601
|
Dietrich PY, Walker PR, Saas P. Death receptors on reactive astrocytes: a key role in the fine tuning of brain inflammation? Neurology 2003; 60:548-54. [PMID: 12607528 DOI: 10.1212/01.wnl.0000042049.74547.7f] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Immune responses protect the CNS against pathogens. However, the fact that there is little dispensable tissue in the brain makes regulation necessary to avoid disastrous immune-mediated damage. Astrocytes respond vigorously to any brain injury (e.g., tumor, stroke, AD, MS, HIV) and are postulated to play an important role in the fine tuning of brain inflammation. The authors propose that astrocytes use death receptors to modulate pro- and anti-inflammatory effects.
Collapse
Affiliation(s)
- Pierre-Yves Dietrich
- Hôpital Universitaire, Division d'Oncologie, Laboratoire d'Immunologie des Tumeurs, Switzerland.
| | | | | |
Collapse
|
602
|
Hao HN, Zhao J, Thomas RL, Parker GC, Lyman WD. Fetal human hematopoietic stem cells can differentiate sequentially into neural stem cells and then astrocytes in vitro. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2003; 12:23-32. [PMID: 12662433 DOI: 10.1089/152581603321210109] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In some rodent models, there is evidence that hematopoietic stem cells (HSC) can differentiate into neural cells. However, it is not known whether humans share this potential, and, if so, what conditions are sufficient for this transdifferentiation to occur. We addressed this question by assessing the ability of fetal human liver CD34(+)/CD133(+)/CD3(-) hematopoietic stem cells to generate neural cells and astrocytes in culture. We cultured fetal liver-derived hematopoietic stem cells in human astrocyte culture-conditioned medium or using a method wherein growing human astrocytes were separated from cultured, nonadherent hematopoietic stem cells by a semipermeable membrane in a double-chamber co-culture system. Hematopoietic stem cell cultures were probed for neural progenitor cell marker expression (nestin and bone morphogenic protein-2 [BMP-2]) during growth in both culture conditions. RT-PCR, western blotting, and immunocytochemistry assays showed that cells cultured in either condition expressed nestin mRNA and protein and BMP-2 mRNA. HSC similarly cultured in nonconditioned medium or in the absence of astrocytes did not express either marker. Cells expressing these neural markers were transferred and cultured on poly-D-lysine-coated dishes with nonconditioned growth medium for further study. Immunocytochemistry demonstrated that these cells differentiated into astrocytes after 8 days in culture as indicated by their morphology and expression of the astrocytic markers glial fibrillary acidic protein (GFAP) and S100, as well as by their rate of proliferation, which was identical to that of freshly isolated fetal brain astrocytes. These findings demonstrate that neural precursor gene expression can be induced when human hematopoietic stem cells are exposed to a suitable microenvironment. Furthermore, the neural stem cells generated in this environment can then differentiate into astrocytes. Therefore, human hematopoietic stem cells may be an alternative resource for generation of neural stem cells for therapy of central nervous system defects resulting from disease or trauma.
Collapse
Affiliation(s)
- Hsiao-Nan Hao
- Department of Pediatrics, Wayne State University School of Medicine and Children's Research Center of Michigan, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
603
|
Enge M, Wilhelmsson U, Abramsson A, Stakeberg J, Kühn R, Betsholtz C, Pekny M. Neuron-specific ablation of PDGF-B is compatible with normal central nervous system development and astroglial response to injury. Neurochem Res 2003; 28:271-9. [PMID: 12608700 DOI: 10.1023/a:1022421001288] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Members of the PDGF family have multiple roles during embryogenesis and in a variety of pathological situations in the adult. One of the major sites of PDGF-B expression in adult mammals are postmitotic CNS neurons. Combined with reported neurotrophic and neuroprotective effects of exogenously administered PDGFs, this has led to the speculation that PDGF-B may have a role in CNS development, in maintenance, or in response to CNS injury. To test these hypotheses, we developed mice in which PDGF-B was ablated genetically in postmitotic neurons at sites where PDGF-B is normally expressed. We found that these mice develop to adulthood without apparent defects. We demonstrate PDGF-B expression in the postnatal mouse hippocampus and forebrain cortex. We show that neuron-specific knockout of PDGF-B does not influence the astroglial and angiogenic responses to injury in the hippocampus or forebrain cortex. We conclude that the role of neuron-derived PDGF-B remains obscure. A role for neuron-derived PDGF-B, if existing, might be redundant with other CNS growth factors. Alternatively, other and more specific analyses of CNS functions in the normal and injured states will be required to demonstrate such a role.
Collapse
Affiliation(s)
- Maria Enge
- Department of Medical Biochemistry, Göteborg University, Göteborg, Sweden
| | | | | | | | | | | | | |
Collapse
|
604
|
Affiliation(s)
- Gülgün Tezel
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Kentucky, USA
| | | |
Collapse
|
605
|
Asnaghi V, Gerhardinger C, Hoehn T, Adeboje A, Lorenzi M. A role for the polyol pathway in the early neuroretinal apoptosis and glial changes induced by diabetes in the rat. Diabetes 2003; 52:506-11. [PMID: 12540628 DOI: 10.2337/diabetes.52.2.506] [Citation(s) in RCA: 180] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We tested the hypothesis that the apoptosis of inner retina neurons and increased expression of glial fibrillary acidic protein (GFAP) observed in the rat after a short duration of diabetes are mediated by polyol pathway activity. Rats with 10 weeks of streptozotocin-induced diabetes and GHb levels of 16 +/- 2% (mean +/- SD) showed increased retinal levels of sorbitol and fructose, attenuation of GFAP immunostaining in astrocytes, appearance of prominent GFAP expression in Müller glial cells, and a fourfold increase in the number of apoptotic neurons when compared with nondiabetic rats. The cells undergoing apoptosis were immunoreactive for aldose reductase. Sorbinil, an inhibitor of aldose reductase, prevented all abnormalities. Intensive insulin treatment also prevented most abnormalities, despite reducing GHb only to 12 +/- 1%. Diabetic mice, known to have much lower aldose reductase activity in other tissues when compared with rats, did not accumulate sorbitol and fructose in the retina and were protected from neuronal apoptosis and GFAP changes in the presence of GHb levels of 14 +/- 2%. This work documents discrete cellular consequences of polyol pathway activity in the retina, and it suggests that activation of the pathway and "retinal neuropathy" require severe hyperglycemia and/or high activity of aldose reductase. These findings have implications for how to evaluate the role of the polyol pathway in diabetic retinopathy.
