451
|
Ishida Y, Nagai A, Kobayashi S, Kim SU. Upregulation of protease-activated receptor-1 in astrocytes in Parkinson disease: astrocyte-mediated neuroprotection through increased levels of glutathione peroxidase. J Neuropathol Exp Neurol 2006; 65:66-77. [PMID: 16410750 DOI: 10.1097/01.jnen.0000195941.48033.eb] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In the present study, we investigated the expression of protease-activated receptors (PARs), receptors for thrombin, in substantia nigra pars compacta (SNpc) of Parkinson disease (PD) brains and cultures of human neurons, astrocytes, oligodendrocytes, and microglia as determined by immunocytochemistry and reverse transcriptase-polymerase chain reaction (RT-PCR). Expression of PAR-1 was demonstrated only in glial fibrillary acidic protein-positive astrocytes in SNpc, and the number of astrocytes expressing PAR-1 increased in SNpc of PD as compared with nonneurologic control brain. Immunoreactivity for thrombin and prothrombin was stronger in astrocytes and the vessel walls in SNpc of PD brains. PAR-1 was expressed in human astrocytes and neurons, but not in oligodendrocytes or microglia as determined by RT-PCR. We investigated thrombin-mediated activation of human astrocytes. Thrombin treatment activates human astrocytes and induces morphologic change and a marked increase in proliferation of astrocytes. Increased expression of glial cell line-derived growth factor and glutathione peroxidase (GPx) but no change in the expression of nerve growth factor and inflammatory cytokines/chemokine (IL-1beta, IL-6, IL-8, MCP-1) was found in thrombin/PAR-activated astrocytes. Next, we studied the neuroprotective effect exerted by thrombin-activated astrocytes in human cerebral neuron x human neuroblastoma hybrid neurons. Although thrombin showed neurotoxicity against human hybrid neurons in a dose-dependent manner, the conditioned media derived from thrombin-pretreated astrocyte cultures promoted the survival of human hybrid neurons. The protective effect was completely inhibited with a GPx inhibitor, mercaptosuccinic acid, indicating that GPx released from thrombin/PAR-activated astrocytes is responsible for neuroprotection of hybrid neurons against thrombin cytotoxicity. The present study suggests that the increased expression of PAR-1 in astrocytes in SNpc of PD brain is the restorative move taken by the brain to provide neuroprotection against neuronal degeneration and cell death of dopaminergic neurons caused by noxious insults during the progression of PD pathology.
Collapse
Affiliation(s)
- Yuri Ishida
- Department of Neurology and Department of Laboratory Medicine, Shimane University School of Medicine, Izumo, Japan
| | | | | | | |
Collapse
|
452
|
Jalabi W, Boehm N, Grucker D, Ghandour MS. Recovery of myelin after induction of oligodendrocyte cell death in postnatal brain. J Neurosci 2006; 25:2885-94. [PMID: 15772348 PMCID: PMC6725149 DOI: 10.1523/jneurosci.2748-04.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A transgenic mouse line (Oligo-TTK) was established to monitor oligodendrocyte cell death and myelin formation in the CNS. The expression of a conditionally toxic gene, the herpes simplex virus-1 thymidine kinase (HSV1-TK), was made under control of the MBP (myelin basic protein) gene promoter. A truncated form of the HSV1-TK (TTK) gene was used to avoid both bystander effect resulting from leaking in thymidine kinase activity and sterility in transgenic males observed in previous transgenic mice. The transgene was expressed in the CNS with a restricted localization in oligodendrocytes. Oligodendrocyte proliferation and myelin formation are therefore tightly controlled experimentally by administration of ganciclovir (GCV) via the induction of oligodendrocyte cell death. The most severe and irreversible hypomyelination was obtained when GCV was given daily from postnatal day 1 (P1) to P30. Oligodendrocyte plasticity and myelin recovery were analyzed in another phenotype generated by GCV treatment from P1 to P15. In this model, after dysmyelination, an apparent normal behavior was restored with no visible pathological symptoms by P30. Proliferating cells, which may be implicated in myelin repair in this model, are detected primarily in myelin tracts expressing the oligodendrocyte phenotype. Therefore, the endogenous potential of oligodendrocytes to remyelinate was clearly demonstrated in the mice of this study.
Collapse
MESH Headings
- Age Factors
- Analysis of Variance
- Animals
- Animals, Newborn
- Antiviral Agents/pharmacology
- Brain/cytology
- Brain/growth & development
- Bromodeoxyuridine/metabolism
- Cell Death/drug effects
- Cell Death/physiology
- Demyelinating Diseases/chemically induced
- Demyelinating Diseases/metabolism
- Disease Models, Animal
- Ganciclovir/pharmacology
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Gene Expression Regulation, Developmental/physiology
- Herpesvirus 1, Human/physiology
- Immunohistochemistry/methods
- In Situ Hybridization/methods
- Male
- Mice
- Mice, Transgenic
- Microscopy, Electron, Transmission/methods
- Myelin Basic Protein/genetics
- Myelin Basic Protein/metabolism
- Myelin Sheath/metabolism
- Oligodendroglia/drug effects
- Oligodendroglia/physiology
- Promoter Regions, Genetic/physiology
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Thymidine Kinase/genetics
Collapse
Affiliation(s)
- Walid Jalabi
- Institut de Physique Biologique, Unité Mixte de Recherche 7004, Université Louis Pasteur/Centre National de la Recherche Scientifique, Faculté de Médecine, 67085 Strasbourg, France
| | | | | | | |
Collapse
|
453
|
Mohri I, Taniike M, Okazaki I, Kagitani-Shimono K, Aritake K, Kanekiyo T, Yagi T, Takikita S, Kim HS, Urade Y, Suzuki K. Lipocalin-type prostaglandin D synthase is up-regulated in oligodendrocytes in lysosomal storage diseases and binds gangliosides. J Neurochem 2006; 97:641-51. [PMID: 16515539 DOI: 10.1111/j.1471-4159.2006.03753.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lipocalin-type prostaglandin (PG) D synthase (L-PGDS) is a dually functional protein, acting both as a PGD2-synthesizing enzyme and as an extracellular transporter of various lipophilic small molecules. L-PGDS is expressed in oligodendrocytes (OLs) in the central nervous system and is up-regulated in OLs of the twitcher mouse, a model of globoid cell leukodystrophy (Krabbe's disease). We investigated whether up-regulation of L-PGDS is either unique to Krabbe's disease or is a more generalized phenomenon in lysosomal storage disorders (LSDs), using LSD mouse models of Tay-Sachs disease, Sandhoff disease, GM1 gangliosidosis and Niemann-Pick type C1 disease. Quantitative RT-PCR revealed that L-PGDS mRNA was up-regulated in the brains of all these mouse models. In addition, strong L-PGDS immunoreactivity was observed in OLs, but not in either astrocytes or microglia in these models. Thus, up-regulation of L-PGDS appears to be a common response of OLs in LSDs. Moreover, surface plasmon resonance analyses revealed that L-PGDS binds GM1 and GM2 gangliosides, accumulated in neurons in the course of LSD, with high affinities (KD = 65 and 210 nm, respectively). This suggests that L-PGDS may play a role in scavenging harmful lipophilic substrates in LSD.
Collapse
Affiliation(s)
- Ikuko Mohri
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
454
|
Hsu JYC, Xu XM. Early profiles of axonal growth and astroglial response after spinal cord hemisection and implantation of Schwann cell-seeded guidance channels in adult rats. J Neurosci Res 2006; 82:472-83. [PMID: 16240391 DOI: 10.1002/jnr.20662] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We previously demonstrated that transplantation of Schwann cell-seeded channels promoted the regrowth of injured axons in the adult spinal cord. It is not clear, however, whether injured axons recapitulate the developmental scenarios to accomplish regeneration. In the present study, we investigated the early events associated with axonal regrowth after spinal cord hemisection at the eighth thoracic level and implantation of a Schwann cell-seeded minichannel in adult rats. Animals were sacrificed at postoperative days (PO) 2, 4, 7, and 14. Anterograde tracing with fluoro-ruby showed that regenerating axons grew into the graft prior to PO2 and reached the distal end of the channel at PO7. These axons expressed both embryonic neural cell adhesion molecule (E-NCAM) and growth associated protein-43 (GAP-43). Although the expression of E-NCAM decreased by PO7, that of GAP-43 remained high throughout the first 2 weeks after implantation. A close relation of vimentin-positive astroglia to the growing axons in the host tissue suggested a contact-mediated role of these cells in axon guidance. Aggregation of glial fibrillary acidic protein (GFAP)-positive astrocytes together with the increased expression of chondroitin sulfate proteoglycans (CSPGs) starting at PO7 appeared to inhibit axonal growth at the host-graft interface. Thus, adult regenerating axons and astroglia do express developmentally related molecules that may facilitate axonal growth into a permissive graft at the early phase of injury and regeneration. These results suggest that molecules and astroglia essential to development are both important in influencing axonal regrowth in the adult spinal cord.
Collapse
Affiliation(s)
- Jung-Yu C Hsu
- Department of Anatomy and Neurobiology, Saint Louis University School of Medicine, Missouri, USA
| | | |
Collapse
|
455
|
Shamekh R, Newcomb J, Mallery J, Cassady CJ, Saporta S, Cameron DF, Sanberg PR, Willing AE. Survival of rat or mouse ventral mesencephalon neurons after cotransplantation with rat sertoli cells in the mouse striatum. Cell Transplant 2006; 14:551-64. [PMID: 16358409 DOI: 10.3727/000000005783982747] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transplanting cells across species (xenotransplantation) for the treatment of Parkinson's disease has been considered an option to alleviate ethical concerns and shortage of tissues. However, using this approach leads to decreased cell survival; the xenografted cells are often rejected. Sertoli cells (SCs) are testis-derived cells that provide immunological protection to developing germ cells and can enhance survival of both allografted and xenografted cells. It is not clear whether these cells will maintain their immunosuppressive support of cografted cells if they are transplanted across species. In this study, we investigated the immune modulatory capacity of SCs and the feasibility of xenografting these cells alone or with allografted and xenografted neural tissue. Transplanting xenografts of rat SCs into the mouse striatum with either rat or mouse ventral mesencephalon prevented astrocytic infiltration of the graft site, although all transplants showed activated microglia within the core of the graft. Surviving tyrosine hydroxylase-positive neurons were observed in all conditions, but the size of the grafts was small at best. SCs were found at 1 and 2 weeks posttransplant. However, few SCs were found at 2 months posttransplant. Further investigation is under way to characterize the immune capabilities of SCs in a xenogeneic environment.
Collapse
Affiliation(s)
- R Shamekh
- Center of Excellence for Aging & Brain Repair, University of South Florida, College of Medicine, Tampa 33612, USA
| | | | | | | | | | | | | | | |
Collapse
|
456
|
Muranyi M, Ding C, He Q, Lin Y, Li PA. Streptozotocin-induced diabetes causes astrocyte death after ischemia and reperfusion injury. Diabetes 2006; 55:349-55. [PMID: 16443767 DOI: 10.2337/diabetes.55.02.06.db05-0654] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diabetes exacerbates neuronal cell death induced by cerebral ischemia. One contributing factor is enhanced acidosis during ischemia. Astrocytes are vulnerable to hypoxia under acidic conditions in vitro and may be targets of ischemia under diabetic conditions. The objective of this study was to determine whether diabetes would cause damage to astrocytes after an ischemic brain injury in vivo. Diabetic and nondiabetic rats were subjected to 5 min of forebrain ischemia and followed by 30 min, 6 h, or 1 or 3 days of recovery. The results showed that ischemia caused activation of astrocytes in nondiabetic rats. In contrast, diabetes caused astrocyte activation in early stage of reperfusion and astrocyte death in late stage of reperfusion. Remarkable astrocyte death was preceded by increased DNA oxidation. Further studies revealed that increased astrocyte damage coincided with enhanced production of free radicals. These data suggest that hyperglycemic ischemia worsens outcome in astrocytes, as it does in neurons.
