neutrophil accumulation in the area of the cortical contusion (p < 0.005). In contrast, delayed induced hyperglycemia (i.e., 4 h, 24 h) did not significantly affect histopathological outcome or neutrophil accumulation. Taken together, these findings indicate that acute but not delayed hyperglycemia aggravates histopathological outcome and increased accumulation of PMNLs. Posttraumatic hyperglycemia in the acute phase may worsen traumatic outcome by enhancing secondary injury processes, including inflammation.
Collapse
Affiliation(s)
- Kosaku Kinoshita
- Department of Neurological Surgery, University of Miami School of Medicine, Miami, Florida 33101, USA
| | | | | | | | | |
Collapse
|
21
|
Mukhtar M, Harley S, Chen P, BouHamdan M, Patel C, Acheampong E, Pomerantz RJ. Primary isolated human brain microvascular endothelial cells express diverse HIV/SIV-associated chemokine coreceptors and DC-SIGN and L-SIGN. Virology 2002; 297:78-88. [PMID: 12083838 DOI: 10.1006/viro.2002.1376] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chemokines have received increasing attention due to their inhibitory activities on human immunodeficiency virus type-1 (HIV-1) and simian immunodeficiency virus (SIV) replication and the potential for chemokine receptors to assist in HIV-1/SIV entry into permissive cells. Besides CD4, which is the major receptor for HIV-1 and SIV, a number of chemokine receptors including but not limited to APJ, CCR3, CXCR4, and CCR5 may be coreceptors for HIV-1/SIV, not only in peripheral blood and lymphoid tissues but also in the central nervous system (CNS). The present studies reveal the lack of CD4, but the significant expression of various chemokine receptors, APJ, CCR3, CXCR4, and CCR5, plus C-type lectins DC-SIGN and L-SIGN on isolated primary human brain microvascular endothelial cells (MVECs). As these MVECs do not express CD4, this suggests a CD4-independent HIV/SIV entry/infection of these cells, which are the major cells constituting the human blood-brain barrier. We also found that chemokines for cognate chemokine receptors individually were unable to block binding of HIV-1 to brain MVECs. These results reveal that in primary isolated brain MVECs viral attachment is mediated by a possible previously unknown receptor(s) or by cooperative activity of various receptors. Moreover, mRNA transcripts for DC-SIGN/L-SIGN, as well as DC-SIGN protein expression, suggest the capability of MVECs to attach viral particles on cell surfaces, even though polyclonal antisera for DC-SIGN did not affect viral binding to these cells. These data will assist in further understanding lentiviral entry into the CNS.
Collapse
MESH Headings
- Base Sequence
- Brain/blood supply
- Brain/virology
- Cell Adhesion Molecules
- Cells, Cultured
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/virology
- Fluorescent Antibody Technique
- HIV-1/metabolism
- Humans
- Lectins/genetics
- Lectins/metabolism
- Lectins, C-Type
- Molecular Sequence Data
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- RNA, Messenger/analysis
- Receptors, Antigen/genetics
- Receptors, Antigen/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Alignment
- Simian Immunodeficiency Virus/metabolism
- Viral Proteins/metabolism
- Virus Replication
Collapse
Affiliation(s)
- Muhammad Mukhtar
- The Dorrance H. Hamilton Laboratories, Center for Human Virology, Division of Infectious Diseases, Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Iarlori C, Reale M, De Luca G, Di Iorio A, Feliciani C, Tulli A, Conti P, Gambi D, Lugaresi A. Interferon beta-1b modulates MCP-1 expression and production in relapsing-remitting multiple sclerosis. J Neuroimmunol 2002; 123:170-9. [PMID: 11880161 DOI: 10.1016/s0165-5728(01)00487-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) seems to be involved in the pathogenesis of multiple sclerosis (MS). We found that in unstimulated (PHA(-)) and PHA-stimulated (PHA(+)) peripheral blood mononuclear cells (PBMC), MCP-1 and TNFalpha levels are higher in stable untreated MS patients. Interferon gamma (IFNgamma) is higher in relapsing patients in PHA(-) cultures and in stable patients in PHA(+) cultures. Chronic IFNbeta-1b treatment down-regulates TNFalpha, IFNgamma and MCP-1 production except for TNFalpha in relapsing patients. IFNbeta-1b, in vitro, increases MCP-1, TNFalpha and IFNgamma spontaneous production in all patients. Multivariate analysis suggests that MCP-1 production is dependent from clinical status and not from TNFalpha and IFNgamma production. Logistic regression analysis shows that MCP-1 production is significantly modified by treatment. Further studies are needed to clarify the role of MCP-1 in MS.
Collapse
Affiliation(s)
- Carla Iarlori
- Department of Oncology and Neuroscience, Neuroimmunology Unit, University "G. d'Annunzio", Via dei Vestini, I-66013, Chieti, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Little AR, O'Callagha JP. Astrogliosis in the adult and developing CNS: is there a role for proinflammatory cytokines? Neurotoxicology 2001; 22:607-18. [PMID: 11770882 DOI: 10.1016/s0161-813x(01)00032-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Astrogliosis, characterized by the enhanced expression of GFAP, represents a remarkably homotypic response of astrocytes to all types of injuries of the CNS, including injuries of the developing CNS. As such, astrocytes serve as microsensors of the injured microenvironment regardless of their location in the CNS. The diversity of insults that engender astrogliosis and the brain-wide nature of the astrocytic response suggest that common injury factors serve as the trigger of this cellular reaction. One prominent theme that has emerged in recent years is that proinflammatory cytokines and chemokines serve as a stimulus for induction of astrogliosis. Here we present a brief critique of this hypothesis based on a review of literature and some of our own recentfindings. Studies of astrocytes, in vitro, clearly indicate that these cell types are responsive to a variety of growth factors, including cytokines and chemokines. A somewhat different picture, however, can be seen from data obtained in vivo. It is true that trauma and diseases of the nervous system, as well as some exposures to neurotoxic chemicals, can be associated with the expression in brain of large varieties of cytokines and chemokines. That these same conditions result in astrogliosis has fostered the circumstantial link between cytokine/chemokine expression and the induction of astrogliosis. Several lines of evidence argue against this view, including (a) suppression of cytokine expression does not suppress gliosis, (b) gliosis can occur in the absence of enhanced expression of cytokines, (c) elevations in brain cytokines can occur in the absence of gliosis and (d) the patterns of cytokine expression in the adult and developing CNS are more consistent with a trophic role for these chemical messengers rather than a role in the induction of inflammation. Enhanced expression of cytokines and chemokines after brain injury appear to be signal transduction events unrelated to the induction of astrogliosis.
