1
|
Mechanisms of Thrombosis and Thrombolysis. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
2
|
Plasminogen Activators in Neurovascular and Neurodegenerative Disorders. Int J Mol Sci 2021; 22:ijms22094380. [PMID: 33922229 PMCID: PMC8122722 DOI: 10.3390/ijms22094380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
The neurovascular unit (NVU) is a dynamic structure assembled by endothelial cells surrounded by a basement membrane, pericytes, astrocytes, microglia and neurons. A carefully coordinated interplay between these cellular and non-cellular components is required to maintain normal neuronal function, and in line with these observations, a growing body of evidence has linked NVU dysfunction to neurodegeneration. Plasminogen activators catalyze the conversion of the zymogen plasminogen into the two-chain protease plasmin, which in turn triggers a plethora of physiological events including wound healing, angiogenesis, cell migration and inflammation. The last four decades of research have revealed that the two mammalian plasminogen activators, tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA), are pivotal regulators of NVU function during physiological and pathological conditions. Here, we will review the most relevant data on their expression and function in the NVU and their role in neurovascular and neurodegenerative disorders.
Collapse
|
3
|
Mechanisms of Thrombosis and Thrombolysis. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
4
|
The plasminogen activation system in neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2015; 1862:395-402. [PMID: 26493446 DOI: 10.1016/j.bbadis.2015.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/08/2015] [Accepted: 10/15/2015] [Indexed: 01/30/2023]
Abstract
The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
5
|
Woo JH, Lee JH, Kim H, Park SJ, Joe EH, Jou I. Control of Inflammatory Responses: a New Paradigm for the Treatment of Chronic Neuronal Diseases. Exp Neurobiol 2015; 24:95-102. [PMID: 26113788 PMCID: PMC4479815 DOI: 10.5607/en.2015.24.2.95] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/30/2015] [Accepted: 04/30/2015] [Indexed: 12/26/2022] Open
Abstract
The term 'inflammation' was first introduced by Celsus almost 2000 years ago. Biological and medical researchers have shown increasing interest in inflammation over the past few decades, in part due to the emerging burden of chronic and degenerative diseases resulting from the increased longevity that has arisen thanks to modern medicine. Inflammation is believed to play critical roles in the pathogenesis of degenerative brain diseases, including Alzheimer's disease and Parkinson's disease. Accordingly, researchers have sought to combat such diseases by controlling inflammatory responses. In this review, we describe the endogenous inflammatory stimulators and signaling pathways in the brain. In particular, our group has focused on the JAK-STAT pathway, identifying anti-inflammatory targets and testing the effects of various anti-inflammatory drugs. This work has shown that the JAK-STAT pathway and its downstream are negatively regulated by phosphatases (SHP2 and MKP-1), inhibitory proteins (SOCS1 and SOCS3) and a nuclear receptor (LXR). These negative regulators are controlled at various levels (e.g. transcriptional, post-transcriptional and post-translational). Future study of these proteins could facilitate the manipulation of the inflammatory response, which plays ubiquitous, diverse and ambivalent roles under physiological and pathological conditions.
Collapse
Affiliation(s)
- Joo Hong Woo
- Department of Pharmacology, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Korea
| | - Jee Hoon Lee
- Department of Pharmacology, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Korea
| | - Hyunmi Kim
- Department of Pharmacology, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Korea. ; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 443-721, Korea
| | - Soo Jung Park
- Department of Pharmacology, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Korea
| | - Eun-Hye Joe
- Department of Pharmacology, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Korea. ; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 443-721, Korea
| | - Ilo Jou
- Department of Pharmacology, and Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Korea. ; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 443-721, Korea
| |
Collapse
|
6
|
Mechanisms of Thrombosis and Thrombolysis. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
The Cerebral Microvasculature and Responses to Ischemia. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
8
|
Yin J, Sakamoto K, Zhang H, Ito Z, Imagama S, Kishida S, Natori T, Sawada M, Matsuyama Y, Kadomatsu K. Transforming growth factor-beta1 upregulates keratan sulfate and chondroitin sulfate biosynthesis in microglias after brain injury. Brain Res 2009; 1263:10-22. [PMID: 19368826 DOI: 10.1016/j.brainres.2009.01.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 01/07/2009] [Accepted: 01/22/2009] [Indexed: 11/28/2022]
Abstract
After injury to the adult central nervous system, levels of extracellular matrix molecules increase at the injury site and may inhibit the repair of injured axons. Among these molecules, the importance of proteoglycans, particularly their chondroitin sulfate chains, has been highlighted. We have recently reported that keratan sulfate-deficient mice show better axonal regeneration after injury. Here, we investigated the regulation of keratan sulfate and chondroitin sulfate biosynthesis after neuronal injuries. Several key enzymes required for glycosaminoglycan biosynthesis (beta3GlcNAcT-7 and GlcNAc6ST-1 for keratan sulfate; CS synthase-1 and C6ST-1 for chondroitin sulfate) were expressed at significantly higher levels in the lesion 7 days after a knife-cut injury was made to the cerebral cortex in adult mice. These increases were accompanied by increased expression of TGF-beta(1) and bFGF. Since microglias at the injury sites expressed both keratan sulfate and chondroitin sulfate, the effects of these cytokines were examined in microglias. TGF-beta(1) induced the expression of the above-named enzymes in microglias, and consequently induced keratan sulfate and chondroitin sulfate biosynthesis as well as the expression of the chondroitin/keratan sulfate proteoglycan aggrecan in these cells. TGF-beta(1) also induced bFGF expression in microglias. bFGF in turn induced TGF-beta(1) expression in astrocytes. Astrocyte-conditioned medium following bFGF stimulation indeed induced keratan sulfate and chondroitin sulfate production in microglias. This production was blocked by TGF-beta(1)-neutralizing antibody. Taken together, our data indicate that the biosyntheses of keratan sulfate and chondroitin sulfate are upregulated in common by TGF-beta(1) in microglias after neuronal injuries.