Collapse
Affiliation(s)
- Veronica Asnaghi
- Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | |
Collapse
|
606
|
Teismann P, Tieu K, Cohen O, Choi DK, Wu DC, Marks D, Vila M, Jackson-Lewis V, Przedborski S. Pathogenic role of glial cells in Parkinson's disease. Mov Disord 2003; 18:121-9. [PMID: 12539204 DOI: 10.1002/mds.10332] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of the dopaminergic neurons in the substantia nigra pars compacta (SNpc). The loss of these neurons is associated with a glial response composed mainly of activated microglial cells and, to a lesser extent, of reactive astrocytes. This glial response may be the source of trophic factors and can protect against reactive oxygen species and glutamate. Alternatively, this glial response can also mediate a variety of deleterious events related to the production of pro-oxidant reactive species, and pro-inflammatory prostaglandin and cytokines. We discuss the potential protective and deleterious effects of glial cells in the SNpc of PD and examine how those factors may contribute to the pathogenesis of this disease.
Collapse
Affiliation(s)
- Peter Teismann
- Neuroscience Research, Movement Disorder Division, Department of Neurology, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
607
|
Zawadzka M, Kaminska B. Immunosuppressant FK506 affects multiple signaling pathways and modulates gene expression in astrocytes. Mol Cell Neurosci 2003; 22:202-9. [PMID: 12676530 DOI: 10.1016/s1044-7431(03)00036-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Brain injury is often associated with proliferation and hypertrophic response of glial cells (reactive gliosis). We have previously reported immunosuppressant effects on survival of glioma cells and adult reactive astrocytes. In the present study, we demonstrate growth-inhibitory effect of FK506 on cortical astrocytes from newborn rats. FK506 inhibits Erk and PI-3K/Akt signaling, two crucial pro-survival pathways. The levels of phosphorylated Akt and p42/44 Erk decline in few hours after FK506 addition. Furthermore, in FK506-treated astrocyte cultures the levels of mRNA encoding PDGF, bFGF, and CNTF decreased. Downregulation of growth factor expression by FK506 may play a role in the inhibition of mitogenic/hypertrophic responses. FasL mRNA level was elevated and interaction of FasL with Fas receptor expressed in astrocytes may trigger cell death. Interestingly, expression of BDNF increased in a dose-dependent manner in FK506-treated astrocytes. Upregulation of BDNF mRNA and protein level in astrocytes exposed to FK506 may underlie neuroprotective action of FK506.
Collapse
Affiliation(s)
- Malgorzata Zawadzka
- Department of Cellular Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur Str., 02-093, Warsaw, Poland
| | | |
Collapse
|
608
|
Dawson J, Miltz W, Mir AK, Wiessner C. Targeting monocyte chemoattractant protein-1 signalling in disease. Expert Opin Ther Targets 2003; 7:35-48. [PMID: 12556201 DOI: 10.1517/14728222.7.1.35] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) has been implicated in many inflammatory and autoimmune diseases. The G-protein-coupled receptor CCR-2B is probably the most important MCP-1 receptor in vivo, and loss of MCP-1 effector function alone is sufficient to impair monocytic trafficking in inflammation models. MCP-1 signalling appears to be a relevant target, especially in rheumatoid arthritis (RA). In RA patients, MCP-1 is produced by synovial cells and infiltrating monocytes, plasma MCP-1 concentrations correlate with swollen joint count, and elevated serum MCP-1 concentrations were found in juvenile RA in patients with active disease. Modulation of MCP-1 signalling in experimental RA showed beneficial effects on inflammation and joint destruction. With respect to chronic neuroinflammation, a critical role for MCP-1 has been established in animal models for multiple sclerosis. In acute neuroinflammation, experimental evidence for a detrimental role of MCP-1 in stroke and excitotoxic injury has been found. Several selective small molecular weight CCR-2B antagonists and MCP-1-blocking antibodies have been described. The proof for the validity of targeting MCP-1 signalling in disease, however, has yet to be established in clinical trials.
Collapse
MESH Headings
- Alzheimer Disease/drug therapy
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antirheumatic Agents/pharmacology
- Antirheumatic Agents/therapeutic use
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/physiopathology
- Central Nervous System/drug effects
- Central Nervous System/injuries
- Chemokine CCL2/antagonists & inhibitors
- Chemokine CCL2/deficiency
- Chemokine CCL2/genetics
- Chemokine CCL2/physiology
- Drug Design
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Humans
- Inflammation/drug therapy
- Mice
- Mice, Inbred MRL lpr
- Mice, Knockout
- Models, Molecular
- Molecular Structure
- Multiple Sclerosis/drug therapy
- Obesity/drug therapy
- Receptors, CCR2
- Receptors, Chemokine/antagonists & inhibitors
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Janet Dawson
- Arthritis and Bone Metabolism Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | | |
Collapse
|
609
|
Dienel GA, Cruz NF, Ball K, Popp D, Gokden M, Baron S, Wright D, Wenger GR. Behavioral training increases local astrocytic metabolic activity but does not alter outcome of mild transient ischemia. Brain Res 2003; 961:201-12. [PMID: 12531487 DOI: 10.1016/s0006-8993(02)03945-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Functional neurological outcome after transient ischemia might be improved by timely therapeutic intervention. To determine if restorative behavioral therapy influences damage, improves task learning, or alters astrocyte metabolic activity after ischemia, rats (food-restricted to 85% of free-feeding weight) were (a) first trained to respond on one of two levers under a fixed-ratio 20 schedule of food presentation (FR20), then (b) subjected to sham manipulation of carotid arteries or 10 min ischemia by four-vessel occlusion, followed by (c) 4 days of operant testing or inactivity, (d) then all rats were tested under a FR20 lever reversal task for 4 weeks, and (e) 3 days after the last behavioral session astrocyte metabolism was assayed by local uptake of [2-14C]acetate. Mild loss of hippocampal neurons occurred in ischemic rats with or without training after ischemia. Glial fibrillary acidic protein-positive astrocytes were present in similar numbers throughout brains of sham control and ischemic rats. Mild ischemia did not impair learning, and no changes in FR20 reversal learning were detected in sham vs. ischemic rats. Net [14C]acetate uptake was unaffected by ischemia but [14C]acetate uptake increased 15-24% (P<0.05; n=12-15/group) in specific structures (caudate, primary motor and sensorimotor cortex, CA1 hippocampus, subcortical white matter) in the pooled groups of rats that had 4 days FR20 testing vs. inactivity before reversal learning. 'Behavioral therapy' (operant testing on the 4 days immediately following either sham manipulation or ischemia) did not alter ischemic outcome, but was associated with higher acetate utilization in regions involved in motor activities.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock 72205, USA.