Collapse
Affiliation(s)
- A R Little
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | | |
Collapse
|
24
|
Restrepo BI, Alvarez JI, Castaño JA, Arias LF, Restrepo M, Trujillo J, Colegial CH, Teale JM. Brain granulomas in neurocysticercosis patients are associated with a Th1 and Th2 profile. Infect Immun 2001; 69:4554-60. [PMID: 11401999 PMCID: PMC98532 DOI: 10.1128/iai.69.7.4554-4560.2001] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neurocysticercosis (NCC) is a common central nervous system (CNS) infection caused by Taenia solium metacestodes. Despite the well-documented importance of the granulomatous response in the pathogenesis of this infection, there is limited information about the types of cells and cytokines involved. In fact, there has been limited characterization of human brain granulomas with any infectious agent. In the present study a detailed histological and immunohistochemical analysis of the immune response was performed on eight craniotomy specimens where a granuloma surrounded each T. solium metacestode. The results indicated that in all the specimens there was a dying parasite surrounded by a mature granuloma with associated fibrosis, angiogenesis, and an inflammatory infiltrate. The most abundant cell types were plasma cells, B and T lymphocytes, macrophages, and mast cells. Th1 cytokines were prevalent and included gamma interferon, interleukin-18 (IL-18), and the immunosuppressive, fibrosis-promoting cytokine transforming growth factor beta. The Th2 cytokines IL-4, IL-13, and IL-10 were also present. These observations indicate that a chronic immune response is elicited in the CNS environment with multiple cell types that together secrete inflammatory and anti-inflammatory cytokines. In addition, both collagen type I and type III deposits were evident and could contribute to irreversible nervous tissue damage in NCC patients.
Collapse
Affiliation(s)
- B I Restrepo
- Molecular Parasitology Group, Corporación para Investigaciones Biológicas, Medellín, Colombia, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Glabinski AR, O'Bryant S, Selmaj K, Ransohoff RM. CXC chemokine receptors expression during chronic relapsing experimental autoimmune encephalomyelitis. Ann N Y Acad Sci 2001; 917:135-44. [PMID: 11268337 DOI: 10.1111/j.1749-6632.2000.tb05377.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chemokines are small proinflammatory cytokines that possess the ability to stimulate migration of inflammatory cells towards the tissue site of inflammation. Previous reports showed that several chemokines may be involved in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of autoimmune central nervous system (CNS) inflammation. Inflammatory cells respond to chemotactic chemokine gradient through the chemokine receptors (ChRs). The goal of this study was to analyze expression of ChRs belonging to CXC subfamily during different stages of chronic relapsing EAE. We found significantly increased expression of CXCR2 and CXCR4 in the spinal cord during the first and second disease attacks. The kinetics of this expression in CNS and blood suggests that CXCR2 is expressed by leukocytes migrating from the blood, but CXCR4 is expressed mainly by CNS parenchymal cells. Those results support the interpretation that chemokine-chemokine receptor interactions may play an important role in the development of CNS autoimmune inflammation.
Collapse
Affiliation(s)
- A R Glabinski
- Department of Neurology, Medical University of Lodz, ul. Kopcinskiego 22, 90-153 Lodz, Poland.
| | | | | | | |
Collapse
|
26
|
Andjelkovic AV, Pachter JS. Characterization of binding sites for chemokines MCP-1 and MIP-1alpha on human brain microvessels. J Neurochem 2000; 75:1898-906. [PMID: 11032879 DOI: 10.1046/j.1471-4159.2000.0751898.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The presence of binding sites for the beta chemokines monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1alpha (MIP-1alpha) has recently been identified on human brain microvessels. We extend these findings in this report to reveal that such sites exemplify characteristics of the recognized major receptors for MCP-1 and MIP-1alpha: CCR2, and CCR1 and CCR5, respectively. Specifically, labeled MCP-1 binding to isolated brain microvessels was inhibited by unlabeled MCP-1 and MCP-3, the latter another CCR2 ligand, but not by MIP-1alpha. Inhibition of labeled MIP-1alpha binding was achieved with unlabeled MIP-1alpha and RANTES, the latter a beta chemokine that binds to both CCR1 and CCR5, but not by MCP-1. Labeled MIP-1alpha binding was also antagonized by unlabeled MCP-3, which is also recognized by CCR1, and MIP-1beta, which is a ligand for CCR5. Labeled MCP-1 and MIP-1alpha were further observed to be internalized within the endothelial cells of brain microvessels, following their binding to the microvascular surface at 37 degrees C. Additionally, exposure of microvessels to unlabeled MCP-1 or MIP-1alpha was accompanied by the initial loss and subsequent recovery of surface binding sites for these chemokines, which occurred on a time scale consistent with ligand-induced endocytosis and recycling. These collective features bear striking similarity to those that characterize interactions of MCP-1 and MIP-1alpha with their receptors on leukocytes and underscore the concept of cognate chemokine receptors on brain microvascular endothelium.