Collapse
Affiliation(s)
- Jiarong Yin
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Samson AL, Nevin ST, Croucher D, Niego B, Daniel PB, Weiss TW, Moreno E, Monard D, Lawrence DA, Medcalf RL. Tissue-type plasminogen activator requires a co-receptor to enhance NMDA receptor function. J Neurochem 2008; 107:1091-101. [PMID: 18796005 PMCID: PMC3198853 DOI: 10.1111/j.1471-4159.2008.05687.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glutamate is the main excitatory neurotransmitter of the CNS. Tissue-type plasminogen activator (tPA) is recognized as a modulator of glutamatergic neurotransmission. This attribute is exemplified by its ability to potentiate calcium signaling following activation of the glutamate-binding NMDA receptor (NMDAR). It has been hypothesized that tPA can directly cleave the NR1 subunit of the NMDAR and thereby potentiate NMDA-induced calcium influx. In contrast, here we show that this increase in NMDAR signaling requires tPA to be proteolytically active, but does not involve cleavage of the NR1 subunit or plasminogen. Rather, we demonstrate that enhancement of NMDAR function by tPA is mediated by a member of the low-density lipoprotein receptor (LDLR) family. Hence, this study proposes a novel functional relationship between tPA, the NMDAR, a LDLR and an unknown substrate which we suspect to be a serpin. Interestingly, whilst tPA alone failed to cleave NR1, cell-surface NMDARs did serve as an efficient and discrete proteolytic target for plasmin. Hence, plasmin and tPA can affect the NMDAR via distinct avenues. Altogether, we find that plasmin directly proteolyses the NMDAR whilst tPA functions as an indirect modulator of NMDA-induced events via LDLR engagement.
Collapse
Affiliation(s)
- Andre L. Samson
- Australian Centre for Blood Diseases, Monash University, AMREP, Melbourne, Australia
| | - Simon T. Nevin
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - David Croucher
- School of Biological Sciences, University of Wollongong, NSW, Australia
| | - Be’eri Niego
- Australian Centre for Blood Diseases, Monash University, AMREP, Melbourne, Australia
| | - Philip B. Daniel
- Australian Centre for Blood Diseases, Monash University, AMREP, Melbourne, Australia
| | - Thomas W. Weiss
- Australian Centre for Blood Diseases, Monash University, AMREP, Melbourne, Australia
| | - Eliza Moreno
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Denis Monard
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | | | - Robert L. Medcalf
- Australian Centre for Blood Diseases, Monash University, AMREP, Melbourne, Australia
| |
Collapse
|
10
|
Differential Regulation of Matrix Metalloproteinase-9 and Tissue Plasminogen Activator Activity by the Cyclic-AMP System in Lipopolysaccharide-stimulated Rat Primary Astrocytes. Neurochem Res 2008; 33:2324-34. [DOI: 10.1007/s11064-008-9737-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Accepted: 04/29/2008] [Indexed: 11/26/2022]
|
11
|
Massey JM, Amps J, Viapiano MS, Matthews RT, Wagoner MR, Whitaker CM, Alilain W, Yonkof AL, Khalyfa A, Cooper NGF, Silver J, Onifer SM. Increased chondroitin sulfate proteoglycan expression in denervated brainstem targets following spinal cord injury creates a barrier to axonal regeneration overcome by chondroitinase ABC and neurotrophin-3. Exp Neurol 2007; 209:426-45. [PMID: 17540369 PMCID: PMC2270474 DOI: 10.1016/j.expneurol.2007.03.029] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2007] [Revised: 03/24/2007] [Accepted: 03/27/2007] [Indexed: 01/11/2023]
Abstract
Increased chondroitin sulfate proteoglycan (CSPG) expression in the vicinity of a spinal cord injury (SCI) is a primary participant in axonal regeneration failure. However, the presence of similar increases of CSPG expression in denervated synaptic targets well away from the primary lesion and the subsequent impact on regenerating axons attempting to approach deafferented neurons have not been studied. Constitutively expressed CSPGs within the extracellular matrix and perineuronal nets of the adult rat dorsal column nuclei (DCN) were characterized using real-time PCR, Western blot analysis and immunohistochemistry. We show for the first time that by 2 days and through 3 weeks following SCI, the levels of NG2, neurocan and brevican associated with reactive glia throughout the DCN were dramatically increased throughout the DCN despite being well beyond areas of trauma-induced blood brain barrier breakdown. Importantly, regenerating axons from adult sensory neurons microtransplanted 2 weeks following SCI between the injury site and the DCN were able to regenerate rapidly within white matter (as shown previously by Davies et al. [Davies, S.J., Goucher, D.R., Doller, C., Silver, J., 1999. Robust regeneration of adult sensory axons in degenerating white matter of the adult rat spinal cord. J. Neurosci. 19, 5810-5822]) but were unable to enter the denervated DCN. Application of chondroitinase ABC or neurotrophin-3-expressing lentivirus in the DCN partially overcame this inhibition. When the treatments were combined, entrance by regenerating axons into the DCN was significantly augmented. These results demonstrate both an additional challenge and potential treatment strategy for successful functional pathway reconstruction after SCI.