| | | | | | | | | | | | | | | |
Collapse
|
610
|
Yamagata K, Tagami M, Torii Y, Takenaga F, Tsumagari S, Itoh S, Yamori Y, Nara Y. Sphingosine 1-phosphate induces the production of glial cell line-derived neurotrophic factor and cellular proliferation in astrocytes. Glia 2003; 41:199-206. [PMID: 12509810 DOI: 10.1002/glia.10180] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a platelet-derived bioactive sphingolipid that evokes a variety of biological responses. To understand the role of S1P in the central nervous system, we have examined the effect of S1P on the production of glial cell line-derived neurotrophic factor (GDNF) and growth regulation of cortical astrocytes from rat embryo. Moreover, we examined the possibility that the expression of GDNF is regulated differently in cultured astrocytes from the stroke-prone spontaneously hypertensive rat (SHRSP) than in those from Wistar kyoto rats (WKY). The mRNA expression was quantitated by RT-PCR based on the fluorescent TaqMan methodology. A new instrument capable of measuring fluorescence in real time was used to quantify gene amplification in astrocytes. GDNF protein was investigated by enzyme-linked immunosorbent assay. S1P induced the expression of GDNF mRNA and the production of GDNF protein in a dose-dependent manner in WKY astrocytes. Moreover, S1P increased cell numbers and induced the proliferation of astrocytes. In addition, the level of mRNA expression and protein production of GDNF was significantly lower in SHRSP than WKY astrocytes following exposure to S1P. These findings revealed that S1P augments GDNF protein production and cellular growth in astrocytes. Also, our results indicate that production in SHRSP astrocytes was attenuated in response to S1P compared with that observed in WKY. We conclude that S1P specifically triggers a cascade of events that regulate the production of GDNF and cell growth in astrocytes. Our results also suggest that the reduced expression of GDNF caused by S1P is a factor in the stroke proneness of SHRSP.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Division of Life Science, Graduate School of Integrated Science and Art, University of East Asia, Yamaguchi, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
611
|
Runquist M, Alonso G. Gabaergic signaling mediates the morphological organization of astrocytes in the adult rat forebrain. Glia 2003; 41:137-51. [PMID: 12509804 DOI: 10.1002/glia.10166] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Previous studies have provided evidence that the morphological organization of immature astrocytes is influenced by the inhibitory neuronal transmitter gamma amino-butyric acid (GABA). The present study was designed to determine whether the occurrence of differential organization of mature astrocytes throughout various regions of the adult brain is related to differential GABAergic signaling. For this we first used Western blotting and high-performance liquid chromatography to quantify the levels of the astrocytic protein glial fibrillary acidic protein (GFAP) and GABA, respectively, within the same tissue punches taken from different forebrain regions of the adult rat, as well as immunocytochemistry for GFAP, GABA, or glutamate decarboxylase to visualize the morphological organization of astrocytes and of GABAergic axons in these regions. These data indicate that GFAP and GABA contents are correlated throughout the different forebrain regions analyzed, and that the regions containing the highest densities in GABAergic terminals are those that contain astrocytes exhibiting the highest degree of stellation. Secondly, we chronically increased GABAergic signaling in vivo by the systemic administration of an inhibitor of GABA transaminase or by the intracerebroventricular infusion of muscimol, a potent agonist of GABA(A) receptors. Our data show that in both cases, the GFAP content of the different forebrain regions is significantly augmented, in close association with a marked increase in the number of astrocytic processes and with their degree of branching. Taken together, these data strongly suggest that GABAergic signaling mediates the morphological organization of astrocytes and their expression of GFAP in the adult brain.
Collapse
|
612
|
Przedborski S, E. Goldman J. Pathogenic role of glial cells in Parkinson's disease. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1569-2558(03)31043-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
613
|
Kálmán M. Glial reaction and reactive glia. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1569-2558(03)31035-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
614
|
Abstract
Enteric glial cells (EGCs) represent an extensive but relatively poorly described cell population within the gastrointestinal tract. Accumulating data suggest that EGCs represent the morphological and functional equivalent of CNS astrocytes within the enteric nervous system (ENS). The EGC network has trophic and protective functions toward enteric neurons and is fully implicated in the integration and the modulation of neuronal activities. Moreover, EGCs seem to be active elements of the ENS during intestinal inflammatory and immune responses, sharing with astrocytes the ability to act as antigen-presenting cells and interacting with the mucosal immune system via the expression of cytokines and cytokine receptors. Transgenic mouse systems have demonstrated that specific ablation of EGC by chemical ablation or autoimmune T-cell targeting induces an intestinal pathology that shows similarities to the early intestinal immunopathology of Crohn's disease. EGCs may also share with astrocytes the ability to regulate tissue integrity, thereby postulating that similar interactions to those observed for the blood-brain barrier may also be partly responsible for regulating mucosal and vascular permeability in the gastrointestinal tract. Disruption of the EGC network in Crohn's disease patients may represent one possible cause for the enhanced mucosal permeability state and vascular dysfunction that are thought to favor mucosal inflammation.