Collapse
Affiliation(s)
- A V Andjelkovic
- Blood-Brain Barrier Laboratory, Department of Pharmacology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | |
Collapse
|
27
|
Zhang W, Smith C, Howlett C, Stanimirovic D. Inflammatory activation of human brain endothelial cells by hypoxic astrocytes in vitro is mediated by IL-1beta. J Cereb Blood Flow Metab 2000; 20:967-78. [PMID: 10894180 DOI: 10.1097/00004647-200006000-00009] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Leukocyte infiltration into the brain contributes to the development of ischemic brain damage and is mediated by endothelial/leukocyte adhesion molecules, cytokines, and chemokines released by ischemic brain cells. In this study, we provide evidence that human astrocytes (FHAs) subjected to in vitro hypoxia produce proinflammatory mediator(s) capable of up-regulating inflammatory genes, including intercellular adhesion molecule-1, interleukin (IL)-1beta, tumor necrosis factor-alpha, IL-8, and monocyte chemotactic protein-1 (MCP-1) in human cerebromicrovascular endothelial cells (HCECs). FHAS were exposed to hypoxia in an anaerobic chamber for 4 hours, followed by reoxygenation for 24 hours. Astrocyte-conditioned media (ACM) collected from normoxic FHAS or FHAS subjected to hypoxia/reoxygenation were applied to HCEC cultures for 4 to 24 hours. Semiquantitative reverse transcription-polymerase chain reaction, immunocytochemistry, and enzyme-linked immunosorbent assay demonstrated up-regulation of intercellular adhesion molecule-1 in HCECs exposed to hypoxic ACM. A pronounced elevation in cytokine IL-1beta and tumor necrosis factor-alpha, and chemokine IL-8 and MCP-1 mRNA, accompanied by increased release of immunoreactive cytokines and chemokines into cell media was observed in HCECs exposed to hypoxic ACM. Hypoxia/reoxygenation induced a transient (4 to 18 hours of reoxygenation) up-regulation of IL-1beta mRNA in FHAS and a two- to threefold increase in IL-1beta levels secreted into ACM. Pretreatment of FHAS with 10 micromol/L dexamethasone inhibited both hypoxia-induced expression/secretion of IL-1beta and the ability of hypoxic ACM to induce inflammatory phenotype in HCECs. The ability of hypoxic ACM to up-regulate inflammatory genes in HCECs was inhibited in the presence of IL-1 receptor antagonist (IL-1Ra) and by pretreating ACM with the blocking anti-IL-1beta antibody. These findings strongly implicate IL-1beta secreted by hypoxic astrocytes in triggering inflammatory activation of HCECs and thereby influencing inflammatory responses at the site of the blood-brain barrier.
Collapse
Affiliation(s)
- W Zhang
- Cellular Neurobiology Group, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario
| | | | | | | |
Collapse
|
28
|
Woodroofe N, Cross AK, Harkness K, Simpson JE. The role of chemokines in the pathogenesis of multiple sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 468:135-50. [PMID: 10635025 DOI: 10.1007/978-1-4615-4685-6_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Affiliation(s)
- N Woodroofe
- Biomedical Research Centre, Sheffield Hallam University, South Yorkshire, U.K.
| | | | | | | |
Collapse
|
29
|
|
30
|
Zhang W, Smith C, Shapiro A, Monette R, Hutchison J, Stanimirovic D. Increased expression of bioactive chemokines in human cerebromicrovascular endothelial cells and astrocytes subjected to simulated ischemia in vitro. J Neuroimmunol 1999; 101:148-60. [PMID: 10580798 DOI: 10.1016/s0165-5728(99)00137-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Leukocyte infiltration into the brain has been implicated in the development of ischemic brain damage. In this study, simulated in vitro ischemia/reperfusion and IL-1beta were found to up-regulate both the expression of intercellular adhesion molecule- (ICAM-1) in cultured human cerebromicrovascular endothelial cells (HCEC) and the adhesion of allogenic neutrophils to HCEC. Both HCEC and human fetal astrocytes (FHAS) also responded to IL-1beta and to in vitro ischemia/reperfusion by a pronounced up-regulation of IL-8 and MCP-1 mRNA and by increased release of IL-8 and MCP-1 in cell culture media. FHAS were found to release 30-times higher levels of MCP-1 than HCEC under both basal and ischemic conditions. However, 100 u/ml IL-1beta induced greater stimulation of both IL-8 and MCP-1 secretion in HCEC (50 and 20 times above controls, respectively) than in FHAS (three and two times above controls, respectively). IL-8 was the principal neutrophil chemoattractant released from IL-1beta-treated HCEC, since IL-8 antibody completely inhibited neutrophil chemotaxis enticed by HCEC media. However, the IL-8 antibody neutralized only 50% of IL-1beta-stimulated neutrophil chemoattractants released from FHAS, and 40%-60% of ischemia-stimulated chemotactic activity released by either HCEC or FHAS. These results suggest that simulated in vitro ischemia, in addition to IL-8 and MCP-1, stimulates secretion of other bioactive chemokines from HCEC and FHAS.
Collapse
Affiliation(s)
- W Zhang
- Institute for Biological Sciences, National Research Council of Canada, Ottawa
| | | | | | | | | | | |
Collapse
|
31
|
Andjelkovic AV, Spencer DD, Pachter JS. Visualization of chemokine binding sites on human brain microvessels. J Biophys Biochem Cytol 1999; 145:403-12. [PMID: 10209033 PMCID: PMC2133113 DOI: 10.1083/jcb.145.2.403] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The chemokines monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1alpha (MIP-1alpha) aid in directing leukocytes to specific locales within the brain and spinal cord during central nervous system inflammation. However, it remains unclear how these chemokines exert their actions across a vascular barrier, raising speculation that interaction with endothelial cells might be required. Therefore, experiments were performed to determine whether binding domains for these chemokines exist along the outer surface of brain microvessels, a feature that could potentially relay chemokine signals from brain to blood. Using a biotinylated chemokine binding assay with confocal microscopy and three-dimensional image reconstruction, spatially resolved binding sites for MCP-1 and MIP-alpha around human brain microvessels were revealed for the first time. Binding of labeled MCP-1 and MIP-1alpha could be inhibited by unlabeled homologous but not heterologous chemokine, and was independent of the presence of heparan sulfate, laminin, or collagen in the subendothelial matrix. This is the first evidence of specific and separate binding domains for MCP-1 and MIP-1alpha on the parenchymal surface of microvessels, and highlights the prospect that specific interactions of chemokines with microvascular elements influence the extent and course of central nervous system inflammation.