Collapse
Affiliation(s)
- Marianna Muranyi
- Department of Complementary and Alternative Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, 96822, USA
| | | | | | | | | |
Collapse
|
457
|
DERVAN AG, TOTTERDELL S, LAU YS, MEREDITH GE. Altered Striatal Neuronal Morphology Is Associated with Astrogliosis in a Chronic Mouse Model of Parkinson's Disease. Ann N Y Acad Sci 2006. [DOI: 10.1111/j.1749-6632.2003.tb07487.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
458
|
Weisman GA, Wang M, Kong Q, Chorna NE, Neary JT, Sun GY, González FA, Seye CI, Erb L. Molecular determinants of P2Y2 nucleotide receptor function: implications for proliferative and inflammatory pathways in astrocytes. Mol Neurobiol 2006; 31:169-83. [PMID: 15953819 DOI: 10.1385/mn:31:1-3:169] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2004] [Accepted: 11/15/2004] [Indexed: 01/05/2023]
Abstract
In the mammalian nervous system, P2 nucleotide receptors mediate neurotransmission, release of proinflammatory cytokines, and reactive astrogliosis. Extracellular nucleotides activate multiple P2 receptors in neurons and glial cells, including G protein-coupled P2Y receptors and P2X receptors, which are ligand-gated ion channels. In glial cells, the P2Y2 receptor subtype, distinguished by its ability to be equipotently activated by ATP and UTP, is coupled to pro-inflammatory signaling pathways. In situ hybridization studies with rodent brain slices indicate that P2Y2 receptors are expressed primarily in the hippocampus and cerebellum. Astrocytes express several P2 receptor subtypes, including P2Y2 receptors whose activation stimulates cell proliferation and migration. P2Y2 receptors, via an RGD (Arg-Gly-Asp) motif in their first extracellular loop, bind to alphavbeta3/beta5 integrins, whereupon P2Y2 receptor activation stimulates integrin signaling pathways that regulate cytoskeletal reorganization and cell motility. The C-terminus of the P2Y2 receptor contains two Src-homology-3 (SH3)-binding domains that upon receptor activation, promote association with Src and transactivation of growth factor receptors. Together, our results indicate that P2Y2 receptors complex with both integrins and growth factor receptors to activate multiple signaling pathways. Thus, P2Y2 receptors present novel targets to control reactive astrogliosis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry and Neuroscience Program, University of Missouri-Columbia, Columbia, MO, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
459
|
Sharif A, Canton B, Junier MP, Chneiweiss H. PEA-15 Modulates TNFα Intracellular Signaling in Astrocytes. Ann N Y Acad Sci 2006; 1010:43-50. [PMID: 15033692 DOI: 10.1196/annals.1299.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PEA-15 is a small protein (15 kDa) that was first identified as an abundant phosphoprotein in brain astrocytes and subsequently shown to be widely expressed in different tissues and highly conserved among mammals. It is composed of an N-terminal death effector domain (DED) and a C-terminal tail of irregular structure. PEA-15 is regulated by multiple calcium-dependent phosphorylation pathways. PEA-15 is ideally positioned to play a major role in signal integration. Accordingly, it has been demonstrated that PEA-15 diverts astrocytes from TNFalpha-triggered apoptosis and regulates the actions of the ERK MAP kinase cascade by binding to ERK and altering its subcellular localization. Expression of PEA-15 directs TNFalpha outcomes toward survival, whereas its absence allows the development of the cytokine-induced cell death.
Collapse
Affiliation(s)
- Ariane Sharif
- INSERM U114, Department de Neuropharmacologie, Collège de France, 75231 Paris Cedex 05, France
| | | | | | | |
Collapse
|
460
|
González FA, Weisman GA, Erb L, Seye CI, Sun GY, Velázquez B, Hernández-Pérez M, Chorna NE. Mechanisms for inhibition of P2 receptors signaling in neural cells. Mol Neurobiol 2006; 31:65-79. [PMID: 15953812 DOI: 10.1385/mn:31:1-3:065] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2004] [Accepted: 11/15/2004] [Indexed: 12/24/2022]
Abstract
Trophic factors are required to ensure neuronal viability and regeneration after neural injury. Although abundant information is available on the factors that cause the activation of astrocytes, little is known about the molecular mechanisms underlying the regulation of this process. Nucleotides released into the extracellular space from injured or dying neural cells can activate astrocytes via P2 nucleotide receptors. After a brief historical review and update of novel P2 receptor antagonists, this article focuses on recent advancements toward understanding molecular mechanisms that regulate G protein-coupled P2Y receptor signaling. Among P2Y receptor subtypes, the heptahelical P2Y2 nucleotide receptor interacts with vitronectin receptors via an RGD sequence in the first extracellular loop, and this interaction is required for effective signal transduction to activate mitogen-activated protein kinases ERK1/2, to mobilize intracellular calcium stores via activation of phospholipase C, protein kinase C isoforms, and to activate focal adhesion kinase and other signaling events. Ligation of vitronectin receptors with specific antibodies caused an inhibition of P2Y2 receptor-induced ERK1/2 and p38 phosphorylation and P2Y2 receptor-induced cytoskeleton rearrangement and DNA synthesis. Structure-function studies have identified agonist-induced phosphorylation of the C-terminus of the P2Y2 receptor, an important mechanism for receptor desensitization. Understanding selective mechanisms for regulating P2Y2 receptor signaling could provide novel targets for therapeutic strategies in the management of brain injury, synaptogenesis, and neurological disorders.
Collapse
Affiliation(s)
- Fernando A González
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Puerto Rico.
| | | | | | | | | | | | | | | |
Collapse
|
461
|
Falsig J, Pörzgen P, Lund S, Schrattenholz A, Leist M. The inflammatory transcriptome of reactive murine astrocytes and implications for their innate immune function. J Neurochem 2006; 96:893-907. [PMID: 16405499 DOI: 10.1111/j.1471-4159.2005.03622.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Upon injury, astrocytes assume an activated state associated with the release of inflammatory mediators. To model this, we stimulated murine primary astrocytes with a complete inflammatory cytokine mix consisting of TNF-alpha, IL-1beta and IFN-gamma. We analysed the transcriptional response of 480 genes at 4 and 16 h after stimulation on a chip designed to give a representative overview over the inflammation-relevant part of the transcriptome of macrophage-like cells. The list of the 182 genes found to be significantly regulated in astrocytes revealed an intriguing co-ordinate regulation of genes linked to the biological processes of antiviral/antimicrobial defence, antigen presentation and facilitation of leucocyte invasion. The latter group was characterized by very high up-regulations of chemokine genes. We also identified regulations of a thymidylate kinase and an interferon-regulated protein with a tetratricopeptide motive, both up to now only known from macrophages. The transcriptional regulations were confirmed on the protein level by a proteomic analysis. These findings taken together suggest that activated astrocytes in brain behave similarly in many respects to inflamed macrophages in the periphery.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Astrocytes/drug effects
- Astrocytes/immunology
- Cell Cycle/genetics
- Cell Death/genetics
- Cells, Cultured
- Cerebral Cortex/cytology
- Chemokine CCL5/genetics
- Chemokine CCL5/metabolism
- Cytokines/pharmacology
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay/methods
- Female
- Glial Fibrillary Acidic Protein/metabolism
- Immunity, Innate/genetics
- Immunity, Innate/physiology
- Immunohistochemistry/methods
- Inflammation/genetics
- Inflammation/metabolism
- Mice
- Mice, Inbred C57BL
- Models, Immunological
- Nitrites/metabolism
- Oligonucleotide Array Sequence Analysis/methods
- Pregnancy
- Proteomics/methods
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
- Stress, Physiological/genetics
- Time Factors
Collapse
Affiliation(s)
- Jeppe Falsig
- Institute of Neuropathology, University of Zürich, Schmelzbergstrasse 12, CH-8032 Zürich, Switzerland.
| | | | | | | | | |
Collapse
|
462
|
Hoshi A, Nakahara T, Kayama H, Yamamoto T. Ischemic tolerance in chemical preconditioning: Possible role of astrocytic glutamine synthetase buffering glutamate-mediated neurotoxicity. J Neurosci Res 2006; 84:130-41. [PMID: 16688719 DOI: 10.1002/jnr.20869] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glutamine synthetase (GS), localized to astrocyte is a key enzyme in the glutamate-glutamine pathway in the brain. 3-Nitropropionic acid (3-NPA) is an irreversible inhibitor of succinate dehydrogenase in the tricarboxylic-acid cycle, and provides ischemic tolerance to the brain. So far, there have been no reports on the relationship of astrocytic GS and ischemic tolerance by chemical preconditioning. In order to test the hypothesis that astrocytes serve a pivotal role in 3-NPA-induced chemical preconditioning, we have investigated the temporal profile of GS expression in astrocyte parallel with those of glial fibrillary acidic protein and heat-shock protein 70 (HSP70). In our rat model of permanent focal ischemia, preconditioning with 3-NPA singnificantly reduced the subsequent neurological deficits and infarct volume within 24-72 hours after treatment. Immunohistochemically, protoplasmic astrocytes in the cortex and striatum were activated in terms of upregulation of GS and more abundant protoplasmic processes with 3-NPA preconditioning, however, HSP70 expression could not be induced. Thus, the activation of astrocytes and upregulation of GS play an important role in 3-NPA-induced preconditioning but HSP70 does not. In view of glutamate being imposed on the cerebral ischemic damage, the astrocytic GS may contribute to 3-NPA-induced ischemic tolerance.
Collapse
Affiliation(s)
- Akihiko Hoshi
- Department of Neurology, Fukushima Medical University, Fukushima, Japan
| | | | | | | |
Collapse
|
463
|
Kimura N, Takahashi M, Tashiro T, Terao K. Amyloid β up-regulates brain-derived neurotrophic factor production from astrocytes: Rescue from amyloid β-related neuritic degeneration. J Neurosci Res 2006; 84:782-9. [PMID: 16862545 DOI: 10.1002/jnr.20984] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Astrocytes, the most abundant type of glia in the brain, are considered to play a key role in Alzheimer's disease (AD) pathologies. In a cell culture study, we have previously shown that astroglial responses against amyloid beta (Abeta) occur before obvious neuronal damage could be detected, suggesting the possibility that astrocytes might be an attractive therapeutic target for treating AD. In the present study, we investigated astroglial gene expression changes in response to Abeta to elucidate further the role of astrocytes in Abeta toxicity. By using real-time PCR and ELISA analyses, we found that Abeta rapidly induced astrocytes to produce brain-derived neurotrophic factor (BDNF). Abeta42 was more effective than Abeta40 in increasing astroglial BDNF production. Moreover, BDNF treatment rescued the neuronally differentiated human neuroblastoma cells from neuritic degeneration caused by Abeta toxicity. This is the first study to demonstrate that astrocytes are capable of increasing the production of a particular neurotrophic factor in response to Abeta. Our findings also identify BDNF as a potential therapeutic agent for preventing Abeta-related neuritic degeneration.