Collapse
Affiliation(s)
- Julie Cabarrocas
- Institut National de la Santé et de la Recherche Médicale U546, Pitié-Salpêtrière Hospital, Paris, France
| | | | | |
Collapse
|
615
|
Function and dysfunction of enteric glia. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1569-2558(03)31014-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
616
|
de Freitas MS, Spohr TCLS, Benedito AB, Caetano MS, Margulis B, Lopes UG, Moura-Neto V. Neurite outgrowth is impaired on HSP70-positive astrocytes through a mechanism that requires NF-kappaB activation. Brain Res 2002; 958:359-70. [PMID: 12470872 DOI: 10.1016/s0006-8993(02)03682-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the adult central nervous system (CNS), prominent reactive astrocytosis is seen in acute traumatic brain injury, neurodegenerative diseases and a variety of viral infections. Reactive astrocytes synthesize a number of factors that could play different roles in neuronal regeneration. In this study, the effects of thermal stress were evaluated on nuclear factor-kappaB (NF-kappaB) activation and proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) secretion in primary astrocytic cultures. The ability of HSP70-positive astrocytes to support or inhibit neurite outgrowth was investigated in neuron-astrocyte cocultures. Cultured astrocytes from cerebral cortex of rats were exposed to transient hyperthermia (42 degrees C/30 min) and incubated at 37 degrees C for different periods of recovery. During HSP70 accumulation, astrocytes extended large and thick processes associated to rearrangement of glial fibrillary acidic protein (GFAP) filaments and an increase in protein synthesis and GFAP, suggesting an astrogliosis event. A delay of NF-kappaB activation appeared closely related to TNF-alpha secretion by HSP70-positive astrocytes. These cells demonstrated a functional shift from neurite growth-promoting to non-permissive substrate. We also found that gliotoxin, a specific NF-kappaB inhibitor, partially abrogated the inhibitory ability of reactive astrocytes. These findings may suggest a involvement of NF-kappaB and TNF-alpha in modulating the failure of HSP70-positive astrocytes to provide functional support to neuritic outgrowth.
Collapse
Affiliation(s)
- Marta S de Freitas
- Departamento de Farmacologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, RJ, Brazil
| | | | | | | | | | | | | |
Collapse
|
617
|
Segovia J. Gene therapy for Parkinson's disease: current status and future potential. AMERICAN JOURNAL OF PHARMACOGENOMICS : GENOMICS-RELATED RESEARCH IN DRUG DEVELOPMENT AND CLINICAL PRACTICE 2002; 2:135-46. [PMID: 12083948 DOI: 10.2165/00129785-200202020-00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Parkinson's disease appears to be a good candidate for gene therapy. The primary biochemical defect associated with the disease has been clearly determined as an absence of dopamine in the caudate-putamen, and the anatomical region where the neuropathologic hallmark of the disease, death of the nigral dopamine-producing neurons, occurs, remains circumscribed. Based on the biochemical and anatomical information gathered on Parkinson's disease, different gene therapy strategies have been devised to treat it. The first, and most explored strategy so far, consists in engineering cells to produce levodopa or dopamine so they will replace dopaminergic neurotransmission. Several types of cells have been employed in these experiments, and behavioral recovery has been reported in animal models of the disease. However, this approach cannot prevent neuronal death, nor reconstruct brain circuits. Another strategy is to protect cells by transferring genes that encode neurotrophic factors. Effort is now being concentrated into this research area, and promising results have recently been reported. Finally, an additional strategy aims at generating cells with a dopaminergic phenotype so they will be capable of replacing the missing dopaminergic neurons in biochemical, anatomical and functional terms. This has the potential to become an important constituent for an effective cure. Gene therapy holds significant promise for the treatment of neurodegenerative disorders, and Parkinson's disease treatment will benefit greatly from the knowledge and information arising from gene therapy research.
Collapse
Affiliation(s)
- José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Distrito Federal, Mexico.
| |
Collapse
|
618
|
Corcoran J, So PL, Maden M. Absence of retinoids can induce motoneuron disease in the adult rat and a retinoid defect is present in motoneuron disease patients. J Cell Sci 2002; 115:4735-41. [PMID: 12432062 DOI: 10.1242/jcs.00169] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We generated retinoid-deficient adult rats by the removal of retinoids from their diet. We show that their motoneurons undergo neurodegeneration and that there is an accumulation of neurofilaments and an increase in astrocytosis, which is associated with motoneuron disease. These effects are mediated through the retinoic acid receptor alpha. The same receptor deficit is found in motoneurons from patients suffering from spontaneous amyotrophic lateral sclerosis. Furthermore, we show that there is a loss of expression of the retinaldehyde dehydrogenase enzyme II in motoneurons. Therefore, we propose that a defect in the retinoid signalling pathway is in part be responsible for some types of motoneuron disease.
Collapse
Affiliation(s)
- Jonathan Corcoran
- MRC Centre for Developmental Neurobiology, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK.
| | | | | |
Collapse
|
619
|
Saravia FE, Revsin Y, Gonzalez Deniselle MC, Gonzalez SL, Roig P, Lima A, Homo-Delarche F, De Nicola AF. Increased astrocyte reactivity in the hippocampus of murine models of type 1 diabetes: the nonobese diabetic (NOD) and streptozotocin-treated mice. Brain Res 2002; 957:345-53. [PMID: 12445977 DOI: 10.1016/s0006-8993(02)03675-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Diabetes can be associated with cerebral dysfunction in humans and animal models of the disease. Moreover, brain anomalies and alterations of the neuroendocrine system are present in type 1 diabetes (T1D) animals, such as the spontaneous nonobese diabetic (NOD) mouse model and/or the pharmacological streptozotocin (STZ)-induced model. Because of the prevalent role of astrocytes in cerebral glucose metabolism and their intimate connection with neurones, we investigated hippocampal astrocyte alterations in prediabetic and diabetic NOD mice and STZ-treated diabetic mice. The number and cell area related to the glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes were quantified in the stratum radiatum region of the hippocampus by computerized image analysis in prediabetic (2, 4 and 8 weeks of age) and diabetic (16-week-old) NOD female mice, age and sex-matched lymphocyte-deficient NODscid and C57BL/6 control mice and, finally, STZ-induced diabetic and vehicle-treated nondiabetic 16-week-old C57BL/6 female mice. Astrocyte number was higher early in life in prediabetic NOD and NODscid mice than in controls, when transient hyperinsulinemia and low glycemia were found in these strains. The number and cell area of GFAP(+) cells further increased after the onset of diabetes in NOD mice. Similarly, in STZ-treated diabetic mice, the number of GFAP(+) cells and cell area were higher than in vehicle-treated mice. In conclusion, astrocyte changes present in genetic and pharmacological models of T1D appear to reflect an adaptive process to alterations of glucose homeostasis.