Collapse
Affiliation(s)
- A V Andjelkovic
- Blood-Brain Barrier Laboratory, Department of Pharmacology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | |
Collapse
|
32
|
Asensio VC, Lassmann S, Pagenstecher A, Steffensen SC, Henriksen SJ, Campbell IL. C10 is a novel chemokine expressed in experimental inflammatory demyelinating disorders that promotes recruitment of macrophages to the central nervous system. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:1181-91. [PMID: 10233856 PMCID: PMC1866572 DOI: 10.1016/s0002-9440(10)65370-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/13/1999] [Indexed: 10/18/2022]
Abstract
Chemokines may be important in the control of leukocytosis in inflammatory disorders of the central nervous system. We studied cerebral chemokine expression during the evolution of diverse neuroinflammatory disorders in transgenic mice with astrocyte glial fibrillary acidic protein-targeted expression of the cytokines IL-3, IL-6, or IFN-alpha and in mice with experimental autoimmune encephalomyelitis. Distinct chemokine gene expression patterns were observed in the different central nervous system inflammatory models that may determine the phenotype and perhaps the functions of the leukocytes that traffic into the brain. Notably, high expression of C10 and C10-related genes was found in the cerebellum and spinal cord of GFAP-IL3 mice with inflammatory demyelinating disease and in mice with experimental autoimmune encephalomyelitis. In both these neuroinflammatory models, C10 RNA and protein expressing cells were predominantly macrophage/microglia and foamy macrophages present within demyelinating lesions as well as in perivascular infiltrates and meninges. Intracerebroventricular injection of recombinant C10 protein promoted the recruitment of large numbers of Mac-1(+) cells and, to a much lesser extent, CD4(+) lymphocytes into the meninges, choroid plexus, ventricles, and parenchyma of the brain. Thus, C10 is a prominent chemokine expressed in the central nervous system in experimental inflammatory demyelinating disease that, we show, also acts as a potent chemotactic factor for the migration of these leukocytes to the brain.
Collapse
Affiliation(s)
- V C Asensio
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
33
|
Nataf S, Davoust N, Barnum SR. Kinetics of anaphylatoxin C5a receptor expression during experimental allergic encephalomyelitis. J Neuroimmunol 1998; 91:147-55. [PMID: 9846831 DOI: 10.1016/s0165-5728(98)00169-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this study, we investigated the expression of the C5aR in spinal cords of Lewis rats with experimental allergic encephalomyelitis (EAE). Using in situ hybridization (ISH) we analyzed the kinetics of C5aR at different time points of EAE (preclinical stage, clinical peak, remission phase). We observed that C5aR mRNA was readily detected in the CNS of EAE rats at all the stages of the disease. Using a combination of ISH and immunohistochemistry, we formally demonstrated that C5aR is strongly expressed on microglial cells and hypertrophic astrocytes during EAE. The potential involvement of C5a receptor in EAE physiopathology is discussed.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/immunology
- Female
- Gene Expression/immunology
- Kinetics
- Macrophages/chemistry
- Macrophages/immunology
- Microglia/chemistry
- Microglia/immunology
- Monocytes/chemistry
- Monocytes/immunology
- Multiple Sclerosis/immunology
- RNA, Messenger/analysis
- Rats
- Rats, Inbred Lew
- Receptor, Anaphylatoxin C5a
- Receptors, Complement/genetics
- Receptors, Complement/immunology
- Spinal Cord/blood supply
- Spinal Cord/cytology
- Spinal Cord/immunology
Collapse
Affiliation(s)
- S Nataf
- Department of Microbiology, University of Alabama at Birmingham, 35294, USA
| | | | | |
Collapse
|
34
|
Kelder W, McArthur JC, Nance-Sproson T, McClernon D, Griffin DE. Beta-chemokines MCP-1 and RANTES are selectively increased in cerebrospinal fluid of patients with human immunodeficiency virus-associated dementia. Ann Neurol 1998; 44:831-5. [PMID: 9818943 DOI: 10.1002/ana.410440521] [Citation(s) in RCA: 248] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Human immunodeficiency virus-associated dementia (HAD) is associated with increased numbers of activated central nervous system (CNS) macrophages. Chemokines, which regulate infiltration of macrophages, were measured in the cerebrospinal fluid (CSF) of human immunodeficiency virus (HIV)-negative and HIV-positive individuals with and without neurological disease. Monocyte chemotactic protein (MCP)-1 and RANTES (but not MCP-3), macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, or interleukin-8 (IL-8) was higher in HAD. MCP-1 correlated with CSF viral load and severity of dementia, and it increased over time in patients who developed dementia.