Collapse
Affiliation(s)
- Nobuyuki Kimura
- Laboratory of Disease Control, Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Tsukuba-shi, Ibaraki, Japan.
| | | | | | | |
Collapse
|
464
|
Adori C, Andó RD, Kovács GG, Bagdy G. Damage of serotonergic axons and immunolocalization of Hsp27, Hsp72, and Hsp90 molecular chaperones after a single dose of MDMA administration in Dark Agouti rat: Temporal, spatial, and cellular patterns. J Comp Neurol 2006; 497:251-69. [PMID: 16705678 DOI: 10.1002/cne.20994] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, "ecstasy") causes long-term disturbance of the serotonergic system. We examined the temporal, spatial, and cellular distribution of three molecular chaperones, Hsp27, Hsp72, and Hsp90, 3 and 7 days after treatment with 7.5, 15, and 30 mg/kg single intraperitoneal (i.p.) doses of MDMA in Dark Agouti rat brains. Furthermore, we compared the immunostaining patterns of molecular chaperones with serotonergic axonal-vulnerability evaluated by tryptophan-hydroxylase (TryOH) immunoreactivity and with astroglial-activation detected by GFAP-immunostaining. There was a marked reduction in TryOH-immunoreactive axon density after MDMA treatment in all examined areas at both time points. Three days after treatment, a significant dose-dependent increase in Hsp27-immunoreactive protoplasmic astrocytes was found in the cingulate, frontal, occipital, and pyriform cortex, and in the hippocampus CA1. However, there was no increase in astroglial Hsp27-immunoreactivity in the caudate putamen, lateral septal nucleus, or anterior hypothalamus. A significant increase in the GFAP immunostaining density of protoplasmic astrocytes was found only in the hippocampus CA1. In addition, numerous strong Hsp72-immunopositive neurons were found in some brain areas only 3 days after treatment with 30 mg/kg MDMA. Increased Hsp27-immunoreactivity exclusively in the examined cortical areas reveals that Hsp27 is a sensitive marker of astroglial response to the effects of MDMA in these regions of Dark Agouti rat brain and suggests differential responses in astroglial Hsp27-expression between distinct brain areas. The co-occurrence of Hsp27 and GFAP response exclusively in the hippocampus CA1 may suggest the particular vulnerability of this region. The presence of strong Hsp72-immunopositive neurons in certain brain areas may reflect additional effects of MDMA on nonserotonergic neurons.
Collapse
Affiliation(s)
- Csaba Adori
- Laboratory of Neurochemistry and Experimental Medicine, National Institute of Psychiatry and Neurology, Budapest, Hungary
| | | | | | | |
Collapse
|
465
|
Masilamoni JG, Jesudason EP, Baben B, Jebaraj CE, Dhandayuthapani S, Jayakumar R. Molecular chaperone alpha-crystallin prevents detrimental effects of neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2005; 1762:284-93. [PMID: 16443350 DOI: 10.1016/j.bbadis.2005.11.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Revised: 11/03/2005] [Accepted: 11/11/2005] [Indexed: 12/01/2022]
Abstract
Silver nitrate administration stimulates immune activation, inflammation and deterioration in cell function. It is well established that hippocampal and cortical tissue are susceptible to degeneration in responses to insult such as oxidative stress or infection. This study was designed to investigate the prophylactic effect of alpha-crystallin, a major chaperone lens protein comprising of alpha-A and alpha-B subunits in inflammation induced mice. Mice were divided into three groups (n=6 in each), control, inflammation and alpha-crystallin treated. Our result shows that alpha-crystallin pretreatment effectively diminished systemic inflammation induced glial fibrillary acidic protein (GFAP) and nuclear factor kappa B (NFkappaB) expression in the mice neocortex, reversed elevated intracellular calcium levels, acetylcholine esterase activity and depletion of glucose. Furthermore it also significantly prevented nitric oxide (P<0.05) and lipid peroxide production in the plasma, liver, neocortex and hippocampus of the inflammation-induced mice. In order to demonstrate the direct *OH and nitric oxide radical scavenging ability of alpha-crystallin, an In vitro experiment using primary astrocyte culture subjected to lipopolysaccharide (LPS), a well-known inflammatory stimuli were also carried out. This study reiterates that alpha-crystallin therapy may serve as a potent pharmacological agent in neuroinflammation.
Collapse
Affiliation(s)
- J G Masilamoni
- Bio-Organic and Neurochemistry Laboratory, Central Leather Research Institute, Adyar, Chennai-600 020, India
| | | | | | | | | | | |
Collapse
|
466
|
Okamoto M, Wang X, Baba M. HIV-1-infected macrophages induce astrogliosis by SDF-1alpha and matrix metalloproteinases. Biochem Biophys Res Commun 2005; 336:1214-20. [PMID: 16169519 DOI: 10.1016/j.bbrc.2005.08.251] [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] [Received: 08/26/2005] [Accepted: 08/31/2005] [Indexed: 11/17/2022]
Abstract
Brain macrophages/microglia and astrocytes are known to be involved in the pathogenesis of HIV-1-associated dementia (HAD). To clarify their interaction and contribution to the pathogenesis, HIV-1-infected or uninfected macrophages were used as a model of brain macrophages/microglia, and their effects on human astrocytes in vitro were examined. The culture supernatants of HIV-1-infected or uninfected macrophages induced significant astrocyte proliferation, which was annihilated with a neutralizing antibody to stromal cell-derived factor (SDF)-1alpha or a matrix metalloproteinase (MMP) inhibitor. In these astrocytes, CXCR4, MMP, and tissue inhibitors of matrix metalloproteinase mRNA expression and SDF-1alpha production were significantly up-regulated. The supernatants of infected macrophages were always more effective than those of uninfected cells. Moreover, the enhanced production of SDF-1alpha was suppressed by the MMP inhibitor. These results indicate that the activated and HIV-1-infected macrophages can indirectly induce astrocyte proliferation through up-regulating SDF-1alpha and MMP production, which implies a mechanism of astrogliosis in HAD.
Collapse
Affiliation(s)
- Mika Okamoto
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan
| | | | | |
Collapse
|
467
|
Sun W, Gerhardinger C, Dagher Z, Hoehn T, Lorenzi M. Aspirin at low-intermediate concentrations protects retinal vessels in experimental diabetic retinopathy through non-platelet-mediated effects. Diabetes 2005; 54:3418-26. [PMID: 16306357 DOI: 10.2337/diabetes.54.12.3418] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The prevention of diabetic retinopathy requires drugs that leverage the benefits of glycemic control without adding the burden of side effects. Aspirin at dosages of 1-1.5 g/day has prevented manifestations of diabetic retinal microangiopathy in a clinical trial as well as in studies with dogs. Because lower and safer doses of aspirin could be used if its beneficial effects on retinopathy were due to antithrombotic effects, we compared the effects of a selective antiplatelet drug (clopidogrel) to those of aspirin in streptozotocin-induced diabetic rats. Clopidogrel did not prevent neuronal apoptosis, glial reactivity, capillary cell apoptosis, or acellular capillaries in the retina of diabetic rats. Aspirin, at doses yielding serum levels (<0.6 mmol/l) well below the anti-inflammatory range for humans, prevented apoptosis of capillary cells and the development of acellular capillaries but did not prevent neuroglial abnormalities. The aldose reductase inhibitor sorbinil, used as the benchmark for the effect of the other drugs, prevented all abnormalities. The diabetic rat retina showed increased expression of the transcription factor CCAAT/enhancer-binding protein-beta, one of the known targets of low-intermediate concentrations of aspirin. Thus we found a spectrum of drug efficacy on the prevention of experimental diabetic retinopathy, ranging from the absent effect of a selective antiplatelet drug to the prevention of all abnormalities by an aldose reductase inhibitor. Aspirin at low-intermediate concentrations selectively prevented microangiopathy. The minimal effective dose of aspirin should now be sought.
Collapse
Affiliation(s)
- Wei Sun
- Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, Massachusetts 02114, USA
| | | | | | | | | |
Collapse
|
468
|
Abstract
The enteric nervous system is composed of both neurons and glia. Recent evidence indicates that enteric glia-which vastly outnumber enteric neurons-are actively involved in the control of gastrointestinal functions: they contain neurotransmitter precursors, have the machinery for uptake and degradation of neuroligands, and express neurotransmitter-receptors which makes them well suited as intermediaries in enteric neurotransmission and information processing in the ENS. Novel data further suggest that enteric glia have an important role in maintaining the integrity of the mucosal barrier of the gut. Finally, enteric glia may also serve as a link between the nervous and immune systems of the gut as indicated by their potential to synthesize cytokines, present antigen and respond to inflammatory insults. The role of enteric glia in human disease has not yet been systematically studied, but based on the available evidence it is predictable that enteric glia are involved in the etiopathogenesis of various pathological processes in the gut, particularly such with neuroinflammatory or neurodegenerative components.
Collapse
Affiliation(s)
- A Rühl
- Department of Human Biology, Technical University of Munich, Germany.
| |
Collapse
|
469
|
Varlet P. [Histology and oligodendrogenesis of glial cells]. Neurochirurgie 2005; 51:229-38. [PMID: 16292166 DOI: 10.1016/s0028-3770(05)83483-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Oligodendrogenesis is a complex and dynamic phenomenon. Knowledge of the underlying molecular control mechanisms advances steadily, especially in rodents. While the parallelism with human oligodendrogenesis is not fully established, the main characteristics are recognized. Neuroepithelial cells of the neural tube participate in both gliogenesis and neurogenesis. Oligodendrogenesis begins after neurogenesis and stops after birth. It is a focal phenomenon under the control of specific morphogenic proteins, and can generate precursors which are able to proliferate and migrate in the same time. Five steps of oligodendrogliogenesis follow one another acquiring and loosing proteinic markers. They lead to intricated maturation steps for generating myelinizing oligodendrocytes, NG2 cells and precursors of quiescient adult oligodendrocytes.
Collapse
Affiliation(s)
- P Varlet
- Laboratoire de Neuropathologie, Centre Hospitalier Sainte-Anne, Paris.
| |
Collapse
|
470
|
Zurich MG, Lengacher S, Braissant O, Monnet-Tschudi F, Pellerin L, Honegger P. Unusual astrocyte reactivity caused by the food mycotoxin ochratoxin A in aggregating rat brain cell cultures. Neuroscience 2005; 134:771-82. [PMID: 15994020 DOI: 10.1016/j.neuroscience.2005.04.030] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2004] [Revised: 02/21/2005] [Accepted: 04/13/2005] [Indexed: 11/26/2022]
Abstract
Ochratoxin A (OTA), a mycotoxin and widespread food contaminant, is known for its patent nephrotoxicity and potential neurotoxicity. Previous observations in vitro showed that in the CNS, glial cells were particularly sensitive to OTA. In the search for the molecular mechanisms underlying OTA neurotoxicity, we investigated the relationship between OTA toxicity and glial reactivity, in serum-free aggregating brain cell cultures. Using quantitative reverse transcriptase-polymerase chain reaction to analyze changes in gene expression, we found that in astrocytes, non cytotoxic concentrations of OTA down-regulated glial fibrillary acidic protein, while it up-regulated vimentin and the peroxisome proliferator-activated receptor-gamma expression. OTA also up-regulated the inducible nitric oxide synthase and the heme oxygenase-1. These OTA-induced alterations in gene expression were more pronounced in cultures at an advanced stage of maturation. The natural peroxisome proliferator-activated receptor-gamma ligand, 15-deoxy-delta(12,14) prostaglandin J2, and the cyclic AMP analog, bromo cyclic AMP, significantly attenuated the strong induction of peroxisome proliferator-activated receptor-gamma and inducible nitric oxide synthase, while they partially reversed the inhibitory effect of OTA on glial fibrillary acidic protein. The present results show that OTA affects the cytoskeletal integrity of astrocytes as well as the expression of genes pertaining to the brain inflammatory response system, and suggest that a relationship exists between the inflammatory events and the cytoskeletal changes induced by OTA. Furthermore, these results suggest that, by inducing an atypical glial reactivity, OTA may severely affect the neuroprotective capacity of glial cells.