Collapse
Affiliation(s)
- Flavia E Saravia
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologa y Medicina Experimental, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
620
|
Wu DC, Tieu K, Cohen O, Choi DK, Vila M, Jackson-Lewis V, Teismann P, Przedborski S. Glial cell response: A pathogenic factor in Parkinson's disease. J Neurovirol 2002; 8:551-8. [PMID: 12476349 DOI: 10.1080/13550280290100905] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The loss of these neurons is associated with a glial response composed mainly of activated microglial cells and, to a lesser extent, of reactive astrocytes. This glial response may be the source of trophic factors and can protect against reactive oxygen species and glutamate. Alternatively, this glial response can also mediate a variety of deleterious events related to the production of pro-oxidant reactive species, proinflammatory prostaglandin, and cytokines. In this review, the authors discuss the potential protective and deleterious effects of glial cells in the SNpc of PD and examine how these factors may contribute to the pathogenesis of this disease.
Collapse
Affiliation(s)
- Du Chu Wu
- Neuroscience Research, Movement Disorder Division, Department of Neurology, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
621
|
Nikaido T, Iseki K, Mori T, Takaki H, Yokoya S, Hagino S, Takeda J, Zhang Y, Takeuchi M, Kikuchi SI, Wanaka A. Expression of OASIS, a CREB/ATF family transcription factor, in CNS lesion and its transcriptional activity. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 108:129-38. [PMID: 12480185 DOI: 10.1016/s0169-328x(02)00521-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We reported the expression patterns of a novel member of the CREB/ATF family, OASIS, in central nervous system (CNS) lesions and its transcriptional activity. OASIS gene expression was upregulated in the stab-injured spinal cord. Double labeling experiments revealed that the distribution of OASIS mRNA-positive cells overlapped with a population of GFAP-immunoreactive cells. This finding suggested that OASIS might regulate expression of important downstream molecules in certain subset of the reactive astrocytes (e.g. inhibitory substances in injured brain). In gel shift assays, OASIS was able to specifically bind to CRE as CREB family members were. We then examined transcriptional activity of full-length OASIS with GAL4-UAS-luciferase reporter assay in COS7 cells. OASIS protein activated transcription, but did not inhibit basal transcription driven by AdML promoter. To determine critical portion(s) of the OASIS protein in transcriptional activation, we examined the activity of various deletion constructs of OASIS gene. The assay revealed that a strong transcriptional activation domain lay in the N-terminal region where acidic amino acids clustered and a possible repression domain, which had not been reported for other CREB/ATF family members, lay in the more C-terminal region. We therefore proposed that OASIS protein positively regulated gene transcription in a subset of reactive astrocytes, and thereby influenced the reaction of injured CNS tissues.
Collapse
Affiliation(s)
- Takuya Nikaido
- Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
622
|
Bal-Price A, Moneer Z, Brown GC. Nitric oxide induces rapid, calcium-dependent release of vesicular glutamate and ATP from cultured rat astrocytes. Glia 2002; 40:312-23. [PMID: 12420311 DOI: 10.1002/glia.10124] [Citation(s) in RCA: 172] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Nitric oxide (NO; 1 microM) or an NO donor (500 microM diethylenetriamine-nitric oxide, DETA-NONOate) caused rapid glutamate and ATP release from cultured rat cortical astrocytes. NO-induced glutamate release was prevented by calcium chelators (EGTA or BAPTA-AM) and an inhibitor of vesicular exocytosis (botulinum neurotoxin C, BoTx-C), but not by a glutamate transport inhibitor, L-trans-pyrrolidine-2,4-dicarboxylate (t-PDC), a cyclooxygenase inhibitor (indomethacin), or an inhibitor of soluble guanylate cyclase 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ), and was not induced by mitochondrial respiratory inhibitors (myxothiazol or azide). Similarly to glutamate, NO-induced ATP release was also completely blocked by BAPTA-AM and BoTx-C, suggesting again a vesicular, calcium-dependent mechanism of release. Addition of DETA-NONOate (500 microM) to fura-2-loaded astrocytes induced a rapid, transient increase in intracellular calcium levels followed by a lower, sustained level of calcium entry. The latter was blocked by gadolinium (1 microM), an inhibitor of capacitative Ca(2+) entry. Thus, NO appears to cause rapid exocytosis of vesicular glutamate and ATP from astrocytes by raising intracellular calcium levels. Astrocytes activated by lipopolysaccharide/endotoxin and interferon-gamma to express inducible NO synthase (iNOS) maintained substantially higher extracellular glutamate levels than nonactivated cells or activated cells treated with an iNOS inhibitor (1400W), but the rate of glutamate uptake by these cells was similar. This suggests that NO from inflammatory-activated astrocytes causes release of astrocytic glutamate. NO-induced release of astrocytic glutamate and ATP may be important in physiological or pathological communication between astrocytes and neurons.