Collapse
Affiliation(s)
- James M. Massey
- M.D./Ph.D. Program, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Jeremy Amps
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Mariano S. Viapiano
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520
| | - Russell. T. Matthews
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520
| | - Michelle R. Wagoner
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Christopher M. Whitaker
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Warren Alilain
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Alicia L. Yonkof
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
| | - Abdelnaby Khalyfa
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Nigel G. F. Cooper
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106
- Address for correspondence, proofs, and reprint requests: Stephen M. Onifer, Ph.D., Spinal Cord and Brain Injury Research Center, University of Kentucky, BBSRB B365, 741 South Limestone Street, Lexington, KY, 40536-0509, U.S.A., TELEPHONE: (859) 323-5226, FAX: (859) 257-5737, EMAIL:
| | - Stephen M. Onifer
- Department of Anatomical Sciences & Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Neurological Surgery, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
- Department of Kentucky Spinal Cord Injury Research Center, School of Medicine, University of Louisville, Louisville, Kentucky, 40292
| |
Collapse
|
12
|
Kucharova K, Lukacova N, Pavel J, Radonak J, Hefferan MP, Kolesar D, Kolesarova M, Marsala M, Marsala J. Spatiotemporal alterations of the NO/NOS neuronal pools following transient abdominal aorta occlusion: morphological and biochemical studies in the rabbit. Cell Mol Neurobiol 2006; 26:1295-310. [PMID: 16786431 DOI: 10.1007/s10571-006-9089-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2005] [Accepted: 05/09/2006] [Indexed: 10/24/2022]
Abstract
1. Brief interruption of spinal cord blood flow resulting from transient abdominal aortic occlusion may lead to degeneration of specific spinal cord neurons and to irreversible loss of neurological function. The alteration of nitric oxide/nitric oxide synthase (NO/NOS) pool occurring after ischemic insult may play a protective or destructive role in neuronal survival of affected spinal cord segments. 2. In the present study, the spatiotemporal changes of NOS following transient ischemia were evaluated by investigating neuronal NOS immunoreactivity (nNOS-IR), reduced nicotinamide adenine dinucleotide phosphate diaphorase (NADPHd) histochemistry, and calcium-dependent NOS (cNOS) conversion of [(3)H] l-arginine to [(3)H] l-citrulline. 3. The greatest levels of these enzymes and activities were detected in the dorsal horn, which appeared to be most resistant to ischemia. In that area, the first significant increase in NADPHd staining and cNOS catalytic activity was found immediately after a 15-min ischemic insult. 4. Increases in the ventral horn were observed later (i.e., after a 24-h reperfusion period). While the most intense increase in nNOS-IR was detected in surviving motoneurons of animals with a shorter ischemic insult (13 min), the greatest increase of cNOS catalytic activity and NADPHd staining of the endothelial cells was found after stronger insult (15 min). 5. Given that the highest levels of nNOS, NADPHd, and cNOS were found in the ischemia-resistant dorsal horn, and nNOS-IR in surviving motoneurons, it is possible that NO production may play a neuroprotective role in ischemic/reperfusion injury.
Collapse
Affiliation(s)
- K Kucharova
- Institute of Neurobiology, Slovak Academy of Sciences, Kosice, Slovak Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Davies JE, Tang X, Bournat JC, Davies SJA. Decorin promotes plasminogen/plasmin expression within acute spinal cord injuries and by adult microglia in vitro. J Neurotrauma 2006; 23:397-408. [PMID: 16629625 DOI: 10.1089/neu.2006.23.397] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spinal cord scar tissue presents a combined physical and molecular barrier to axon regeneration. Theoretically, spinal cord injuries (SCIs) can be rendered more permissive to axon growth by either suppressing synthesis of misaligned, fibrotic scar tissue and associated axon growth inhibitors, or enzymatically degrading them. We have previously shown that acute infusion of human recombinant decorin core protein into discreet stab injuries of the rat dorsal column pathways effected a major suppression of inflammation, astrogliosis, and multiple axon growth inhibitory chondroitin sulfate proteoglycans, which combined to promote rapid axon growth across the injury site. The high efficiency of chondroitin sulfate proteoglycan (CSPG) core protein suppression (approximately 90%) suggested that decorin may promote CSPG degradation in addition to suppressing CSPG synthesis. As the serine protease plasmin can degrade axon growth inhibitory CSPGs (neurocan and phosphacan) and its zymogen, plasmininogen is synthesized by microglia, we have investigated whether decorin treatment of acute SCIs and cultured adult spinal cord microglia can increase plasminogen/ plasmin synthesis. Infusion of hr-decorin over the first 8 days post-SCI induced 10- and 17-fold increases in plasminogen and plasmin protein levels, respectively, within sites of injury and a threefold increase in microglial plasminogen mRNA in vitro. In addition to potentially degrading multiple axon growth inhibitory components of the glial scar, plasmin is known to play major roles in activating neurotrophins and promoting central nervous system (CNS) plasticity. The wider implications of decorin induction of plasmin in the injured spinal cord for axon regeneration, and recovery of function at acute and chronic time points post-SCI are reviewed.