Collapse
Affiliation(s)
- M-G Zurich
- Department of Physiology, University of Lausanne, Rue du Bugnon 7, CH-1005 Lausanne, Switzerland.
| | | | | | | | | | | |
Collapse
|
471
|
Phillips J, Pearce B. Serum deprivation and re-addition: effects on cyclooxygenase inhibitor sensitivity in cultured glia. Inflammopharmacology 2005; 13:431-9. [PMID: 16280096 DOI: 10.1163/156856005774649368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A number of drugs were assessed for their ability to inhibit stimulus-evoked prostanoid synthesis in cultured glia. These drugs included non-selective cyclooxygenase (COX) inhibitors and those considered to be selective for the inducible isoform of this enzyme (COX-2). Experiments were carried out on normal cultures and those which had been maintained in serum-free growth medium for four days then re-exposed to serum for a further seven days. All of the drugs tested elicited concentration-dependent inhibitions of arachidonic acid (AA)-stimulated thromboxane B(2) (TXB(2)) accumulation in normal cultures with the following rank order of potency: indomethacin > piroxicam > nimesulide = NS398 > ibuprofen >> aspirin > paracetamol. In cultures which had been deprived of serum for four days, basal and AA-stimulated TXB(2) production was considerably reduced, as was the amount of COX immunoreactivity determined by Western blotting. Basal and AA-stimulated TXB(2) production together with COX immunoreactivity were restored to control levels by the re-addition of serum to serum-deprived cultures for 7 days. In these cultures, the rank order of potency was: indomethacin > piroxicam >> ibuprofen > nimesulide = NS398 >> aspirin > paracetamol; however, there were marked charges in the apparent IC(50) values for particular drugs. Indomethacin, piroxicam and aspirin were very similar to control, but the potencies of ibuprofen (3-fold), NS398 (30-fold) and nimesulide (40-fold) were found to be decreased when compared to control. Paracetamol, on the other hand, was found to be almost 3-fold more potent under these conditions. Glia appear to express a COX with a novel sensitivity to particular inhibitors following serum deprivation and re-addition.
Collapse
Affiliation(s)
- James Phillips
- Department of Pharmacology, The School of Pharmacy, 29/39 Brunswick Square, London WC1N 1AX, UK
| | | |
Collapse
|
472
|
Zhang S, Zhu C, Liu Q, Wang W. Effects of chloroquine on GFAP, PCNA and cyclin D1 in hippocampus and cerebral cortex of rats with seizures induced by pentylenetetrazole. ACTA ACUST UNITED AC 2005; 25:625-8. [PMID: 16696308 DOI: 10.1007/bf02896153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The effects of chloroquine on glial fibrillary acidic protein (GFAP), proliferation cell nuclear antigen (PCNA) and Cyclin D1 in hippocampus and cerebral cortex of rats with seizures induced by pentylenetetrazole (PTZ) were observed in the present study. Forty-eight male adult Sprague-Dawley (SD) rats were randomly divided into control group, chloroquine intervening group, and PTZ group. The behavior and electroencephalogram (EEG) were observed and recorded. GFAP and PCNA were examined with immunohistochemistry. The content of Cyclin D1 in hippocampus and cerebral cortex was inspected with Western blot. The results showed no seizure activity in the control group, severe seizure activity in the PTZ group (IV - V degree), and slight seizure activity (I - IIl degree) in the chloroquine intervening group (P < 0.05). EEG recordings showed no epileptic spikes in the control group, high amplitude with fast frequency in the PTZ group, low-amplitude and slow frequency in the chloroquine intervening group. The expression of GFAP and the positive index of PCNA in the PTZ group were higher than those of control group (P < 0.05 and P < 0.01, respectively). No differences in GFAP expression and PCNA index were observed between chloroquine intervening and control groups (P > 0.05). The content of Cyclin D1 in hippocampus and cerebral cortex was significantly higher in the PTZ group than in control and chloroquine intervening groups (P < 0.05). Therefore, it is considered that chloroquine, by inhibiting the functions and proliferation of glial cells in the hippocampus and cerebral cortex, can alleviate the seizure activities. These results suggest that chloroquine may be an ideal anticonvulsant in preventing and treating epilepsy.
Collapse
Affiliation(s)
- Shuhua Zhang
- Neuroscience Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | | | |
Collapse
|
473
|
Taft JR, Vertes RP, Perry GW. Distribution of GFAP+ astrocytes in adult and neonatal rat brain. Int J Neurosci 2005; 115:1333-43. [PMID: 16048809 DOI: 10.1080/00207450590934570] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Astrocytes can proliferate as a result of trauma to the brain, such as occurs in a variety of diseases. Understanding the normal distribution of astrocytes is necessary before the extent of astrogliosis can be clearly determined. However, little is known about the normal distribution of GFAP+ astrocytes especially during development. This study examined distribution of GFAP+ astrocytes in regions of the cortex, cerebellum, and brainstem of adult and rat pup brains by immunocytochemistry using antibodies against GFAP. The findings showed a differential distribution of GFAP+ astrocytes in the rat brain. A paucity of GFAP expression was found in most regions of the normal adult rat brainstem, whereas GFAP+ astrocytes were abundantly distributed in all areas of the cortex and cerebellum. A similar regional heterogeneity in the distribution of GFAP+ astrocytes was seen in the neonatal rat brain. These findings suggest that the development of the differential pattern of GFAP+ astrocytes seen in the rat brain does not occur postnatally, but instead is present at birth and appears to be determined during fetal development.
Collapse
Affiliation(s)
- Janna R Taft
- Department of Psychology and Center for Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL 24450, USA.
| | | | | |
Collapse
|
474
|
Melø TM, Nehlig A, Sonnewald U. Metabolism is normal in astrocytes in chronically epileptic rats: a (13)C NMR study of neuronal-glial interactions in a model of temporal lobe epilepsy. J Cereb Blood Flow Metab 2005; 25:1254-64. [PMID: 15902201 DOI: 10.1038/sj.jcbfm.9600128] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The aim of the present work was to study potential disturbances in metabolism and interactions between neurons and glia in the lithium-pilocarpine model of temporal lobe epilepsy. Rats chronically epileptic for 1 month received [1-(13)C]glucose, a substrate for neurons and astrocytes, and [1,2-(13)C]acetate, a substrate for astrocytes only. Analyses of extracts from cerebral cortex, cerebellum, and hippocampal formation (hippocampus, amygdala, entorhinal, and piriform cortices) were performed using (13)C and (1)H nuclear magnetic resonance spectroscopy and HPLC. In the hippocampal formation of epileptic rats, levels of glutamate, aspartate, N-acetyl aspartate, adenosine triphosphate plus adenosine diphosphate and glutathione were decreased. In all regions studied, labeling from [1,2-(13)C]acetate was similar in control and epileptic rats, indicating normal astrocytic metabolism. However, labeling of glutamate, GABA, aspartate, and alanine from [1-(13)C]glucose was decreased in all areas possibly reflecting neuronal loss. The labeling of glutamine from [1-(13)C]glucose was decreased in cerebral cortex and cerebellum and unchanged in hippocampal formation. In conclusion, no changes were detected in glial-neuronal interactions in the hippocampal formation while in cortex and cerebellum the flow of glutamate to astrocytes was decreased, indicating a disturbed glutamate-glutamine cycle. This is, to our knowledge, the first study showing that metabolic disturbances are confined to neurons inside the epileptic circuit.
Collapse
Affiliation(s)
- Torun M Melø
- Department of Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | | | | |
Collapse
|
475
|
Koyama Y, Baba A, Matsuda T. Endothelins stimulate the expression of neurotrophin-3 in rat brain and rat cultured astrocytes. Neuroscience 2005; 136:425-33. [PMID: 16181740 DOI: 10.1016/j.neuroscience.2005.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Revised: 07/31/2005] [Accepted: 08/01/2005] [Indexed: 10/25/2022]
Abstract
Endothelins play a role in the regulation of astrocytic functions in brain pathologies such as hyperplasia and neurotrophic factor production. The present study examined the effects of endothelins on production of neurotrophin-3, a member of the neurotrophin family of neurotrophic factors, in cultured astrocytes and rat brain. Quantitative reverse transcription-PCR analysis of mRNA copy numbers showed that cultured astrocytes expressed comparable numbers of neurotrophin-3 and neurotrophin-4/5 mRNA copies to nerve growth factor and brain-derived neurotrophic factor. Endothelin-1 (100 nM) and Ala1,3,11,15-endothelin-1 (an endothelinB receptor agonist, 100 nM) caused a transient increase in neurotrophin-3 mRNA levels, but not in neurotrophin-4/5 levels, in cultured astrocytes. The increases in mRNA levels were accompanied with that in extracellular release of neurotrophin-3. The effects of endothelin-1 on neurotrophin-3 mRNA levels were reduced by BQ788, an endothelinB receptor antagonist. I.c.v. administration of 500 pmol/day Ala1,3,11,15-endothelin-1 increased mRNA and peptide levels of neurotrophin-3 in rat caudate putamen and cerebrum. On the other hand, neurotrophin-3 production in hippocampus was not affected by Ala1,3,11,15-endothelin-1. Immunohistochemical examination of Ala1,3,11,15-endothelin-1-infused rats showed that neurotrophin-3 was mainly expressed in glial fibrillary acidic protein-positive astrocytes in caudate putamen and cerebrum. endothelin-induced increases in neurotrophin-3 expression in cultured astrocytes were inhibited by chelation of intracellular Ca2+ and PD98095 (an ERK inhibitor). These results suggest that endothelin is an extracellular signal that stimulates astrocytic neurotrophin-3 production in brain pathologies.
Collapse
Affiliation(s)
- Y Koyama
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-Oka 1-6 Suita, 565-0871, Japan
| | | | | |
Collapse
|
476
|
Bauer A, Zilles K, Matusch A, Holzmann C, Riess O, von Hörsten S. Regional and subtype selective changes of neurotransmitter receptor density in a rat transgenic for the Huntington's disease mutation. J Neurochem 2005; 94:639-50. [PMID: 16033418 DOI: 10.1111/j.1471-4159.2005.03169.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Huntington's disease (HD) is an autosomal dominantly inherited progressive neurodegenerative disorder caused by a CAG/polyglutamine repeat expansion in the gene encoding the huntingtin protein. We have recently generated a rat model transgenic for HD, which displays a slowly progressive phenotype resembling the human adult-onset type of disease. In this study we systematically assessed the distribution and density of 17 transmitter receptors in the brains of 2-year-old rats using quantitative multi-tracer autoradiography and high-resolution positron emission tomography. Heterozygous animals expressed increased densities of M(2) acetylcholine (increase of 148 +/- 16% of controls; p > 0.001; n = 7), nicotine (increase of 149 +/- 16% of controls; p > 0.01; n = 6), and alpha(2) noradrenergic receptors (increase of 141 +/- 15% of controls; p > 0.001; n = 6), respectively. Densities of these receptors were decreased in homozygous animals. Decreases of receptor density in both hetero- and homozygous animals were found for M1 acetylcholine, 5-HT 2A serotonin, A 2A adenosine, D1 and D2 dopamine, and GABA(A) receptors, respectively. Other investigated receptor systems showed small changes or were not affected. The present data suggest that the moderate increase of CAG/polyglutamine repeat expansions in the present rat model of Huntington's disease is characterized by subtype-selective and region-specific changes of neuroreceptor densities. In particular, there is evidence for a contribution of predominantly presynaptically localized cholinergic and noradrenergic receptors in the response to Huntington's disease pathology.