Collapse
Affiliation(s)
- Anna Bal-Price
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| | | | | |
Collapse
|
623
|
Venero JL, Santiago M, Tomás-Camardiel M, Matarredona ER, Cano J, Machado A. DCG-IV but not other group-II metabotropic receptor agonists induces microglial BDNF mRNA expression in the rat striatum. Correlation with neuronal injury. Neuroscience 2002; 113:857-69. [PMID: 12182892 DOI: 10.1016/s0306-4522(02)00232-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We have previously described a neuroprotective action of (2S,2'R,3'R)-2-(2'3'-dicarboxycyclopropyl)glycine (DCG-IV), an agonist for group-II metabotropic receptors, on dopaminergic nerve terminals against the degeneration induced by 1-methyl-4-phenylpyridinium (MPP+). This effect was accompanied by an up-regulation of brain-derived neurotrophic factor (BDNF) mRNA expression in the rat striatum. We have now analyzed the phenotypic nature of the BDNF mRNA-expressing cells in response to intrastriatal injection of DCG-IV. Dual in situ hybridization and immunohistochemistry revealed that microglial cells but not astrocytes were responsible for this induction. Subsequent analysis demonstrated that this effect was accompanied by striking loss of striatal glutamic acid decarboxylase (GAD) mRNA and massive appearance of internucleosomal DNA fragmentation, a hallmark of apoptosis. A dose-response study demonstrated that doses of DCG-IV as low as 5 nmol was very toxic in terms GAD mRNA and apoptosis. 0.5 nmol of DCG-IV did not induce toxicity at all in terms of GAD mRNA and apoptosis. Activation of group-II metabotropic receptors in striatum with N-Acetyl-Asp-Glu (NAAG; a mGlu3 agonist) and (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate (a mGlu2 and mGlu3 agonist) did not induce neither loss of GAD mRNA nor appearance of apoptosis (doses up to 20 nmol). In additional experiments, NAAG, in contrast to DCG-IV, failed to protect the striatal dopaminergic system against the degeneration induced by MPP+ as studied by microdialysis. Finally, we studied the mechanism by which DCG-IV is highly toxic. For that, selective antagonists of either metabotropic--(R,S)-alpha-methyl-4-carboxyphenylglycine and LY 341495--or ionotropic (N-methyl-D-aspartate, NMDA)--DL-2-amino-5-phosphonovaleric acid (AP-5) glutamate receptors --were co-administered with DCG-IV. Only AP-5 highly protected the striatum against the degeneration induced by DCG-IV. Since DCG-IV also activates the NMDA receptor at concentrations higher than 3 microM, it is conceivable that a intrastriatal concentration equal or higher than 3 microM after a single striatal injection of 5-20 nmol of DCG-IV. Our findings suggest that much caution must be exerted when testing the numerous neuroprotective effects ascribed to group-II metabotropic receptor activation, in particular when using DCG-IV. We conclude that the neuroprotectant capability of a given compound on a specific system does not exclude the possibility of inducing toxicity on a different one.
Collapse
Affiliation(s)
- J L Venero
- Departamento de Bioquímica, Bromatología y Toxicología, Facultad de Farmacia, Universidad de Sevilla, C/Prof. García González s/n, 41012 Sevilla, Spain
| | | | | | | | | | | |
Collapse
|
624
|
Bates KA, Fonte J, Robertson TA, Martins RN, Harvey AR. Chronic gliosis triggers Alzheimer's disease-like processing of amyloid precursor protein. Neuroscience 2002; 113:785-96. [PMID: 12182886 DOI: 10.1016/s0306-4522(02)00230-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease is a progressively dementing illness characterized by the extracellular accumulation and deposition of beta-amyloid. Early onset Alzheimer's disease is linked to mutations in three genes, all of which lead to increased beta-amyloid production. Inflammatory changes and gliosis may also play a role in the disease process, but the importance of these reactive events remains unclear. We recently reported that chronic cortical gliosis in heterotopic fetal rat cortical transplants is associated with significant changes in the levels of some of the proteins implicated in the pathogenesis of Alzheimer's disease. Because rodent beta-amyloid does not form extracellular amyloid deposits, we have now extended this model of chronic cortical gliosis to transgenic mice expressing the Swedish mutant form of human amyloid precursor protein. In addition, apolipoprotein E knockout mice were used to elucidate the role of this protein in reactive gliosis. The expression of mutant and murine proteins was assayed 6 or 10 months after transplantation using immunohistochemical and western blot methods. Heterotopic transplantation of fetal cortex onto the midbrain of neonatal mice consistently resulted in reactive gliosis, independent of apolipoprotein E status. In contrast, in homotopic cortex-to-cortex grafts there was little alteration in glial reactivity, a result similar to that obtained previously in rats. By 10 months post-transplantation the level of presenilin-1 expression was lower in heterotopic grafts than in host cortex and there was increased expression of transgenic amyloid precursor protein, but only in the gliotic cortex-to-midbrain grafts. Most importantly, increased levels of beta-amyloid, and particularly its precursor, C-99, were selectively found in these heterotopic transplants. Our results show that chronic gliosis is associated with altered processing of the amyloid precursor protein in vivo and thus may initiate or exacerbate pathological changes associated with Alzheimer's disease.
Collapse
Affiliation(s)
- K A Bates
- School of Anatomy and Human Biology, The University of Western Australia, Nedlands, WA 6009, Australia
| | | | | | | | | |
Collapse
|
625
|
Veznedaroglu E, Van Bockstaele EJ, O'Connor MJ. Extravascular collagen in the human epileptic brain: a potential substrate for aberrant cell migration in cases of temporal lobe epilepsy. J Neurosurg 2002; 97:1125-30. [PMID: 12450035 DOI: 10.3171/jns.2002.97.5.1125] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Several lines of evidence have demonstrated a number of cellular changes that occur within the hippocampus in patients with temporal lobe epilepsy (TLE). These include aberrant migration of granule cells and sprouting of mossy fibers, processes that have been linked to the hyperexcitability phenomenon observed in cases of TLE. In the present study the authors examined brain tissues obtained in patients undergoing temporal lobectomy surgery and in patients at autopsy (normal human control specimens), and compared the subcellular composition of regions of the hippocampus containing dispersed granule cells. METHODS Six human hippocampi were obtained in patients undergoing temporal lobectomies for intractable seizures. The patients ranged in age from 24 to 50 years. Two of the six patients had a history of head trauma and one had experienced a febrile seizure during childhood. Immediately following excision from the brain, the tissue was placed in an acrolein-paraformaldehyde fixative. The hippocampi were processed along with six human brain control specimens obtained at autopsy for light and electron microscopic evaluation. The tissues were then labeled for collagen types I through IV. Positive collagen labeling was identified, with the aid of both light and electron microscopy, in the parenchyma of all patients with TLE but not in the control tissues. CONCLUSIONS The authors report the first localization of collagen outside of the vasculature and meninges in the brains of patients with TLE. Recent evidence of collagen's chemoattractant properties and its role in epileptogenesis in animal models suggests that collagen may play a role in cellular migration and seizure activity in a subset of patients. Further studies with a larger series of patients are warranted.