Collapse
Affiliation(s)
- Jeannette E Davies
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
14
|
Kadiu I, Glanzer JG, Kipnis J, Gendelman HE, Thomas MP. Mononuclear phagocytes in the pathogenesis of neurodegenerative diseases. Neurotox Res 2006; 8:25-50. [PMID: 16260384 DOI: 10.1007/bf03033818] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Brain mononuclear phagocytes (MP, bone marrow monocyte-derived macrophages, perivascular macrophages, and microglia) function to protect the nervous system by acting as debris scavengers, killers of microbial pathogens, and regulators of immune responses. MP are activated by a variety of environmental cues and such inflammatory responses elicit cell injury and death in the nervous system. MP immunoregulatory responses include secretion of neurotoxic factors, mobilization of adaptive immunity, and cell chemotaxis. This incites tissue remodelling and blood-brain barrier dysfunction. As disease progresses, MP secretions engage neighboring cells in a vicious cycle of autocrine and paracrine amplification of inflammation leading to tissue injury and ultimately destruction. Such pathogenic processes tilt the balance between the relative production of neurotrophic and neurotoxic factors and to disease progression. The ultimate effects that brain MP play in disease revolves "principally" around their roles in neurodegeneration. Importantly, common functions of brain MP in neuroimmunity link highly divergent diseases (for example, human immunodeficiency virus type-one associated dementia, Alzheimer's disease and Parkinson's disease). Research into this process from our own laboratories and those of others seek to harness MP inflammatory processes with the intent of developing therapeutic interventions that block neurodegenerative processes and improve the quality of life in affected people.
Collapse
Affiliation(s)
- I Kadiu
- Laboratory of Neuroregeneration, Department of Pharmacology and Experimental Neuroscience, Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | | | |
Collapse
|
15
|
von Bernhardi R, Eugenín J. Microglial reactivity to β-amyloid is modulated by astrocytes and proinflammatory factors. Brain Res 2004; 1025:186-93. [PMID: 15464759 DOI: 10.1016/j.brainres.2004.07.084] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2004] [Indexed: 12/24/2022]
Abstract
The brains of Alzheimer's disease (AD) patients present activated glial cells, amyloid plaques and dystrophic neurites. The core of amyloid plaques is composed of aggregated amyloid peptide (Abeta), a peptide known to activate glial cells and to have neurotoxic effects. We evaluated the capability of glial cells to mediate Abeta(1-42) cytotoxicity in hippocampal cultures. Conditioned media obtained from microglial cultures exposed to Abeta induced apoptosis of hippocampal cells. This pro-apoptotic effect was not observed in hippocampal cultures exposed to conditioned media obtained from mixed glial (astrocytes and microglia) cultures that had been exposed to Abeta. Microglia exposed to Abeta responded with reactive morphological changes, induction of iNOS, elevated nitric oxide production and decreased reductive metabolism. All these responses were attenuated by the presence of astrocytes. This astrocyte modulation was however, not observed when glial cells were exposed to proinflammatory factors (LPS+Interferon-gamma) alone or in combination with Abeta. Our results suggest that astrocytes and proinflammatory molecules are determining factors in the response of microglia to Abeta.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile.
| | | |
Collapse
|
16
|
Hamann G, del Zoppo GJ. The Cerebral Microvasculature and Responses to Ischemia. Stroke 2004. [DOI: 10.1016/b0-44-306600-0/50045-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
17
|
del Zoppo GJ, Kalafut M. Mechanisms of Thrombosis and thrombolysis. Stroke 2004. [DOI: 10.1016/b0-44-306600-0/50046-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
Fu KY, Light AR, Maixner W. Long-lasting inflammation and long-term hyperalgesia after subcutaneous formalin injection into the rat hindpaw. THE JOURNAL OF PAIN 2003; 2:2-11. [PMID: 14622781 DOI: 10.1054/jpai.2001.9804] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Subcutaneous formalin injection is widely used as a nociceptive stimulus in the rat. This procedure evokes overt behaviors that last about 90 minutes. However, little is known about the duration of paw inflammation and alterations in pain sensitivity to noxious stimuli after 2 hours. We studied the nociceptive responses to thermal and mechanical stimuli 2 hours to 4 weeks after formalin injection into the dorsal or plantar side of the hindpaw. Thirty-two adult male Sprague-Dawley rats were divided into 3 groups: In group I, 50 microL of 5% formalin was injected into the plantar side (n = 12); in group II, 50 microL of 5% formalin was injected into the dorsal side (n = 12); in group III, 50 microL saline was injected into the dorsal or plantar side of the hindpaw (n = 8). Nociceptive responses to thermal and mechanical stimuli applied to the dorsal or plantar surfaces of the injected and the contralateral hindpaws were recorded. The injection of formalin into the rat's hindpaw produced a hypoalgesic region around the injection site. In contrast, hyperalgesic responses to thermal and mechanical stimulation were induced on the opposite surface of the injected hindpaw as well as in the contralateral noninjected hindpaw. The hyperalgesic responses, which were observed 2 hours after formalin administration, were enhanced 1 to 3 days after injection and lasted 3 to 4 weeks. These results suggest that peripheral inflammation after subcutaneous formalin injection produces a long-lasting sensitization. Possible mechanisms for these changes in nociception are discussed.