Collapse
Affiliation(s)
- Andreas Bauer
- Institute of Medicine, Research Center Jülich, Jülich, Germany.
| | | | | | | | | | | |
Collapse
|
477
|
Veyrac A, Didier A, Colpaert F, Jourdan F, Marien M. Activation of noradrenergic transmission by alpha2-adrenoceptor antagonists counteracts deafferentation-induced neuronal death and cell proliferation in the adult mouse olfactory bulb. Exp Neurol 2005; 194:444-56. [PMID: 16022870 DOI: 10.1016/j.expneurol.2005.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Revised: 03/02/2005] [Accepted: 03/07/2005] [Indexed: 11/20/2022]
Abstract
The olfactory bulb is the target of neural progenitor cells that are generated in the subventricular zone of the lateral ventricle in the adult brain. This permanent neurogenesis is likely influenced by olfactory input to the bulb since previous studies have shown that cell proliferation and/or apoptotic death are stimulated by naris closure or surgical transection of the olfactory nerve. Since the olfactory bulb is densely innervated by noradrenergic afferents originating in the locus coeruleus, we have studied the impact of pharmacologically activating this noradrenergic system on cell death and proliferation following unilateral olfactory axotomy in the adult mouse olfactory bulb. We found that noradrenaline release in the olfactory bulb was significantly increased by intraperitoneal injections of the selective alpha(2)-adrenoceptor antagonists, dexefaroxan (0.63 mg/kg) and 5-fluoro-methoxyidazoxan (F 14413; 0.16 mg/kg). A chronic treatment with either compound for 7 days following olfactory axotomy significantly reduced neuronal death, glial activation and cell proliferation in the deafferented olfactory bulb. These data (1) confirm that alpha(2)-adrenoceptor antagonists, presumably by facilitating central noradrenergic transmission, afford neuroprotection in vivo, as previously shown in models of cerebral ischemia, excitotoxicity and devascularization-induced neurodegeneration, and (2) support a role of the locus coeruleus noradrenergic system in promoting survival of neurons in areas of the brain where neurogenesis persists in the adult.
Collapse
Affiliation(s)
- Alexandra Veyrac
- Laboratoire Neurosciences et Systèmes Sensoriels, CNRS-UMR 5020, Université Claude Bernard-Lyon 1, 50 Avenue Tony Garnier, F-69366 Lyon, France
| | | | | | | | | |
Collapse
|
478
|
Mander P, Brown GC. Activation of microglial NADPH oxidase is synergistic with glial iNOS expression in inducing neuronal death: a dual-key mechanism of inflammatory neurodegeneration. J Neuroinflammation 2005; 2:20. [PMID: 16156895 PMCID: PMC1232863 DOI: 10.1186/1742-2094-2-20] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2005] [Accepted: 09/12/2005] [Indexed: 12/20/2022] Open
Abstract
Background Inflammation-activated glia are seen in many CNS pathologies and may kill neurons through the release of cytotoxic mediators, such as nitric oxide from inducible NO synthase (iNOS), and possibly superoxide from NADPH oxidase (NOX). We set out to determine the relative role of these species in inducing neuronal death, and to test the dual-key hypothesis that the production of both species simultaneously is required for significant neuronal death. Methods Primary co-cultures of cerebellar granule neurons and glia from rats were used to investigate the effect of NO (from iNOS, following lipopolysaccharide (LPS) and/or cytokine addition) or superoxide/hydrogen peroxide (from NOX, following phorbol 12-myristate 13-acetate (PMA), ATP analogue (BzATP), interleukin-1β (IL-1β) or arachidonic acid (AA) addition) on neuronal survival. Results Induction of glial iNOS caused little neuronal death. Similarly, activation of NOX alone resulted in little or no neuronal death. However, if NOX was activated (by PMA or BzATP) in the presence of iNOS (induced by LPS and interferon-γ) then substantial delayed neuronal death occurred over 48 hours, which was prevented by inhibitors of iNOS (1400W), NOX (apocynin) or a peroxynitrite decomposer (FeTPPS). Neurons and glia were also found to stain positive for nitrotyrosine (a putative marker of peroxynitrite) only when both iNOS and NOX were simultaneously active. If NOX was activated by weak stimulators (IL-1β, AA or the fibrillogenic prion peptide PrP106-126) in the presence of iNOS, it caused microglial proliferation and delayed neurodegeneration over 6 days, which was prevented by iNOS or NOX inhibitors, a peroxynitrite decomposer or a NMDA-receptor antagonist (MK-801). Conclusion These results suggest a dual-key mechanism, whereby glial iNOS or microglial NOX activation alone is relatively benign, but if activated simultaneously are synergistic in killing neurons, through generating peroxynitrite. This mechanism may mediate inflammatory neurodegeneration in response to cytokines, bacteria, ATP, arachidonate and pathological prions, in which case neurons may be protected by iNOS or NOX inhibitors, or scavengers of NO, superoxide or peroxynitrite.
Collapse
Affiliation(s)
- Palwinder Mander
- Biochemistry Department, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Guy C Brown
- Biochemistry Department, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
| |
Collapse
|
479
|
Höltje M, Hoffmann A, Hofmann F, Mucke C, Grosse G, Van Rooijen N, Kettenmann H, Just I, Ahnert-Hilger G. Role of Rho GTPase in astrocyte morphology and migratory response during in vitro wound healing. J Neurochem 2005; 95:1237-48. [PMID: 16150054 DOI: 10.1111/j.1471-4159.2005.03443.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Small Rho GTPases are key regulators of the cytoskeleton in a great variety of cells. Rho function mediates morphological changes as well as locomotor activity. Using astrocyte cultures established from neonatal mice we investigated the role of Rho in process formation during astrocyte stellation. Using a scratch-wound model, we examined the impact of Rho on a variety of morphological and functional variables such as stellation and migratory activity during wound healing. C3 proteins are widely used to study cellular Rho functions. In addition, C3 derived from Clostridium botulinum (C3bot) is considered selectively to promote neuronal regeneration. Because the latter requires a balanced activity of neurones and glial cells, the effects of C3 protein on glial cells such as astrocytes have to be considered carefully. Low nanomolar concentrations of C3 proteins significantly promoted process outgrowth and increased process branching. Besides enzymatic inactivation of Rho by ADP-ribosylation, changes in protein levels of the various Rho GTPases may also contribute to the observed effects. Furthermore, incubation of scratch-wounded astrocyte cultures with C3bot accelerated wound healing. By inhibiting the Rho downstream effector ROCK with the selective inhibitor Y27632 we were able to demonstrate that the accelerated wound closure resulted from both enhanced polarized process formation and increased migratory activity of astrocytes into the lesion site. These results suggest that Rho negatively regulates astrocytic process growth and migratory responses after injury and that its inactivation by C3bot in nanomolar concentrations promotes astrocyte migration.
Collapse
Affiliation(s)
- Markus Höltje
- Charité-Universitätsmedizin Berlin, Centrum für Anatomie, AG Funktionelle Zellbiologie, Humboldt-Universität zu Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
480
|
Foote AK, Blakemore WF. Repopulation of oligodendrocyte progenitor cell-depleted tissue in a model of chronic demyelination. Neuropathol Appl Neurobiol 2005; 31:374-83. [PMID: 16008821 DOI: 10.1111/j.1365-2990.2005.00647.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Some, but not all, chronically demyelinated multiple sclerosis (MS) lesions are depleted of oligodendrocyte progenitor cells (OPCs) suggesting that OPCs are destroyed during the process of demyelination and some factor impedes OPC repopulation of the depleted tissue. The chronically demyelinated axons in MS lie in an astrocytic environment and it has been proposed that this might impede entry of OPCs into such regions. By depleting a short length of spinal cord of its OPCs using 40 Gy of X-irradiation in both normal rats and rats with progressive myelin loss accompanied by an astrocytosis (taiep rats), we investigated whether such changes affect the ability of OPCs to repopulate OPC-depleted tissue. In both taiep and normal rats, the rate of repopulation decreases with age, but no difference was detected in the rate at which OPCs repopulated normally myelinated and chronically demyelinated and astrocytosed tissue. This indicates that, if the astrocytic environment of the taiep central nervous system (CNS) is comparable to that found in MS lesions, then the presence of chronically demyelinated axons and astrocytosis in chronic MS lesions does not represent a barrier to repopulation of the tissue by OPCs. However, similar to the situation in the normal adult rodent CNS, the rate of repopulation by endogenous OPCs in aged taiep rats is very slow, approximately 0.2 mm per week.
Collapse
Affiliation(s)
- A K Foote
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
481
|
Kinsner A, Pilotto V, Deininger S, Brown GC, Coecke S, Hartung T, Bal-Price A. Inflammatory neurodegeneration induced by lipoteichoic acid from Staphylococcus aureus is mediated by glia activation, nitrosative and oxidative stress, and caspase activation. J Neurochem 2005; 95:1132-43. [PMID: 16144539 DOI: 10.1111/j.1471-4159.2005.03422.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this study we investigated the mechanisms of neuronal cell death induced by lipoteichoic acid (LTA) and muramyl dipeptide (MDP) from Gram-positive bacterial cell walls using primary cultures of rat cerebellum granule cells (CGCs) and rat cortical glial cells (astrocytes and microglia). LTA (+/- MDP) from Staphylococcus aureus induced a strong inflammatory response of both types of glial cells (release of interleukin-1beta, tumour necrosis factor-alpha and nitric oxide). The death of CGCs was caused by activated glia because in the absence of glia (treatment with 7.5 microm cytosine-d-arabinoside to inhibit non-neuronal cell proliferation) LTA + MDP did not cause significant cell death (less than 20%). In addition, staining with rhodamine-labelled LTA confirmed that LTA was bound only to microglia and astrocytes (not neurones). Neuronal cell death induced by LTA (+/- MDP)-activated glia was partially blocked by an inducible nitric oxide synthase inhibitor (1400 W; 100 microm), and completely blocked by a superoxide dismutase mimetic [manganese (III) tetrakis (4-benzoic acid)porphyrin chloride; 50 microm] and a peroxynitrite scavenger [5,10,15,20-tetrakis (4-sulfonatophenyl) porphyrinato iron (III); 100 microm] suggesting that nitric oxide and peroxynitrite contributed to LTA-induced cell death. Moreover, neuronal cell death was inhibited by selective inhibitors of caspase-3 (z-DEVD-fmk; 50 microm) and caspase-8 (z-Ile-Glu(O-Me)-Thr-Asp(O-Me) fluoromethyl ketone; 50 microm) indicating that they were involved in LTA-induced neuronal cell death.