Collapse
Affiliation(s)
- Erol Veznedaroglu
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | | | | |
Collapse
|
626
|
Abstract
Cytokines constitute a significant portion of the immuno- and neuromodulatory messengers that can be released by activated microglia. By virtue of potent effects on resident and invading cells, microglial cyto- and chemokines regulate innate defense mechanisms, help the initiation and influence the type of immune responses, participate in the recruitment of leukocytes to the CNS, and support attempts of tissue repair and recovery. Microglia can also receive cyto- and chemokine signals as part of auto- and paracrine communications with astrocytes, neurons, the endothelium, and leukocyte infiltrates. Strong responses and modulatory influences can be demonstrated, adding to the emerging view that microglial behavior is highly dependent on the (cytokine) environment and that reactions to a challenge may vary with the stimulation context. In principle, microglial activation aims at CNS protection. However, failed microglial engagement due to excessive or sustained activation could significantly contribute to acute and chronic neuropathologies. Dysregulation of microglial cytokine production could thereby promote harmful actions of the defense mechanisms, result in direct neurotoxicity, as well as disturb neural cell functions as they are sensitive to cytokine signaling.
Collapse
Affiliation(s)
- Uwe-Karsten Hanisch
- Department of Cellular Neurosciences, Max Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- University of Applied Sciences Lausitz, Senftenberg, Germany
| |
Collapse
|
627
|
Magnaghi V, Veber D, Morabito A, Buccellato FR, Melcangi RC, Scalabrino G. Decreased GFAP-mRNA expression in spinal cord of cobalamin-deficient rats. FASEB J 2002; 16:1820-2. [PMID: 12354698 DOI: 10.1096/fj.02-0231fje] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have demonstrated previously that chronic vitamin B12 [cobalamin (Cbl)] deficiency preferentially affects glial cells in the rat central nervous system (CNS) and severely affects peripheral glial cells independently of and concomitantly with the central neuropathy. In this study, we determined the mRNA levels for myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) in different CNS areas of rats made Cbl-deficient by total gastrectomy, as well as the mRNA levels for glycoprotein Po and peripheral myelin protein (PMP)22 in the sciatic nerve. GFAP-mRNA levels were significantly decreased in the spinal cord (SC) and hypothalamus, but not in the cortex, hippocampus, or striatum of totally gastrectomized (TGX) rats. No differences in GFAP protein levels were found in the SC and hypothalamus of the TGX rats treated or not with Cbl. MBP-mRNA levels were significantly decreased only in the hypothalamus, and the levels of mRNA for both glial markers returned to normal with Cbl replacement therapy. The levels of mRNA for the various myelin proteins in the sciatic nerve were not modified by Cbl deficiency. These results demonstrate that: a) the neurotrophic action of Cbl in rat CNS occurs in a zonal manner; and b) Cbl deficiency does not affect myelin synthesis (with the sole exception of the hypothalamus).
Collapse
Affiliation(s)
- Valerio Magnaghi
- Institute of General Pathology, University of Milan, 20133 Milan, Italy
| | | | | | | | | | | |
Collapse
|
628
|
Yun SJ, Kim MO, Kim SO, Park J, Kwon YK, Kim IS, Lee EH. Induction of TGF-beta-inducible gene-h3 (betaig-h3) by TGF-beta1 in astrocytes: implications for astrocyte response to brain injury. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 107:57-64. [PMID: 12414124 DOI: 10.1016/s0169-328x(02)00447-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Transforming growth factor (TGF)-beta-inducible gene-h3 (betaig-h3) product is a secreted protein that is induced by TGF-beta in several cell types and implicated in various tissue pathologies. The aims of this study were to determine the effect of TGF-beta1 on betaig-h3 expression in cultured astrocytes and to examine whether betaig-h3 is expressed in the brain after traumatic injury. The results showed that betaig-h3 mRNA and protein increased in response to TGF-beta1 in U87 human astrocytoma cells and mouse cortical astrocytes. Treatment with other cytokines, including tumor necrosis factor-alpha and fibroblast growth factor-2, did not enhance the expression of betaig-h3 in astrocytes. betaig-h3 was significantly expressed in reactive astrocytes at the site of a stab wound in the cerebral cortex of adult rats. These results provide an insight into understanding a novel role for betaig-h3 protein in the response of astrocytes to brain injury.
Collapse
Affiliation(s)
- Su-Jin Yun
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-In 449-701, South Korea
| | | | | | | | | | | | | |
Collapse
|
629
|
Saas P, Walker PR, Quiquerez AL, Chalmers DE, Arrighi JF, Liénard A, Boucraut J, Dietrich PY. A self-defence mechanism of astrocytes against Fas-mediated death involving interleukin-8 and CXCR2. Neuroreport 2002; 13:1921-4. [PMID: 12395092 DOI: 10.1097/00001756-200210280-00018] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Astrocytes play multiple roles from passive support to the regulation of inflammation during brain injury. This latter function is in part achieved by responses induced by the triggering of Fas expressed on astrocytes both and. It was previously shown that astrocytes are resistant to Fas-mediated death, responding to Fas triggering by interleukin-8 production. However, the cellular mechanisms by which astrocytes protect themselves from Fas-mediated death are unclear. Here, we show that survival of cultured astrocytes after Fas triggering is governed by the interaction of interleukin-8 with one of its receptors, CXCR2. Furthermore, interleukin-8 secretion and CXCR2 expression are both induced in human astrocytes after Fas stimulation, suggesting a new mechanism of self-defence against Fas-mediated death.