Collapse
Affiliation(s)
- W Kelder
- Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Hygiene and Public Health, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
35
|
Majumder S, Zhou LZ, Chaturvedi P, Babcock G, Aras S, Ransohoff RM. Regulation of human IP-10 gene expression in astrocytoma cells by inflammatory cytokines. J Neurosci Res 1998; 54:169-80. [PMID: 9788276 DOI: 10.1002/(sici)1097-4547(19981015)54:2<169::aid-jnr5>3.0.co;2-c] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Because of its prominent expression in central nervous system inflammatory pathology by astrocytes, we examined the mechanism of human IP-10 (hIP-10) gene induction by interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha) in astrocytoma cells. When present together, IFN-gamma and TNF-alpha induced robust accumulation of hIP-10 mRNA, but hIP-10 mRNA was minimally induced when astrocytoma cells were treated with individual cytokines. This pattern of expression resembled that previously described for murine IP-10 (mIP-10) gene induction in fibroblasts and in rat astroglia. Nuclear run-on experiments showed that the synergistic effect of the cytokines resulted from an increased rate of IP-10 transcriptional initiation. Functional analysis of the hIP-10 promoter after deletion and substitution mutagenesis indicated that an interferon-stimulated response element (ISRE) governed both simple response to IFN-gamma and synergy with TNF-alpha. Synergistic induction of hIP-10 also required an ISRE-proximal nuclear factor kappa-B (NFkappaB) binding site. TNF-alpha-induced NFkappaB binding activity at this site was composed of RelA (p65) homodimers. Our results document that cis-elements through which cytokines mediate synergistic induction of IP-10 in mouse and human are strictly conserved despite divergence elsewhere within the proximal 5'-flanking region.
Collapse
Affiliation(s)
- S Majumder
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Mehta PS, Bruccoleri A, Brown HW, Harry GJ. Increase in brain stem cytokine mRNA levels as an early response to chemical-induced myelin edema. J Neuroimmunol 1998; 88:154-64. [PMID: 9688337 DOI: 10.1016/s0165-5728(98)00116-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This study examined the early response of pro-inflammatory and regulatory cytokines in the mouse brain following triethyltin (TET)-induced myelin injury characterized by edematous vacuolation. Following an acute intraperitoneal injection of triethyltin (TET) sulfate (3 mg/kg) to 17-day old CD1 mice, significant increases in brain stem TNF-alpha and IL-1alpha mRNA levels occurred at 6 and 24 h, respectively with elevations in TGF-beta1 and MIP-1alpha at 1 h. In the cortex, responses were limited to elevations at 6 h in TNF-alpha, TGF-beta1 and MIP-1alpha. These data suggest that a chemokine/cytokine response can occur with minimal alterations to the integrity of the myelin sheath and may contribute to the initial signaling mechanisms associated with demyelinating disorders.
Collapse
Affiliation(s)
- P S Mehta
- Department of Chemistry, University of North Carolina at Chapel Hill, 27599, USA
| | | | | | | |
Collapse
|
38
|
Cole KE, Strick CA, Paradis TJ, Ogborne KT, Loetscher M, Gladue RP, Lin W, Boyd JG, Moser B, Wood DE, Sahagan BG, Neote K. Interferon-inducible T cell alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med 1998; 187:2009-21. [PMID: 9625760 PMCID: PMC2212354 DOI: 10.1084/jem.187.12.2009] [Citation(s) in RCA: 680] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/1997] [Revised: 03/31/1998] [Indexed: 11/21/2022] Open
Abstract
Chemokines are essential mediators of normal leukocyte trafficking as well as of leukocyte recruitment during inflammation. We describe here a novel non-ELR CXC chemokine identified through sequence analysis of cDNAs derived from cytokine-activated primary human astrocytes. This novel chemokine, referred to as I-TAC (interferon-inducible T cell alpha chemoattractant), is regulated by interferon (IFN) and has potent chemoattractant activity for interleukin (IL)-2-activated T cells, but not for freshly isolated unstimulated T cells, neutrophils, or monocytes. I-TAC interacts selectively with CXCR3, which is the receptor for two other IFN-inducible chemokines, the IFN-gamma-inducible 10-kD protein (IP-10) and IFN-gamma- induced human monokine (HuMig), but with a significantly higher affinity. In addition, higher potency and efficacy of I-TAC over IP-10 and HuMig is demonstrated by transient mobilization of intracellular calcium as well as chemotactic migration in both activated T cells and transfected cell lines expressing CXCR3. Stimulation of astrocytes with IFN-gamma and IL-1 together results in an approximately 400,000-fold increase in I-TAC mRNA expression, whereas stimulating monocytes with either of the cytokines alone or in combination results in only a 100-fold increase in the level of I-TAC transcript. Moderate expression is also observed in pancreas, lung, thymus, and spleen. The high level of expression in IFN- and IL-1-stimulated astrocytes suggests that I-TAC could be a major chemoattractant for effector T cells involved in the pathophysiology of neuroinflammatory disorders, although I-TAC may also play a role in the migration of activated T cells during IFN-dominated immune responses.
Collapse
MESH Headings
- Amino Acid Sequence
- Astrocytes
- Base Sequence
- Calcium/metabolism
- Chemokine CXCL11
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Chemotaxis, Leukocyte
- Chromosomes, Human, Pair 4
- Cloning, Molecular
- DNA, Complementary/genetics
- Desensitization, Immunologic
- Humans
- Interferon-gamma/pharmacology
- Lymphocyte Activation
- Molecular Sequence Data
- Protein Binding
- RNA, Messenger/biosynthesis
- Receptors, CXCR3
- Receptors, Chemokine/metabolism
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- K E Cole
- Department of Molecular Sciences, Central Research Division, Pfizer Inc., Groton, Connecticut 06340, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Rajan AJ, Klein JDS, Brosnan CF. The Effect of γδ T Cell Depletion on Cytokine Gene Expression in Experimental Allergic Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.12.5955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
In experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis, we showed previously that depletion of γδ T cells using the mAb GL3 immediately before disease onset, or during the chronic phase, significantly ameliorated clinical severity. We now report on the effect of γδ T cell depletion on expression of five cytokine genes, IL-1, IL-6, TNF, lymphotoxin, and IFN-γ in spinal cords of mice during the pre-onset, onset, height, and recovery phases of EAE, and on expression of type II nitric oxide synthase. In control animals, the mRNAs for IL-1 and IL-6 rose dramatically at disease onset and peaked before disease height, whereas the mRNAs for TNF, lymphotoxin, and IFN-γ rose more slowly and peaked with peak of disease. In GL3-treated animals, a dramatic reduction in all five cytokines was noted at disease onset, but only IFN-γ remained significantly reduced at a time point equivalent to height of disease in control animals. ELISA data confirmed the reduced levels of IL-1 and IL-6 at disease onset in GL3-treated animals, and pathologic analysis demonstrated a marked reduction in meningeal infiltrates at the same time point. Studies of type II NOS also demonstrated a significant reduction in both mRNA and protein expression at the height of disease in GL3-treated animals. These results suggest that γδ T cells contribute to the pathogenesis of EAE by regulating the influx of inflammatory cells into the spinal cord and by augmenting the proinflammatory cytokine profile of the inflammatory infiltrates.