Collapse
Affiliation(s)
- Agnieszka Kinsner
- European Centre for the Validation of Alternative Methods (ECVAM), European Commission Joint Research Centre, Ispra, Italy
| | | | | | | | | | | | | |
Collapse
|
482
|
Galoyan AA, Sarkissian JS, Chavushyan VA, Sulkhanyan RM, Avakyan ZE, Avetisyan ZA, Grigorian YK, Abrahamyan DO. Neuroprotective action of hypothalamic peptide PRP-1 at various time survivals following spinal cord hemisection. Neurochem Res 2005; 30:507-25. [PMID: 16076021 DOI: 10.1007/s11064-005-2686-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The purpose of the present study was to evaluate the neuroprotective action of proline-rich peptide-1 (PRP-1) produced by hypothalamic nuclei cells (nuclei paraventricularis and supraopticus) following lateral hemisection of spinal cord (SC). The dynamics of rehabilitative shifts were investigated at various periods of postoperative survival (1-2, 3, and 4 weeks), both with administration of PRP-1 and without it (control). We registered evoked spike flow activity in both interneurons and motoneurons of the same segment of transected and symmetric intact sides of SC and below it on the stimulation of mixed (n. ischiadicus), flexor (n. gastrocnemius) and extensor (n. peroneus communis) nerves. In the control group (administration of 0.9% saline as placebo), no significant decrease of post-stimulus activity of neurons was observed on the transected side by the 2nd week. This activity strongly decreased by week 3 postaxotomy, with some increase on the intact side, possibly of compensatory origin. No shifts occurred by the 4th week. Regardless of the period of administration, PRP-1 increased neuronal activity on the transected side, with the same activation levels on both SC sides. These data were confirmed by histochemical investigation. PRP-1 administration, both daily and every other day, for a period of 2-3 weeks led to prevention of scar formation and promotion of the re-growth of white matter nerve fibers in the damaged area. It also resulted in prevention of neuroglial elements degeneration and reduction in gliosis expression in the lesion supporting neuronal survival. Thus, PRP-1 achieved protection against "tissue stress", which was also confirmed by the registration of activity on the level of transection and restoration of the motor activity on the injured side. The obtained data propose the possibility of PRP-1 application in clinical practice for prevention of neurodegeneration of traumatic origin.
Collapse
Affiliation(s)
- Armen A Galoyan
- Buniatian Institute of Biochemistry NAS RA, Yerevan, Republic of Armenia.
| | | | | | | | | | | | | | | |
Collapse
|
483
|
Hamby ME, Uliasz TF, Hewett SJ, Hewett JA. Characterization of an improved procedure for the removal of microglia from confluent monolayers of primary astrocytes. J Neurosci Methods 2005; 150:128-37. [PMID: 16105687 DOI: 10.1016/j.jneumeth.2005.06.016] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2004] [Revised: 06/15/2005] [Accepted: 06/15/2005] [Indexed: 11/26/2022]
Abstract
Cultures of astrocytes can be readily established and are widely used to study the biological functions of these glial cells in isolation. Unfortunately, contamination by microglia can confound results from such studies. Herein, a simple and highly effective modification of a common procedure to remove microglia from astrocyte cultures is described. After becoming confluent, astrocytes were exposed to a mitotic inhibitor for 5-6 days then treated with 50-75 mM l-leucine methyl ester (LME) for 60-90 min. Unlike previous protocols that employed lower LME concentrations on subconfluent cultures or during passage of astrocytes, this protocol effectively depleted microglia from high-density astrocyte monolayers. This was evidenced by the selective depletion of microglial-specific markers. Purified monolayers appeared morphologically normal 24h after LME treatment and expressed nitric oxide synthase-2 (NOS-2) and cyclooxygenase-2 (COX-2) proteins upon stimulation with LPS plus IFNgamma, albeit to a lower level than unpurified monolayers. This difference could be attributed to removal of contaminating microglia from monolayers and not to astrocyte dysfunction, since LME treatment did not alter global protein synthesis and a reactive phenotype could be induced in the purified monolayers. Thus, this modified protocol selectively depletes microglia from high-density primary astrocyte monolayers without compromising their functional integrity.
Collapse
Affiliation(s)
- Mary E Hamby
- Department of Neuroscience MC 3401, University of Connecticut Health Center, 263 Farmington Avenue, CT 06030-3401, USA
| | | | | | | |
Collapse
|
484
|
Nicchia GP, Srinivas M, Li W, Brosnan CF, Frigeri A, Spray DC. New possible roles for aquaporin-4 in astrocytes: cell cytoskeleton and functional relationship with connexin43. FASEB J 2005; 19:1674-6. [PMID: 16103109 DOI: 10.1096/fj.04-3281fje] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Aquaporin-4 (AQP4), the main water channel in the brain, is expressed in the perivascular membranes of mouse, rat, and human astrocytes. In a previous study, we used small interfering RNA (siRNA) to specifically knock down AQP4 in rat astrocyte primary cultures and found that together with reduced osmotic permeability, AQP4 knockdown (KD) led to altered cell morphology. However, a recent report on primary cultured astrocytes from AQP4 null mice (KO) showed no morphological differences compared with wild types. In this study, we compared the effect of AQP4 KD in mouse, rat, and human astrocyte primary cultures and found that AQP4 KD in human astrocytes resulted in a morphological phenotype similar to that found in rat. In contrast, AQP4 KD in mouse astrocytes caused only very mild morphological changes. The actin cytoskeleton of untreated astrocytes exhibited strong species-specific differences, with F-actin being organized in cortical bands in mouse and in stress fibers in rat and human astrocytes. Surprisingly, as a consequence of AQP4 KD, F-actin cytoskeleton was depolymerized in rat and human whereas it was completely rearranged in mouse astrocytes. Although AQP4 KD induced alterations of the cell cytoskeleton, we found that the expression of dystrophin (DP71), beta-dystroglycan, and alpha-syntrophin was not altered. AQP4 KD in cultured mouse astrocytes produced strong down-regulation of connexin43 (Cx43) with a concomitant reduction in cell coupling while no major alterations in Cx43 expression were found in rat and human cells. Taken together, these results demonstrate that with regard to these properties, human astrocytes in culture are more similar to rat than to mouse astrocytes. Moreover, even though AQP4 KD in mouse astrocytes did not result in a dramatic morphological phenotype, it induced a remarkable rearrangement of F-actin, not related to disruption of the dystrophin complex, indicating a primary role of this water channel in the cytoskeleton changes observed. Finally, the strong down-regulation of Cx43 and cell coupling in AQP4 KD mouse astrocytes indicate that a functional relationship likely exists between water channels and gap junctions in brain astrocytes.
Collapse
Affiliation(s)
- Grazia P Nicchia
- Department of General and Environmental Physiology and Centre of Excellence in Comparative Genomics (CEGBA), University of Bari, Bari, Italy.
| | | | | | | | | | | |
Collapse
|
485
|
Koyama Y, Tsujikawa K, Matsuda T, Baba A. Endothelin increases expression of exon III- and exon IV-containing brain-derived neurotrophic factor transcripts in cultured astrocytes and rat brain. J Neurosci Res 2005; 80:809-16. [PMID: 15898104 DOI: 10.1002/jnr.20512] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The effects of endothelins (ETs) on brain-derived neurotrophic factor (BDNF) production in astrocytes were investigated. ET-1 (100 nM) increased the mRNA level and extracellular release of BDNF in cultured astrocytes. RT-PCR analyses using primer pairs that amplified exon-specific BDNF transcripts revealed that exon III- and exon IV-containing BDNF transcripts existed in cultured astrocytes, whereas exon I- and exon II-containing BDNF transcripts did not. ET-1 and Ala(1,3,11,15)-ET-1, an ET(B) receptor agonist, increased the expressions of the exon III and exon IV transcripts in cultured astrocytes. Intracerebroventricular administration of 500 pmol/day of Ala(1,3,11,15)-ET-1 increased exon III and exon IV BDNF transcripts in the rat striatum. In cultured astrocytes, Ca(2+)-chelation, W-7 (a calmodulin inhibitor), and KN93 (a Ca(2+)/calmodulin kinase inhibitor) inhibited the increases in exon IV BDNF mRNA and CCAAT enhancer-binding protein beta (C/EBPbeta) levels induced by ET-1. The ET-induced increases in exon III BDNF mRNA expression and phosphorylation of cAMP response element binding protein (CREB) were reduced by Ca(2+) chelation, W-7, KN93, PD98059 (a MEK inhibitor), and wortmannin (a phosphatidylinositol 3-kinase inhibitor). These results suggest that ETs stimulate the expressions of exon III and exon IV BDNF transcripts in astrocytes through CREB and C/EBPbeta-mediated mechanisms, respectively.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.
| | | | | | | |
Collapse
|
486
|
Hashimoto K, Parker A, Malone P, Gabelt BT, Rasmussen C, Kaufman PS, Hernandez MR. Long-term activation of c-Fos and c-Jun in optic nerve head astrocytes in experimental ocular hypertension in monkeys and after exposure to elevated pressure in vitro. Brain Res 2005; 1054:103-15. [PMID: 16081055 DOI: 10.1016/j.brainres.2005.06.050] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2005] [Revised: 06/10/2005] [Accepted: 06/14/2005] [Indexed: 11/30/2022]
Abstract
This study investigates whether the immediate early gene (IEG) products c-Fos and c-Jun are activated in vivo in monkeys with experimental glaucoma, and in vitro in cultured human ONH astrocytes exposed to hydrostatic pressure (HP). Three Rhesus monkeys with mild glaucomatous damage (mean intraocular pressure (IOP) 27 +/- 1.3 mm Hg approximately 42 weeks) and three with moderate glaucomatous damage (mean IOP 44 +/- 6.7% mm Hg approximately 11 weeks) were used for this study; the contralateral eye served as normal control (mean IOP 18.6 +/- 1.7 mm Hg). ONH tissues were stained with GFAP, DAPI, and c-Jun or c-Fos, and transcription factor positive and negative nuclei were counted to determine nuclear localization. Cultured human normal and glaucomatous ONH astrocytes exposed to elevated HP served as the in vitro model of elevated pressure. Activation and nuclear localization of c-Fos and c-Jun increased significantly in the monkeys with elevated IOP. These data correlated with axonal loss, reactive astrocytes, and remodeling of the optic disc. Cultured human ONH astrocytes showed increased nuclear localization of c-Fos and c-Jun under exposure to HP. Immunohistochemistry demonstrated that the upstream regulators of c-Fos and c-Jun, ERK-MAPK and MAPKp38 localized to the nuclei of ONH astrocytes in monkeys with experimental glaucoma. Taken together, these results demonstrate c-Fos and c-Jun activation in ONH astrocytes in vivo and in vitro, and that activation of both transcription factors is associated with ERK and MAPKp38 activation in experimental glaucoma, suggesting that activation of transcription factors may participate in the induction and maintenance of the reactive astrocyte phenotype in glaucomatous optic neuropathy.
Collapse
Affiliation(s)
- K Hashimoto
- Division of Ophthalmology and Visual Science, Niigata University Graduate School, 1-757 Asahimachi, Niigata 951-8510, Japan
| | | | | | | | | | | | | |
Collapse
|
487
|
Silvia MT, Licht DJ. Pediatric central nervous system infections and inflammatory white matter disease. Pediatr Clin North Am 2005; 52:1107-26, ix. [PMID: 16009259 DOI: 10.1016/j.pcl.2005.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This article reviews the immunology of the central nervous system and the clinical presentation, diagnosis, and treatment of children with viral or parainfectious encephalitis. The emphasis is on the early recognition of treatable causes of viral encephalitis (herpes simplex virus), and the diagnosis and treatment of acute disseminated encephalomyelitis are described in detail. Laboratory and imaging findings in the two conditions also are described.
Collapse
Affiliation(s)
- Mary T Silvia
- Division of Neurology, The Children's Hospital of Philadelphia, 34th & Civic Center Boulevard, Philadelphia, PA 19104, USA
| | | |
Collapse
|
488
|
Guillemin GJ, Wang L, Brew BJ. Quinolinic acid selectively induces apoptosis of human astrocytes: potential role in AIDS dementia complex. J Neuroinflammation 2005; 2:16. [PMID: 16042813 PMCID: PMC1187916 DOI: 10.1186/1742-2094-2-16] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2005] [Accepted: 07/26/2005] [Indexed: 12/30/2022] Open
Abstract
There is evidence that the kynurenine pathway (KP) and particularly one of its end products, quinolinic acid (QUIN) play a role in the pathogenesis of several major neuroinflammatory diseases, and more particularly AIDS dementia complex (ADC). We hypothesized that QUIN may be involved in astrocyte apoptosis because: 1) apoptotic astrocytes have been observed in the brains of ADC patients, 2) ADC patients have elevated cerebrospinal fluid QUIN concentrations, and 3) QUIN can induce astrocyte death. Primary cultures of human fetal astrocytes were treated with three pathophysiological concentrations of QUIN. Numeration of apoptotic cells was assessed using double immunocytochemistry for expression of active caspase 3 and for nucleus condensation. We found that treatment of human astrocytes with QUIN induced morphological (cell body shrinking) and biochemical changes (nucleus condensation and over-expression of active caspase 3) of apoptosis. After 24 hours of treatment with QUIN 500 nM and 1200 nM respectively 10 and 14% of astrocytes were undergoing apoptosis. This would be expected to lead to a relative lack of trophic support factors with consequent neuronal dysfunction and possibly death. Astroglial apoptosis induced by QUIN provides another potential mechanism for the neurotoxicity of QUIN during ADC.