Collapse
Affiliation(s)
- K Y Fu
- Department of Cell and Molecular Physiology, University of North Carolina-Chapel Hill, 27599-7455, USA.
| | | | | |
Collapse
|
19
|
Abstract
Cerebral microvessels have a unique ultrastructure form, which allows for the close relationship of the endothelium and blood elements to the neurons they serve, via intervening astrocytes. To focal ischemia, the cerebral microvasculature rapidly displays multiple dynamic responses. Immediate events include breakdown of the primary endothelial cell permeability barrier, with transudation of plasma, expression of endothelial cell-leukocyte adhesion receptors, loss of endothelial cell and astrocyte integrin receptors, loss of their matrix ligands, expression of members of several matrix-degrading protease families, and the appearance of receptors associated with angiogenesis and neovascularization. These events occur pari passu with neuron injury. Alterations in the microvessel matrix after the onset of ischemia also suggest links to changes in nonvascular cell viability. Microvascular obstruction within the ischemic territory occurs after occlusion and reperfusion of the feeding arteries ("focal no-reflow" phenomenon). This can result from extrinsic compression and intravascular events, including leukocyte(-platelet) adhesion, platelet-fibrin interactions, and activation of coagulation. All of these events occur in microvessels heterogeneously distributed within the ischemic core. The panorama of acute microvessel responses to focal cerebral ischemia provide opportunities to understand interrelationships between neurons and their microvascular supply and changes that underlie a number of central nervous system neurodegenerative disorders.
Collapse
Affiliation(s)
- Gregory J del Zoppo
- of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MEM 132, La Jolla, CA 92037, U.S.A.
| | | |
Collapse
|
20
|
Yang MS, Park EJ, Sohn S, Kwon HJ, Shin WH, Pyo HK, Jin B, Choi KS, Jou I, Joe EH. Interleukin-13 and -4 induce death of activated microglia. Glia 2002; 38:273-80. [PMID: 12007140 DOI: 10.1002/glia.10057] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
When the brain suffers injury, microglia migrate to the damaged sites and become activated. These activated microglia are not detected several days later and the mechanisms underlying their disappearance are not well characterized. In this study, we demonstrate that interleukin (IL)-13, an anti-inflammatory cytokine, selectively induces cell death of activated microglia in vitro. Cell death was detected 4 days after the coaddition of IL-13 with any one of the microglial activators, lipopolysaccharide (LPS), ganglioside, or thrombin. This cell death occurred in a time-dependent manner. LPS, ganglioside, thrombin, or IL-13 alone did not induce cell death. Among anti-inflammatory cytokines, IL-4 mimicked the effect of IL-13, while TGF-beta did not. Cells treated with IL-13 plus LPS, or IL-13 plus ganglioside, showed the characteristics of apoptosis when analyzed by electron microscopy and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining. Electron micrographs also showed microglia engulfing neighboring dead cells. We propose that IL-13 and IL-4 induce death of activated microglia, and that this process is important for prevention of chronic inflammation that can cause tissue damage.
Collapse
Affiliation(s)
- Myung-Soon Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Studies of extracellular matrix (ECM) biology in the nervous system have mainly focused on laminin, fibronectin and tenascin-R, proteins that are present during nervous system development and normal function. However, during disease, fibrin, which physiologically is not present in the nervous tissue, is detected at nervous tissue lesions. This review summarizes evidence that correlates fibrin deposition with neuropathology and presents recent findings on cellular mechanisms and intracellular signaling pathways regulated by fibrin that might contribute to nervous system disease.
Collapse
Affiliation(s)
- Katerina Akassoglou
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10021, USA
| | | |
Collapse
|
22
|
Teesalu T, Hinkkanen AE, Vaheri A. Coordinated induction of extracellular proteolysis systems during experimental autoimmune encephalomyelitis in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:2227-37. [PMID: 11733372 PMCID: PMC1850601 DOI: 10.1016/s0002-9440(10)63073-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/12/2001] [Indexed: 02/02/2023]
Abstract
Plasminogen activators (PAs) and matrix metalloproteinases (MMPs) are considered to play an important role in the pathogenesis of multiple sclerosis. Experimental autoimmune encephalomyelitis (EAE) is widely used as an animal model of multiple sclerosis. Whereas several studies have addressed the expression of various MMPs and their inhibitors in the pathogenesis of EAE, the expression of the molecules of the PA system during EAE has not been reported previously. The present study was undertaken to investigate the expression of the molecules of the PA system (tPA, uPA, PAI-1, uPAR, LRP), as well as several members of the MMP family and their inhibitors in the course of actively induced EAE in BALB/c mice. During clinical EAE, the PA system was up-regulated in the central nervous system at several levels. Induction of expression of tPA and PAI-1 transcripts was detected in activated astrocytes in the white matter. Inflammatory cells expressed uPA receptor, uPAR. In situ zymography demonstrated the presence of increased tPA and uPA activities in the areas of the inflammatory damage. Accumulation of fibrin, fibronectin, and vitronectin immunoreactivity was seen in perivascular matrices of symptomatic animals. In addition, transcription of MT1-MMP and metalloelastase (in inflammatory cells), and TIMP-1 (in activated astrocytes) was induced during EAE. Increased gelatinolytic activity was detected at the sites of inflammatory cell accumulation by in situ zymography of fluorescently labeled gelatin; substrate gel zymography identified the up-regulated gelatinolytic activity as gelatinase B. Overall, our study demonstrates concurrent induction of PA and MMP systems during active EAE, supporting further the concept that the neuroinflammatory damage in EAE involves altered balance between multiple extracellular proteases and their inhibitors.