Collapse
Affiliation(s)
- Alice J. Rajan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Celia F. Brosnan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
40
|
Ghirnikar RS, Lee YL, Li JD, Eng LF. Chemokine inhibition in rat stab wound brain injury using antisense oligodeoxynucleotides. Neurosci Lett 1998; 247:21-4. [PMID: 9637400 DOI: 10.1016/s0304-3940(98)00268-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Traumatic injury to the central nervous system (CNS) results in the breakdown of the blood-brain barrier and recruitment of hematogenous cells at the site of injury. The role of chemokines in this process has been well recognized and they have been regarded as promising targets for development of anti-inflammatory therapies. The expression of monocyte chemoattractant protein (MCP-1), in particular, has been closely linked to macrophage infiltration following trauma in rat brain. In this study we determined whether inhibition of MCP-1 following stab wound injury would reduce macrophage infiltration. Stab wound injured Sprague-Dawley rats were infused with MCP-1 sense or antisense oligonucleotides using an Alzet miniosmotic pump (1 microl/h for 3 days). Three days following injury, widespread gliosis was observed in both groups of rats as judged by glial fibrillary acidic protein (GFAP) immunoreactivity. Immunohistochemistry showed significantly less staining for MCP-1 in antisense treated animals. In addition, the number of macrophages were reduced by 30% in the antisense compared to the sense treated animals (P < 0.05). These results demonstrate that modulation of MCP-1 expression in stab wound injury directly affects monocytic infiltration and provide a basis for MCP-1 inhibition as a therapeutic strategy for controlling inflammatory events of traumatic brain injury.
Collapse
Affiliation(s)
- R S Ghirnikar
- Department of Pathology, VAPA Health Care System, Palo Alto, CA 94304, USA
| | | | | | | |
Collapse
|
41
|
Glabinski AR, Tuohy VK, Ransohoff RM. Expression of chemokines RANTES, MIP-1alpha and GRO-alpha correlates with inflammation in acute experimental autoimmune encephalomyelitis. Neuroimmunomodulation 1998; 5:166-71. [PMID: 9730682 DOI: 10.1159/000026333] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an investigator-initiated disorder that serves as an animal model for the common human demyelinating disease multiple sclerosis. Both diseases are typified by disseminated perivascular and submeningeal cuffs in the central nervous system (CNS). It was shown recently that chemokines are integral to the pathogenesis of EAE. In the present study we analyzed the gene expression of three chemokines, RANTES, MIP-1alpha and GRO-alpha, at the onset of acute EAE, and correlated that expression with the intensity of inflammatory changes in the CNS. We showed that all three chemokines are upregulated simultaneously with symptom onset of acute EAE, and that chemokine expression correlates with the intensity of inflammation in the CNS. This consistent relationship supports the hypothesis that chemokines are relevant to leukocyte accumulation in CNS parenchyma.
Collapse
Affiliation(s)
- A R Glabinski
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, Ohio, USA.
| | | | | |
Collapse
|
42
|
Simpson JE, Newcombe J, Cuzner ML, Woodroofe MN. Expression of monocyte chemoattractant protein-1 and other beta-chemokines by resident glia and inflammatory cells in multiple sclerosis lesions. J Neuroimmunol 1998; 84:238-49. [PMID: 9628469 DOI: 10.1016/s0165-5728(97)00208-7] [Citation(s) in RCA: 314] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Beta-chemokines induce the directional migration of monocytes and T lymphocytes and are thus associated with chronic inflammation. Using immunocytochemistry and in situ hybridisation (ISH) techniques, we have examined the expression of the beta-chemokines monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES (regulated upon activation, normal T cell expressed and secreted) in post-mortem human brain from multiple sclerosis (MS) cases, at different stages of lesion development. In actively demyelinating MS plaques RANTES expression was restricted to the blood vessel endothelium, perivascular cells and surrounding astrocytes, suggesting a role in the recruitment of inflammatory cells from the circulation. MCP-1 was expressed by astrocytes and macrophages within acute MS lesions, but was restricted to reactive astrocytes in the parenchyma surrounding the lesion. MIP-1alpha was expressed by astrocytes and macrophages within the plaque, while MIP-1beta was expressed by macrophages and microglia within the lesion, and by microglia in surrounding white matter. Glial cells may be stimulated to produce chemokines and continue the local inflammatory response by forming chemotactic gradients to attract T cells and mononuclear phagocytes from the circulation and surrounding tissue.