Collapse
Affiliation(s)
- Gilles J Guillemin
- Centre for Immunology, St Vincent's Hospital, Sydney, Australia
- University of New South Wales, Faculty of Medicine, Sydney, Australia
| | - Lily Wang
- Centre for Immunology, St Vincent's Hospital, Sydney, Australia
| | - Bruce J Brew
- Centre for Immunology, St Vincent's Hospital, Sydney, Australia
- Department of Neurology, St Vincent's Hospital, Sydney, Australia
| |
Collapse
|
489
|
Nishio T, Kawaguchi S, Yamamoto M, Iseda T, Kawasaki T, Hase T. Tenascin-C regulates proliferation and migration of cultured astrocytes in a scratch wound assay. Neuroscience 2005; 132:87-102. [PMID: 15780469 DOI: 10.1016/j.neuroscience.2004.12.028] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2004] [Indexed: 11/23/2022]
Abstract
Tenascin-C (TNC), an extracellular matrix glycoprotein, is involved in tissue morphogenesis like embryogenesis, wound healing or tumorigenesis. Astrocytes are known to play major roles in wound healing in the CNS. To elucidate the roles of TNC in wound closure by astrocytes, we have examined the morphological changes of cultured astrocytes in a scratch wound assay and measured the content of soluble TNC released into the medium. We have also localized the expression of TNC mRNA, TNC, glial fibrillary acidic protein (GFAP), vimentin and integrin beta1. After wounding, glial cells rapidly released the largest TNC isoform and proliferated in the border zones. Subsequently, they became polarized with unidirectional processes and finally migrated toward the denuded area. The proliferating border zone cells and pre-migratory cells intensely expressed TNC mRNA, TNC-, vimentin-, GFAP- and integrin beta1-like immunoreactivity, while the migratory cells showed generally reduced expression except the front. Exogenous TNC enhanced cell proliferation and migration, while functional blocking with anti-TNC or anti-integrin beta1 antibody reduced both of them. These results suggest that mechanical injury induces boundary astrocytes to produce and release TNC that promotes cell proliferation and migration via integrin beta1 in an autocrine/paracrine fashion.
Collapse
Affiliation(s)
- T Nishio
- Department of Integrative Brain Science, Kyoto University Graduate School of Medicine, Yoshida-Konoe, Sakyo, Kyoto 606-8501, Japan.
| | | | | | | | | | | |
Collapse
|
490
|
Hagemann TL, Gaeta SA, Smith MA, Johnson DA, Johnson JA, Messing A. Gene expression analysis in mice with elevated glial fibrillary acidic protein and Rosenthal fibers reveals a stress response followed by glial activation and neuronal dysfunction. Hum Mol Genet 2005; 14:2443-58. [PMID: 16014634 DOI: 10.1093/hmg/ddi248] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Alexander disease is a fatal neurodegenerative disorder resulting from missense mutations of the intermediate filament protein, GFAP. The pathological hallmark of this disease is the formation of cytoplasmic protein aggregates within astrocytes known as Rosenthal fibers. Transgenic mice engineered to over-express wild-type human GFAP develop an encephalopathy with identical aggregates, suggesting that elevated levels of GFAP in addition to mutant protein contribute to the pathogenesis of this disorder. To study further the effects of elevated GFAP and Rosenthal fibers per se, independent of mutations, we performed gene expression analysis on olfactory bulbs of transgenic mice at two different ages to follow the progression of pathology. The expression profiles reveal a stress response that includes genes involved in glutathione metabolism, peroxide detoxification and iron homeostasis. Many of these genes are regulated by the transcription factor Nfe2l2, which is also increased in expression at 3 weeks. An immune-related response occurs with activation of cytokine and cytokine receptor genes, complement components and acute phase response genes. These transcripts are further elevated with age, with additional induction of macrophage-specific markers such as Mac1 and CD68, suggesting activation of microglia. At 4 months, decreased expression of genes for microtubule-associated proteins, vesicular trafficking proteins and neurotransmitter receptors becomes apparent. Interneuron-specific transcription factors including Dlx family members and Pax6 are downregulated as well as Gad1 and Gad2, suggesting impairment of GABAergic granule cells. Together, these data implicate an initial stress response by astrocytes, which results in the activation of microglia and compromised neuronal function.
Collapse
|
491
|
Yano A, Shingo T, Takeuchi A, Yasuhara T, Kobayashi K, Takahashi K, Muraoka K, Matsui T, Miyoshi Y, Hamada H, Date I. Encapsulated vascular endothelial growth factor—secreting cell grafts have neuroprotective and angiogenic effects on focal cerebral ischemia. J Neurosurg 2005; 103:104-14. [PMID: 16121981 DOI: 10.3171/jns.2005.103.1.0104] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Object
The authors evaluated the neuroprotective and angiogenic effects of a continuous and low-dose infusion of vascular endothelial growth factor (VEGF)-165 on cerebral ischemia in rats.
Methods
The authors introduced VEGF complementary (c)DNA into baby hamster kidney (BHK) cells and established a cell line that produces human VEGF165 (BHK-VEGF). The BHK-VEGF cells and BHK cells that had been transfected with an expression vector that did not contain human VEGF165 cDNA (BHK-control) were encapsulated. Both capsules were implanted into rat striata. Six days after capsule implantation, the right middle cerebral artery (MCA) was occluded. Some animals were killed 24 hours after occlusion to measure the volume of the resulting infarct and to perform immunohistochemical studies. Other animals were used for subsequent behavioral studies 1, 7, and 14 days after MCA occlusion.
The encapsulated BHK-VEGF cell grafts significantly reduced the volume of the infarct and the number of apoptotic cells in the penumbral area when compared with the effect of the BHK-control cell capsule. In addition, angiogenesis and gliogenesis significantly increased in the region around the capsule in animals that received BHK-VEGF cell capsules without an increase in focal cerebral blood flow; this did not occur in animals that received the BHK-control cell capsule. In behavioral studies rats that received the BHK-VEGF cell capsule displayed significant recovery while participating in the accelerating rotarod test after stroke.
Conclusions
Continuous intracerebral administration of low-dose VEGF165 through encapsulated grafts of VEGF-producing cells produces neuroprotective and angiogenic effects. These effects improve subsequent motor function.
Collapse
Affiliation(s)
- Akimasa Yano
- Department of Neurological Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
492
|
Yoo YM, Lee U, Kim YJ. Apoptosis and nestin expression in the cortex and cultured astrocytes following 6-OHDA administration. Neurosci Lett 2005; 382:88-92. [PMID: 15911127 DOI: 10.1016/j.neulet.2005.02.070] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2004] [Revised: 02/22/2005] [Accepted: 02/24/2005] [Indexed: 11/19/2022]
Abstract
We used the dopaminergic neurotoxicant, 6-hydroxydopamine (6-OHDA), as a tool to characterize the origins of the astrocytic response to injury. Reactive astrocytes were examined by immunocyto- and histo-chemical visualization of nestin protein in the brain and cultivated cells. Following 6-OHDA (dose-dependent) treatment, the expression of nestin-like immunoreactive cells in the corpus callosum and cerebral cortex was increased compared with that of the control animals, indicating that a significant up-regulation of nestin protein occurred in these regions. In the corpus callosum and cerebral cortex, the majority of the nestin-like immunoreactive cells showed a distribution and pattern similar to those of the glial fibrillary acid protein (GFAP)-immunoreactive cells. Double immunofluorescence measurements showed that 100% of the nestin-like immunoreactive cells expressed GFAP-immunoreactive cells, indicating that these nestin-like immunoreactive cells belong to a reactive population of the astrocytes. In this study, we observed the morphological changes in the astrocytes following 6-OHDA administration, demonstrating that 6-OHDA induced injury leads to a rapid and transient up-regulation of nestin-like immunoreactivity in activated astrocytes.
Collapse
Affiliation(s)
- Young Mi Yoo
- Department of Neurosurgery, Gachon Medical College, Gil Medical Center, 1198 Kuwel-Dong, Incheon 405-220, South Korea.
| | | | | |
Collapse
|
493
|
Schubert KO, Naumann T, Schnell O, Zhi Q, Steup A, Hofmann HD, Kirsch M. Activation of STAT3 signaling in axotomized neurons and reactive astrocytes after fimbria-fornix transection. Exp Brain Res 2005; 165:520-31. [PMID: 15991029 DOI: 10.1007/s00221-005-2330-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Accepted: 03/01/2005] [Indexed: 10/25/2022]
Abstract
It is an open question to what extent neuroprotective mechanisms involving neurotrophic proteins are activated after central nervous system (CNS) lesions. Results from previous studies have indicated that ciliary neurotrophic factor (CNTF) and other members of the family of gp130-associated cytokines have neuroprotective effects on septohippocampal projection neurons axotomized by fimbria-fornix transection (FFT). Here we demonstrate that the transcription factor STAT3, a component of the primary cytokine signal transduction pathway, is transiently activated after FFT in the medial septum and in the lateral septum deafferented by the lesion. Immunocytochemical double-labeling showed nuclear signals for phosphorylated STAT3 in both types (GABAergic and cholinergic) of axotomized medial septal neurons around day 4 postlesion. This response temporally coincided with the cell-type-specific upregulation of the cytokine receptor components CNTF receptor alpha and LIF receptor beta in the same neurons. However, neuronal STAT3-activation was not abolished in CNTF- or LIF-deficient mouse mutants. Furthermore, lesion-induced STAT3 signaling was also found in reactive GFAP-positive astrocytes of the medial and lateral septum. Interestingly, basal GFAP expression was reduced but postlesional upregulation was markedly enhanced in CNTF(-/-) animals. These results demonstrate transient activation of cytokine signaling after CNS lesion and suggest that gp130-associated cytokines have multiple functions in the lesioned CNS by directly acting on axotomized neurons and on reactive astrocytes.
Collapse
Affiliation(s)
- Klaus Oliver Schubert
- Institute of Anatomy and Cell Biology I, University of Freiburg, P.O. Box 111, 79001, Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
494
|
Abstract
Astrocytes become activated (reactive) in response to many CNS pathologies, such as stroke, trauma, growth of a tumor, or neurodegenerative disease. The process of astrocyte activation remains rather enigmatic and results in so-called "reactive gliosis," a reaction with specific structural and functional characteristics. In stroke or in CNS trauma, the lesion itself, the ischemic environment, disrupted blood-brain barrier, the inflammatory response, as well as in metabolic, excitotoxic, and in some cases oxidative crises--all affect the extent and quality of reactive gliosis. The fact that astrocytes function as a syncytium of interconnected cells both in health and in disease, rather than as individual cells, adds yet another dimension to this picture. This review focuses on several aspects of astrocyte activation and reactive gliosis and discusses its possible roles in the CNS trauma and ischemia. Particular emphasis is placed on the lessons learnt from mouse genetic models in which the absence of intermediate filament proteins in astrocytes leads to attenuation of reactive gliosis with distinct pathophysiological and clinical consequences.