Collapse
MESH Headings
- Animals
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Electrophoresis, Polyacrylamide Gel
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Extracellular Matrix/metabolism
- Extracellular Matrix Proteins/analysis
- Female
- Fibrin/analysis
- Fibronectins/analysis
- Gene Expression Regulation
- Immunohistochemistry
- In Situ Hybridization
- Male
- Matrix Metalloproteinase 14
- Matrix Metalloproteinases/genetics
- Matrix Metalloproteinases/metabolism
- Matrix Metalloproteinases, Membrane-Associated
- Metalloendopeptidases/genetics
- Metalloendopeptidases/metabolism
- Mice
- Mice, Inbred BALB C
- Plasminogen Activator Inhibitor 1/genetics
- Plasminogen Activator Inhibitor 1/metabolism
- Plasminogen Activators/genetics
- Plasminogen Activators/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Urokinase Plasminogen Activator
- Tissue Inhibitor of Metalloproteinase-1/genetics
- Tissue Inhibitor of Metalloproteinase-1/metabolism
- Tissue Plasminogen Activator/genetics
- Tissue Plasminogen Activator/metabolism
- Vitronectin/analysis
Collapse
Affiliation(s)
- T Teesalu
- Haartman Institute, University of Helsinki, Helsinki, Finland.
| | | | | |
Collapse
|
23
|
Hosomi N, Lucero J, Heo JH, Koziol JA, Copeland BR, del Zoppo GJ. Rapid differential endogenous plasminogen activator expression after acute middle cerebral artery occlusion. Stroke 2001; 32:1341-8. [PMID: 11387497 DOI: 10.1161/01.str.32.6.1341] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE During focal cerebral ischemia, the microvascular matrix (ECM), which participates in microvascular integrity, is degraded and lost when neurons are injured. Loss of microvascular basal lamina antigens coincides with rapid expression of select matrix metalloproteinases (MMPs). Plasminogen activators (PAs) may also play a role in ECM degradation by the generation of plasmin or by MMP activation. METHODS The endogenous expressions of tissue-type plasminogen activator (tPA), urokinase (uPA), and PA inhibitor-1 (PAI-1) were quantified in 10-microm frozen sections from ischemic and matched nonischemic basal ganglia and in the plasma of 34 male healthy nonhuman primates before and after middle cerebral artery occlusion (MCA:O). RESULTS Within the ischemic basal ganglia, tissue uPA activity and antigen increased significantly within 1 hour after MCA:O (2P<0.005). tPA activity transiently decreased 2 hours after MCA:O (2P=0.01) in concert with an increase in PAI-1 antigen (2P=0.001) but otherwise did not change. The transient decrease in free tPA antigen was marked by an increase in the tPA-PAI-1 complex (2P<0.001). No significant relations to neuronal injury or intracerebral hemorrhage were discerned. CONCLUSIONS The rapid increase in endogenous PA activity is mainly due to significant increases in uPA, but not tPA, within the ischemic basal ganglia after MCA:O. This increase and an increase in PAI-1 coincided with latent MMP-2 generation and microvascular ECM degeneration but not neuronal injury.
Collapse
Affiliation(s)
- N Hosomi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
24
|
Hailer NP, Glomsda B, Blaheta RA. Astrocytic factors down-regulate the expression of major histocompatibility complex-class-II and intercellular adhesion molecule-1 on human monocytes. Neurosci Lett 2001; 298:33-6. [PMID: 11154829 DOI: 10.1016/s0304-3940(00)01711-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Several factors contribute to the maintenance of central nervous system immune privilege and astrocytes have been identified as a major source of immunomodulatory cytokines. To investigate whether hematogenous monocytes are immunologically deactivated by astrocyte-derived factors human monocytes were stimulated with lipopolysaccharide or interferon (IFN)-gamma and treated with the supernatant from pure astrocyte cultures, interleukin (IL)-4, IL-10, or with IL-1-receptor antagonist (1L-1-RA). Flow cytometry demonstrated that the supernatant from astrocyte cultures was the most potent agent in reducing the levels of major histocompatibility complex (MHC)-class-II- as well as intercellular adhesion molecule-1-expression, whereas IL-4, IL-10, and IL-1-RA had only marginal effects. The expression of leukocyte function antigen-1 and very late antigen-4 was not modulated by either factor. In conclusion, astrocytes seem to provide soluble factors that have the capacity to deactivate hematogenous monocytes.