Collapse
Affiliation(s)
- J E Simpson
- Department of Biomedical Sciences, Sheffield Hallam University, City Campus, South Yorkshire, UK
| | | | | | | |
Collapse
|
43
|
Abstract
Hematogenous leukocytes infiltrate the CNS after inflammatory stimuli, including infection, mechanical trauma and excitotoxic neuronal necrosis. However,the role of leukocytic inflammation in promoting or hindering tissue repair is poorly understood. Identification of signals that lead to leukocyte recruitment and activation is essential for the designing of interventions that modulate inflammation, thus improving neurological outcome. Chemokines are small pleiotropic chemoattractant cytokines whose target specificity suggests an important role in determining the cellular composition of inflammatory infiltrates. Chemokine expression profiles in the CNS during autoimmune and post-traumatic inflammation correlate well with the composition of leukocyte infiltrates, and expression studies in systems such as transgenic mice, suggest that chemokines have potent functional attributes in CNS physiology. We propose that selective chemokine expression by CNS cells is crucial for post-traumatic leukocyte accumulation.
Collapse
Affiliation(s)
- R M Ransohoff
- Dept of Neurology, The Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic Foundation, OH 44195, USA
| | | |
Collapse
|
44
|
Ghirnikar RS, Lee YL, Eng LF. Inflammation in traumatic brain injury: role of cytokines and chemokines. Neurochem Res 1998; 23:329-40. [PMID: 9482245 DOI: 10.1023/a:1022453332560] [Citation(s) in RCA: 205] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A traumatic injury to the adult mammalian central nervous system (CNS), such as a stab wound lesion, results in reactive astrogliosis and the migration of hematogenous cells into the damaged neural tissue. The roles of cytokines and growth factors released locally by the damaged endogenous cells are recognized in controlling the cellular changes that occur following CNS injury. However, the role of chemokines, a novel class of chemoattractant cytokines, is only recently being studied in regulating inflammatory cell invasion in the injured/diseased CNS (1). The mRNAs for several chemokines have been shown to be upregulated in experimental allergic encephalomyelitis (EAE), an inflammatory demyelinating disease of the CNS, but chemokine expression in traumatic brain injury has not been studied in detail. Astrocytes have been demonstrated to participate in numerous processes that occur following injury to the CNS. In particular, astrocytic expression of cytokines and growth factors in the injured CNS has been well reviewed (2). Recently a few studies have detected the presence of chemokines in astrocytes following traumatic brain injury (3,4). These studies have suggested that chemokines may represent a promising target for future therapy of inflammatory conditions. This review summarizes the events that occur in traumatic brain injury and discusses the roles of resident and non-resident cells in the expression of growth factors, cytokines and chemokines in the injured CNS.
Collapse
|
45
|
Martiney JA, Cuff C, Litwak M, Berman J, Brosnan CF. Cytokine-induced inflammation in the central nervous system revisited. Neurochem Res 1998; 23:349-59. [PMID: 9482247 DOI: 10.1023/a:1022457500700] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cytokines play an essential role as mediators of the immune response. They usually function as part of a network of interactive signals that either activate, enhance, or inhibit the ensuing reaction. An important contribution of this cytokine cascade is the induction of an inflammatory response that recruits and activates subsets of leukocytes that function as effector cells in the response to the sensitizing antigen. Proinflammatory cytokines activate endothelial cells (EC) to express adhesion molecules and induce the release of members of the chemokine family, thus focusing and directing the inflammatory response to sites of antigen recognition. However, the vasculature of the central nervous system (CNS) is highly specialized and restricts the access of components of the immune system to the CNS compartment. In this review, we address the question as to whether endothelial cells in the CNS respond differently to specific cytokines known to induce either a proinflammatory effect or a regulatory effect in systemic vascular beds.
Collapse
Affiliation(s)
- J A Martiney
- Laboratory of Microbial Pathogenesis, The Picower Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | | | | | |
Collapse
|
46
|
Ehrlich LC, Hu S, Sheng WS, Sutton RL, Rockswold GL, Peterson PK, Chao CC. Cytokine Regulation of Human Microglial Cell IL-8 Production. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.4.1944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
IL-8 involvement in neutrophil activation and chemotaxis may be important in inflammatory responses within the central nervous system, secondary to meningitis, encephalitis, and traumatic injury. The source of IL-8 within the brain during these inflammatory processes, however, is unknown. To explore the role of microglia in the production of IL-8, human fetal microglia, which are the resident macrophages of the brain, were treated with LPS and pro- and anti-inflammatory cytokines to determine their effects on IL-8 production. We found that IL-8 protein levels increased in response to LPS or IL-1β, or to TNF-α, which also corresponded to elevated IL-8 mRNA levels by RT-PCR. Pretreatment with IL-4, IL-10, or TGF-β1 potently inhibited the stimulatory effects of these proinflammatory agents. These findings indicate that human microglia synthesize IL-8 in response to proinflammatory stimuli, and that anti-inflammatory cytokines down-regulate the production of this chemokine. These results may have important therapeutic implications for certain central nervous system insults involving inflammation.