Collapse
Affiliation(s)
- Milos Pekny
- The Arvid Carlsson Institute for Neuroscience, Institute of Clinical Neuroscience, Sahlgrenska Academy at Göteborg University, Göteborg, Sweden
| | - Michael Nilsson
- The Arvid Carlsson Institute for Neuroscience, Institute of Clinical Neuroscience, Sahlgrenska Academy at Göteborg University, Göteborg, Sweden
| |
Collapse
|
495
|
Korolainen MA, Auriola S, Nyman TA, Alafuzoff I, Pirttilä T. Proteomic analysis of glial fibrillary acidic protein in Alzheimer's disease and aging brain. Neurobiol Dis 2005; 20:858-70. [PMID: 15979880 DOI: 10.1016/j.nbd.2005.05.021] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Revised: 05/12/2005] [Accepted: 05/17/2005] [Indexed: 01/04/2023] Open
Abstract
Chronic inflammation is known to play an important role in the heterogeneous pathogenesis of Alzheimer's disease (AD). Activated astrocytes expressing glial fibrillary acidic protein (GFAP) are closely associated with AD pathology, such as tangles, neuritic plaques and amyloid depositions. Altogether, 46 soluble isoforms of GFAP were separated and most of them quantified by two-dimensional immunoblotting in frontal cortices of AD patients and age-matched controls. A 60% increase in the amount of more acidic isoforms of GFAP was observed in AD and these isoforms were both phosphorylated and N-glycosylated, while more basic isoforms were O-glycosylated and exhibited no quantitative differences between post-mortem AD and control brains. These data highlight the importance of exploring isoform-specific levels of proteins in pathophysiological conditions since modifications of proteins determine their activity state, localization, turnover and interaction with other molecules. Mechanisms, structures and functional consequences of modification of GFAP isoforms remain to be clarified.
Collapse
Affiliation(s)
- Minna A Korolainen
- Department of Neuroscience and Neurology, University of Kuopio, Harjulantie 1D, P.O. Box 1627, FIN-70211 Kuopio, Finland.
| | | | | | | | | |
Collapse
|
496
|
Suárez I, Bodega G, Rubio M, Felipo V, Fernández B. Neuronal and inducible nitric oxide synthase expression in the rat cerebellum following portacaval anastomosis. Brain Res 2005; 1047:205-13. [PMID: 15904901 DOI: 10.1016/j.brainres.2005.04.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Revised: 04/05/2005] [Accepted: 04/15/2005] [Indexed: 10/25/2022]
Abstract
In order to determine the role of neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS) in the pathogenesis of experimental hepatic encephalopathy (HE), the expression of both was analyzed in the cerebellum of rats 1 month and 6 months after performing portacaval anastomosis (PCA). In control cerebella, nNOS immunoreactivity was mainly observed in the molecular layer (ML), whereas the Purkinje cells did not express nNOS. However, nNOS expression was detected in the Purkinje cells at 1 month after PCA, correlating with a decrease in nNOS expression in the ML--part of an overall reduction in cerebellar nNOS concentrations (as determined by Western blotting). At 6 months post-PCA, a significant increase in nNOS expression was observed in the ML, as well as increased nNOS immunoreactivity in the Purkinje cells. nNOS immunoreactivity was also observed in the Bergmann glial cells of PCA-treated rats. While no immunoreactivity for iNOS was seen in the cerebella of control rats, iNOS immunoreactivity was significantly induced in the cerebellum 1 month after PCA. In addition, the expression of iNOS was greater at 6 months than at 1 month post-PCA. Immunohistochemical analysis revealed this iNOS to be localized in the Purkinje cells and Bergmann glial cells. The induction of iNOS in astroglial cells has been associated with pathological conditions. Therefore, the iNOS expression observed in the Bergmann glial cells might play a role in the pathogenesis of HE, the harmful effects of PCA being caused by them via the production of excess nitric oxide. These results show that nNOS and iNOS are produced in the Purkinje cells and Bergmann glial cells following PCA, implicating nitric oxide in the pathology of HE.
Collapse
Affiliation(s)
- I Suárez
- Departamento de Biología Celular y Genética, Facultad de Biología, Universidad de Alcalá, 28871 Madrid, Spain.
| | | | | | | | | |
Collapse
|
497
|
Suh HW, Lee HK, Seo YJ, Kwon MS, Shim EJ, Lee JY, Choi SS, Lee JH. Kainic acid (KA)-induced Ca2+/Calmodulin-dependent protein kinase II (CaMK II) expression in the neurons, astrocytes and microglia of the mouse hippocampal CA3 region, and the phosphorylated CaMK II only in the hippocampal neurons. Neurosci Lett 2005; 381:223-7. [PMID: 15896474 DOI: 10.1016/j.neulet.2005.01.089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 01/30/2005] [Accepted: 01/31/2005] [Indexed: 11/20/2022]
Abstract
In the present study, we investigated the role of Ca2+/calmodulin-dependent protein kinase II (CaMK II) and which types of neuronal cells contain CaMK II and phosphorylated CaMK II (p-CaMK II) in the CA3 hippocampal region of mice using confocal immunofluorescence study. KA increased the CaMK II, p-CaMK II, glial fibrillary acidic protein (GFAP) and complement receptor type 3 (OX-42) immunoreactivities (IR) at 30 min after KA treatment in mouse hippocampal area. In studies, nevertheless KA-induced CaMK II is expressed in neurons or astrocytes or microglia, p-CaMK II is expressed only in neurons. Thus, our results suggest that the activated CaMK II in early time may be performed important roles only in neurons but not in the astrocytes and microglia.
Collapse
Affiliation(s)
- Hong-Won Suh
- Department of Pharmacology, Institute of Natural Medicine, College of Medicine, Hallym University, 1 Okchun-Dong, Chunchon, Gangwon-Do 200-702, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
498
|
Veiga S, Azcoitia I, Garcia-Segura LM. Ro5-4864, a peripheral benzodiazepine receptor ligand, reduces reactive gliosis and protects hippocampal hilar neurons from kainic acid excitotoxicity. J Neurosci Res 2005; 80:129-37. [PMID: 15696538 DOI: 10.1002/jnr.20430] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The peripheral-type benzodiazepine receptor (PBR) is a critical component of the mitochondrial permeability transition pore, which is involved in the regulation of cell survival. Different forms of brain injury result in induction of the expression of the PBR in the areas of neurodegeneration, mainly in reactive glial cells. The consequences of induction of PBR expression after brain injury are unknown. To test whether PBR may be involved in the regulation of neuronal survival after injury, we have assessed the effect of two PBR ligands, Ro5-4864 and PK11195, on neuronal loss induced by kainic acid in the hippocampus. Systemic administration of kainic acid to male rats resulted in the induction of a reactive phenotype in astrocytes and microglia and in a significant loss of hilar neurons in the dentate gyrus. Administration of Ro5-4864, before the injection of kainic acid, decreased reactive gliosis in the hilus and prevented hilar neuronal loss. In contrast, PK11195 was unable to reduce reactive gliosis and did not protect hilar neurons from kainic acid. These findings suggest that the PBR is involved in control of neuronal survival and gliosis after brain injury and identify this molecule as a potential target for neuroprotective interventions.
Collapse
|
499
|
Bartnik BL, Spigelman I, Obenaus A. Cell-permeant calcium buffer induced neuroprotection after cortical devascularization. Exp Neurol 2005; 192:357-64. [PMID: 15755553 DOI: 10.1016/j.expneurol.2004.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Revised: 09/24/2004] [Accepted: 11/10/2004] [Indexed: 11/23/2022]
Abstract
An excitotoxic cascade resulting in a significant intracellular calcium load is thought to be a primary mechanism leading to neuronal death after ischemia. One way to protect neurons from injury is through the use of cell-permeant calcium buffers. These molecules have been reported to be neuroprotective via their ability to increase the cell's overall Ca(2+) buffering load as well as by attenuating neurotransmitter release. However, their efficacy when given after injury has yet to be determined. We used diffusion-weighted magnetic resonance imaging (DWI), histological, and immunohistochemical methods to determine the neuroprotective efficacy of 2-aminophenol-N, N, O-triacetic acid acetoxymethyl ester (APTRA-AM) after focal cerebral ischemia. Injured animals were given two injections of APTRA-AM at 1 and 12 h after injury. Animals were imaged prior to injury and then at 12, 24, 48 h and 3 and 7 days after injury. After 7 days the animals were euthanized for correlative cresyl violet histology and immunohistochemistry. Injury resulted in a decrease in the apparent diffusion coefficient (ADC) of the injured area within the first 12 h of injury, which returned to normal by 7 days. In contrast, animals injected with APTRA-AM showed no significant change in the ADC at any time point studied. Tissue analysis showed that APTRA-AM significantly reduced the infarct size by 85% and extent of inflammatory cell infiltration by 94%. The results clearly demonstrate significant neuroprotection by APTRA-AM when given after injury.
Collapse
Affiliation(s)
- Brenda L Bartnik
- Department of Radiation Medicine, Loma Linda University, Radiobiology Program CSP A1010, Loma Linda, CA 92354, USA.
| | | | | |
Collapse
|
500
|
Mani N, Khaibullina A, Krum JM, Rosenstein JM. Astrocyte growth effects of vascular endothelial growth factor (VEGF) application to perinatal neocortical explants: receptor mediation and signal transduction pathways. Exp Neurol 2005; 192:394-406. [PMID: 15755557 DOI: 10.1016/j.expneurol.2004.12.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Revised: 12/03/2004] [Accepted: 12/10/2004] [Indexed: 01/19/2023]
Abstract
The non-angiogenic role of vascular endothelial growth factor (VEGF), and its receptors flt-1 and flk-1, together with downstream signaling pathways were examined in fetal and postnatal rat cerebral cortical organotypic explants. VEGF application in both paradigms caused a significant increase in astroglial proliferation and a dose-dependent increase in GFAP and nestin immunoreactivity. The VEGF receptor flt-1 was observed on most, though not all astrocytes, while flk-1 receptor immunoexpression was absent. Treatment with antisense oligonucleotides (AS-ODNs) to flt-1 resulted in a dramatic decrease in GFAP and nestin immunoreactivity, which further confirmed the role of flt-1 in mediating VEGF's gliotrophic effects, while AS-ODNs to flk-1 had no effect. VEGF-induced gliotrophic effects were found to be mediated by the MAPK/ERK and PI-3 kinase signaling pathways, since the both the MEK1 inhibitor, PD98059 and the PI-3 kinase inhibitor, Wortmannin abolished VEGF-induced astrocytic GFAP(+) expression. Although high dose VEGF application resulted in strong upregulation of both GFAP and nestin immunoreactivity in astrocytes, overlap of the two proteins was not observed in all cells, suggesting that some of the nestin(+) cells might be neural progenitors. Exposure to VEGF resulted in upregulation of both VEGF and bFGF mRNA at the one-day time point, and bFGF protein by 3 days; VEGF activated astrocytes expressed bFGF to a much greater degree than those in untreated explants. The increased expression of bFGF induced by VEGF, may serve in the proliferation of multipotential neural stem/progenitor cells in vitro. VEGF, an established angiogenic factor, appears to play a significant role in the growth and differentiation of astrocytes in the CNS.
Collapse
Affiliation(s)
- Nina Mani
- Department of Anatomy and Cell Biology, Ross Hall 205, The George Washington University Medical Center, 2300 I Street, NW Washington, DC 20037, USA
| | | | | | | |
Collapse
|