Collapse
Affiliation(s)
- N P Hailer
- University Hospital for Orthopaedic Surgery, Friedrichsheim, Marienburgstrasse 2, D-60528 Frankfurt am Main, Germany.
| | | | | |
Collapse
|
25
|
|
26
|
Molina-Holgado F, Toulmond S, Rothwell NJ. Involvement of interleukin-1 in glial responses to lipopolysaccharide: endogenous versus exogenous interleukin-1 actions. J Neuroimmunol 2000; 111:1-9. [PMID: 11063815 DOI: 10.1016/s0165-5728(00)00344-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Interleukin-1beta (IL-1beta) participates in neuroinflammation and neurodegeneration. Its mechanisms of action are not fully understood, but appear to involve complex interactions between neurons and glia. The objective of this study was to determine the involvement of endogenous IL-1beta in inflammatory responses to LPS in cultured mouse glial cells, and compare this to the effects of exogenous IL-1beta. Activation of primary mixed glial cultures by incubation with LPS (1 microgram/ml, 24 h), caused marked (approximately ten-fold) increases in release of NO, twenty-fold increases in PGE(2) and ninety-fold increases of IL-6 release. Incubation with human recombinant IL-1beta (100 ng/ml) also stimulated NO and IL-6 release to a similar extent to LPS, but IL-1beta (1 or 100 ng/ml) caused only modest increases (approximately seven-fold) in PGE(2) release. Co-incubation with IL-1ra inhibited the effects of LPS on NO release (-65%) and IL-6 production (-30%), but failed to reduce PGE(2) release. These results indicate that exogenous IL-1beta induces release of NO, PGE(2) and IL-6 in mixed glial cultures, and that endogenous IL-1beta mediates inflammatory actions of LPS on NO and to a lesser extent IL-6, but not on PGE(2) release in mixed glial cultures. Indeed endogenous IL-1beta appears to inhibit LPS-induced PGE(2) release.
Collapse
Affiliation(s)
- F Molina-Holgado
- School of Biological Sciences 1.124 Stopford Building, The University of Manchester, Oxford Road, M13 9PT, Manchester, UK
| | | | | |
Collapse
|
27
|
Ledeboer A, Brev� JJ, Poole S, Tilders FJ, Van Dam AM. Interleukin-10, interleukin-4, and transforming growth factor-? differentially regulate lipopolysaccharide-induced production of pro-inflammatory cytokines and nitric oxide in co-cultures of rat astroglial and microglial cells. Glia 2000. [DOI: 10.1002/(sici)1098-1136(200004)30:2%3c134::aid-glia3%3e3.0.co;2-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
28
|
Ledeboer A, Brevé JJ, Poole S, Tilders FJ, Van Dam AM. Interleukin-10, interleukin-4, and transforming growth factor-beta differentially regulate lipopolysaccharide-induced production of pro-inflammatory cytokines and nitric oxide in co-cultures of rat astroglial and microglial cells. Glia 2000; 30:134-42. [PMID: 10719355 DOI: 10.1002/(sici)1098-1136(200004)30:2<134::aid-glia3>3.0.co;2-3] [Citation(s) in RCA: 179] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The pro-inflammatory cytokines interleukin-1beta (IL-1beta), IL-6, tumor necrosis factor-alpha (TNF-alpha), and nitric oxide (NO) can be produced by activated glial cells and play a critical role in various neurological diseases. Using primary co-cultures of rat microglial and astroglial cells, we investigated the effects of the anti-inflammatory cytokines transforming growth factor-beta1 (TGF-beta1)/beta2, IL-4, and IL-10 on the production of (pro-) inflammatory mediators after stimulation of the cells with lipopolysaccharide (LPS; 0.1 micrograms/ml, 24 h). IL-10 (10 and 100 ng/ml) and IL-4 (5 and 50 U/ml) suppressed the LPS-induced production of NO, IL-6, and TNF-alpha in a dose-dependent manner, whereas TGF-beta1/beta2 (2 and 20 ng/ml) only suppressed NO production. LPS-induced levels of IL-1beta were suppressed by IL-10, but not by IL-4 and TGF-beta1/beta2. Conversely, co-incubation of the glial cells with LPS and antibodies to TGF-beta1/beta2 selectively enhanced LPS-induced NO production, whereas co-incubation with antibody to IL-10 enhanced LPS-induced production of all pro-inflammatory cytokines and NO. This finding strongly suggests that effective concentrations of TGF-beta1/beta2 and IL-10 are produced by LPS-stimulated glial cell co-cultures. Production of IL-10 in these co-cultures was confirmed by measurement of rat IL-10 by radioimmunoassay. We conclude that anti-inflammatory cytokines affect the production of inflammatory mediators in LPS-activated co-cultures of microglial and astroglial cells differentially.