Collapse
Affiliation(s)
| | - Shuxian Hu
- *Neuroimmunobiology and Host Defense Laboratory, and
- ‡University of Minnesota Medical School, Minneapolis, MN 55404
| | - Wen S. Sheng
- *Neuroimmunobiology and Host Defense Laboratory, and
| | - Richard L. Sutton
- †Neurotrauma Research Laboratory, Minneapolis Medical Research Foundation, and
- ‡University of Minnesota Medical School, Minneapolis, MN 55404
| | - Gaylan L. Rockswold
- †Neurotrauma Research Laboratory, Minneapolis Medical Research Foundation, and
- ‡University of Minnesota Medical School, Minneapolis, MN 55404
| | - Phillip K. Peterson
- *Neuroimmunobiology and Host Defense Laboratory, and
- ‡University of Minnesota Medical School, Minneapolis, MN 55404
| | - Chun C. Chao
- *Neuroimmunobiology and Host Defense Laboratory, and
- ‡University of Minnesota Medical School, Minneapolis, MN 55404
| |
Collapse
|
47
|
Lane TE, Asensio VC, Yu N, Paoletti AD, Campbell IL, Buchmeier MJ. Dynamic Regulation of α- and β-Chemokine Expression in the Central Nervous System During Mouse Hepatitis Virus-Induced Demyelinating Disease. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.2.970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Infection of C57BL/6 mice with the V5A13.1 strain of mouse hepatitis virus (MHV-V5A13.1) results in an acute encephalomyelitis and chronic demyelinating disease with features similar to the human demyelinating disease multiple sclerosis. Chemokines are a family of proinflammatory cytokines associated with inflammatory pathology in various diseases. The kinetics and histologic localization of chemokine production in the central nervous system of MHV-infected mice were examined to identify chemokines that contribute to inflammation and demyelination. Transcripts for the chemokines cytokine-response gene-2 (CRG-2), regulated on activation, normal T cell expressed and secreted (RANTES), macrophage-chemoattractant protein-1 and protein-3 (MCP-1, MCP-3), macrophage-inflammatory protein-1β (MIP-1β), and MIP-2 were detected in the brains of MHV-infected mice at 3 days postinfection (p.i.), and these transcripts were increased markedly in brains and spinal cords at day 7 p.i., which coincides with the occurrence of acute viral encephalomyelitis. By day 35 p.i., RANTES, CRG-2, and MIP-1β were detected in brains and spinal cords of mice with chronic demyelination. CRG-2 mRNA expression colocalized with viral RNA and was associated with demyelinating lesions. Astrocytes were the predominant cell type expressing CRG-2 mRNA. These observations suggest a role for chemokines, notably CRG-2, in the initiation and maintenance of an inflammatory response following infection with MHV, which is important in contributing to demyelination.
Collapse
Affiliation(s)
- Thomas E. Lane
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Valérie C. Asensio
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Naichen Yu
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Alyssa D. Paoletti
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Iain L. Campbell
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Michael J. Buchmeier
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
48
|
Stuve O, Chabot S, Jung SS, Williams G, Yong VW. Chemokine-enhanced migration of human peripheral blood mononuclear cells is antagonized by interferon beta-1b through an effect on matrix metalloproteinase-9. J Neuroimmunol 1997; 80:38-46. [PMID: 9413258 DOI: 10.1016/s0165-5728(97)00134-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The increased migration of peripheral blood mononuclear cells (PBMNCs) across a fibronectin (FN) matrix in response to the chemokines RANTES, MIP-1 alpha and MCP-1 is antagonized by interferon-beta-1b (IFN beta-1b). MCP-1 treatment of PBMNCs elevates their mRNA level and secretion of a matrix degrading enzyme, matrix metalloproteinase (MMP)-9, which is abrogated by IFN beta-1b. The clinical benefits of IFN beta-1b treatment in multiple sclerosis patients may in part be a result of this drug's ability to decrease the migration of PBMNCs in spite of a chemotactic gradient. Furthermore, the elevation of MMP-9 production by PBMNCs may be an important mechanism of action of chemokines.
Collapse
Affiliation(s)
- O Stuve
- Department of Neurology and Neurosurgery, McGill University, Montreal, Que., Canada
| | | | | | | | | |
Collapse
|
49
|
Abstract
Elevated levels of chemokines have been observed in various diseases of the CNS. Little is known, however, about how these chemokines affect parenchymal cells of the CNS. The current studies examine astrocyte chemotaxis to the mouse chemokine macrophage inflammatory protein-1alpha (MIP-1alpha). Murine astrocytes demonstrate directed migration along a chemical gradient in response to 10(-10)-10(-8) M MIP-1alpha. Peak chemotactic responses are noted at 10(-9) M. MIP-1alpha-induced astrocyte migration is specifically inhibitable with pertussis toxin, suggesting a role for Galphai proteins in the signaling process. RT-PCR and in situ hybridization were used to identify expression of the murine CCR1 MIP-1alpha receptor on astrocytes. Astrocytes contain mRNA for CCR1, but messages for CCR4 and the orphan chemokine receptor MIP-1alphaR-like#1 were not detected. The combined results suggest that a functional chemokine receptor is expressed on resident cells of the CNS. We speculate that the interactions of chemokines with astrocytes are involved in inflammatory reactions of the CNS.
Collapse
|
50
|
Abstract
A traumatic injury to the adult mammalian central nervous system (CNS) results in reactive astrogliosis and the migration of hematogenous cells into the damaged neural tissue. Chemokines, a novel class of chemoattractant cytokines, are now being recognized as mediators of the inflammatory changes that occur following injury. The expression of MCP-1 (macrophage chemotactic peptide-1), a member of the beta family of chemokines, has recently been demonstrated in trauma in the rat brain (Berman et al.: J Immunol 156:3017-3023, 1996). Using a stab wound model for mechanical injury, we studied the expression of two other beta chemokines: RANTES (Regulated on Activation, Normal T cell Expressed and Secreted) and MIP-1 beta (macrophage inflammatory protein-1 beta) in the rat brain. The stab wound injury was characterized by widespread gliosis and infiltration of hematogenous cells. Immunohistochemical staining revealed the presence of RANTES and MIP-1 beta in the injured brain. RANTES and MIP-1 beta were both diffusely expressed in the necrotic tissue and were detected as early as 1 day post-injury (dpi). Double-labeling studies showed that MIP-1 beta, but not RANTES, was expressed by reactive astrocytes near the lesion site. In addition, MIP-1 beta staining was also detected on macrophages at the site of injury. The initial expression of the chemokines closely correlated with the appearance of inflammatory cells in the injured CNS, suggesting that RANTES and MIP-1 beta may play a role in the inflammatory events of traumatic brain injury. This study also demonstrates for the first time MIP-1 beta expression in reactive astrocytes following trauma to the rat CNS.
Collapse
Affiliation(s)
- R S Ghirnikar
- Department of Pathology, VAPA Health Care System, Palo Alto, CA 94304, USA
| | | | | | | |
Collapse
|