Collapse
Affiliation(s)
- A Ledeboer
- Department of Pharmacology, Research Institute Neurosciences Free University, Faculty of Medicine, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
29
|
Abstract
Interferon-gamma-inducible 10 kd protein (IP-10) is an ELR (Glu-Leu-Arg)(-) alpha chemokine with known chemotactic effects on T cells and monocytes, as well as anti-viral, anti-angiogenic, and anti-tumor effects. Previous studies have demonstrated that in cultured rat astrocytes and microglia, stimulation with LPS or virus can induce the expression of IP-10. In this study, we determined the pattern of IP-10 gene induction in primary human microglia and astrocytes by cytokines and LPS using ribonuclease protection assay. The expression of IP-10 mRNA was compared with that of other alpha (IL-8) and beta chemokines. The results showed that in human microglia, IP-10 expression was induced equally potently by LPS, IFNbeta or IFNgamma. "Proinflammatory" cytokines IL-1beta or TNFalpha also induced small amounts of IP-10 mRNA. "Anti-inflammatory" cytokines IL-4, IL-10 and TGFbeta were ineffective in inducing IP-10 in microglia. In human astrocytes, induction of IP-10 mRNA by cytokines was similar to that in microglia. LPS, however, was ineffective in inducing IP-10 in human astrocytes. The monocyte chemoattractant beta-chemokine I-309 mRNA was induced in human astrocytes and microglia by IFNbeta or IFNgamma, or by LPS in microglia, showing a tight co-regulation with IP-10 mRNA expression. In contrast to the potent induction of IP-10 and I-309 by IFNs in human glia, the ELR(+) alpha chemokine IL-8 mRNA was induced by IL-1beta and TNFalpha, and to a lesser extent by IFNbeta in microglia. IFNbeta but not IFNgamma was effective in inducing the expression of beta chemokines MIP-1alpha and MIP-1beta in human microglia, with the levels of mRNA similar to those induced by IL-1beta or TNFalpha. Neither MIP-1alpha nor MIP-1beta mRNAs were induced by any stimulation in human astrocytes. The induction of RANTES mRNA in microglia by IFNbeta, IL-1beta or TNFalpha was variable, showing no to low level expression depending on the case, whereas LPS provided a consistent inducing signal. In astrocytes, only cytokine combinations (IFN + IL-1beta) effectively induced the RANTES mRNA. These results demonstrate that distinct sets of chemokine genes are induced in human glial cells by cytokines and interferons. These results may have wide implications for inflammatory, vascular and neoplastic diseases of the CNS.
Collapse
Affiliation(s)
- L L Hua
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
30
|
Tanaka J, Toku K, Sakanaka M, Maeda N. Morphological differentiation of microglial cells in culture: involvement of insoluble factors derived from astrocytes. Neurosci Res 1999; 34:207-15. [PMID: 10576543 DOI: 10.1016/s0168-0102(99)00041-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
It is believed that ramified resting microglial cells in the brain are differentiated from macrophage-like ameboid cells, although the mechanism for the differentiation is not fully understood. In the present study, we investigated whether the differentiation of microglial cells is observable in mixed brain cell culture prepared from newborn rat forebrains. In confluent mixed brain cell culture, both ramified and ameboid microglial cells were simultaneously present. The ramified cells were located in or under the astrocyte monolayer, while the ameboid cells were over the layer as revealed by confocal laser scan microscopy. The majority of ramified cells appeared after the astrocyte layer was completely formed and they downregulated the expression of the major histocompatibility complex antigen. Fibronectin was detected around ramified microglial cells, and laminin was also present in the astrocyte monolayer in mixed brain cell culture, while both proteins were not distributed near ameboid cells over the monolayer. When purified microglial cells were cultured on astrocyte-derived extracellular matrix in serum-free medium, they ramified. These results show that the differentiation of microglial cells is observable in culture and that astrocytes may play pivotal roles in the differentiation mainly by secreting insoluble factors.
Collapse
Affiliation(s)
- J Tanaka
- Department of Physiology, School of Medicine, Ehime University, Shigenobu, Japan.
| | | | | | | |
Collapse
|
31
|
Kataoka K, Asai T, Taneda M, Ueshima S, Matsuo O, Kuroda R, Carmeliet P, Collen D. Nigral degeneration following striato-pallidal lesion in tissue type plasminogen activator deficient mice. Neurosci Lett 1999; 266:220-2. [PMID: 10465713 DOI: 10.1016/s0304-3940(99)00310-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tissue type plasminogen activator (tPA) has been suggested as a key factor in excitotoxic neuronal death in the hippocampus. Transneuronal degeneration of the substantia nigra pars reticulata (SNR) neurons after striato-pallidal lesions is attributable to excess excitatory glutamatergic inputs into the SNR following inhibitory GABAergic deafferentation and tPA may contribute to the mechanism of transneuronal degeneration of the SNR. To examine this possibility, we studied pathological changes in the SNR following striato-pallidal lesions produced by electrocoagulation in tPA-deficient mice. There was no difference in the degree of SNR degeneration, or in microglial activation and proliferation in the degenerating SNR of tPA-deficient and control mice. Our results indicate that tPA does not contribute to transneuronal degeneration in the SNR following striato-pallidal lesions in mice.
Collapse
Affiliation(s)
- K Kataoka
- Department of Neurosurgery, Kinki University, School of Medicine, Osaka-Sayama, Osaka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Damage to the central nervous system (CNS) elicits the activation of both astrocytes and microglia. This review is focused on the principal features that characterize the activation of microglia after CNS injury. It provides a critical discussion of concepts regarding microglial biology that include the relationship between microglia and macrophages, as well as the role of microglia as immunocompetent cells of the CNS. Mechanistic and functional aspects of microgliosis are discussed primarily in the context of microglial neuronal interactions. The controversial issue of whether reactive microgliosis is a beneficial or a harmful process is addressed, and a resolution of this dilemma is offered by suggesting different interpretations of the term 'activated microglia' depending on its usage during in vivo or in vitro experimentation.
Collapse
Affiliation(s)
- W J Streit
- Department of Neuroscience, University of Florida College of Medicine and Brain Institute, Gainesville 32610, USA.
| | | | | |
Collapse
|
33
|
|