Collapse
MESH Headings
- Anti-Bacterial Agents/pharmacology
- Antibodies, Monoclonal
- Apoptosis/drug effects
- Apoptosis/physiology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Cell Survival/drug effects
- Cell Survival/physiology
- Cells, Cultured
- Encephalitis/metabolism
- Encephalitis/physiopathology
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Homeostasis/drug effects
- Homeostasis/physiology
- Humans
- Interleukin-8/metabolism
- Polymyxin B/pharmacology
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptors, Interleukin-8A/antagonists & inhibitors
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Up-Regulation/drug effects
- Up-Regulation/physiology
- fas Receptor/metabolism
- fas Receptor/pharmacology
Collapse
Affiliation(s)
- Philippe Saas
- Laboratory of Tumor Immunology, Division of Oncology, University Hospital, Rue Micheli-du-Crest 24, 1211 Geneva 14, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
630
|
Baydas G, Reiter RJ, Nedzvetskii VS, Nerush PA, Kirichenko SV. Altered glial fibrillary acidic protein content and its degradation in the hippocampus, cortex and cerebellum of rats exposed to constant light: reversal by melatonin. J Pineal Res 2002; 33:134-9. [PMID: 12220326 DOI: 10.1034/j.1600-079x.2002.02110.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Reactive astrocytosis is a well-known phenomenon that occurs rapidly after physical or metabolic injury to the brain. One of the important events during astrocyte differentiation is the increased expression of glial fibrillary acidic protein (GFAP), a member of the family of intermediate filament structural proteins. Free radicals are neurotoxic and free radical scavengers have been shown to protect the brain against neurotoxic damage. In the present study, we examined the effect of melatonin on astrocytic reactivity by determining the expression of the glial marker, GFAP, in different brain regions. Rats were exposed to constant light to reduce endogenous melatonin production; half of the animals were injected with melatonin during the exposure to constant light for 7 days. Western blots showed increases in total and degraded GFAP content in the brain of rats exposed to constant light. Melatonin administration caused a reduction of degraded GFAP content. In addition, melatonin significantly reduced neural tissue lipid peroxidation while constant light significantly enhanced the breakdown of lipids in the brain. Brain glutathione levels decreased significantly as a result of constant light exposure; this reduction was reversed by melatonin administration. These results suggest that melatonin potentially protects both neurons and glial cells from free radicals; melatonin's protective actions are probably related to the antioxidant properties of the indole.
Collapse
Affiliation(s)
- Giyasettin Baydas
- Department of Physiology, College of Medicine, Firat University, Elazig, Turkey.
| | | | | | | | | |
Collapse
|
631
|
Anderson E, Zink W, Xiong H, Gendelman HE. HIV-1-associated dementia: a metabolic encephalopathy perpetrated by virus-infected and immune-competent mononuclear phagocytes. J Acquir Immune Defic Syndr 2002; 31 Suppl 2:S43-54. [PMID: 12394782 DOI: 10.1097/00126334-200210012-00004] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Infection of the nervous system by HIV-1 commonly causes a broad range of cognitive, behavioral, and motor abnormalities called, in its most severe form, HIV-1-associated dementia (HAD). HAD is a metabolic encephalopathy caused by productive viral infection of brain mononuclear phagocytes (MPs) (perivascular and parenchymal brain macrophages and microglia) and sustained by paracrine-amplified, inflammatory, neurotoxic responses. MP neurotoxins are, in large measure, homeostatic secretory products that can have a negative effect on neuronal cell function when produced in abundance. Proinflammatory cytokines, chemokines, platelet-activating factor, arachidonic acid and its metabolites, nitric oxide, quinolinic acid, progeny virions, and viral structural and regulatory proteins are all included as part of these cellular and viral toxic elements. In addition, neuronal damage can occur directly by engaging specific receptors or through inducing widespread inflammatory activities in brain tissue that ultimately induce neuronal demise. The mechanisms for immune-and viral-mediated neural injury in HAD are made more striking by the effects of abused drugs on cognitive function. Ultimately, linkages between neuronal function and disordered MP immunity will provide insights into how HIV-1 infection of the brain leads to compromised mental function as well as providing clues into the pathogenesis of other neurodegenerative disorders.
Collapse
Affiliation(s)
- Eric Anderson
- The Center for Neurovirology and Neurodegenerative Disorders and Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha 68198-5215, USA
| | | | | | | |
Collapse
|
632
|
Proescholdt MG, Chakravarty S, Foster JA, Foti SB, Briley EM, Herkenham M. Intracerebroventricular but not intravenous interleukin-1beta induces widespread vascular-mediated leukocyte infiltration and immune signal mRNA expression followed by brain-wide glial activation. Neuroscience 2002; 112:731-49. [PMID: 12074914 DOI: 10.1016/s0306-4522(02)00048-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interleukin-1beta (IL-1beta) is a pro-inflammatory cytokine that appears in brain and cerebrospinal fluid following peripheral immune challenges and central infections or injury. We examined the consequences of i.c.v. infusion of IL-1beta on mRNA expression of several immune markers and on recruitment of peripheral leukocytes. Awake rats were infused with IL-1beta (100 ng/rat) into the lateral ventricle, and 0.5, 2, 4, 8, 12, or 24 h later, animals were killed and their fresh-frozen brains processed for in situ hybridization and immunohistochemistry. Widespread vascular expression of inhibitory factor kappa(B)alpha (Ikappa(B)alpha, marker of nuclear factor kappa(B)alpha transcriptional activity) and inducible cyclooxygenase (COX-2) mRNAs at 0.5-2 h was credited to movement of IL-1beta along ventricular, subarachnoid, and perivascular pathways to target endothelia that express type 1 IL-1 receptor mRNA. Induction of monocyte chemoattractant protein-1 mRNA and intercellular adhesion molecule-1 (ICAM-1) immunostaining on endothelia began at 0.5-2 h. Leukocytes (neutrophils and monocytes, recognized by morphology and CD45 and ED1 immunostaining) appeared in meninges and blood vessels at 2-4 h and diffusely penetrated the parenchyma at 8-24 h. The leukocytes strongly expressed IL-1beta and inducible nitric oxide synthase mRNAs. Beginning at 4-12 h, astrocytes (glial acidic fibrillary protein mRNA and protein and c-fos mRNA) and microglia (ionized calcium-binding adaptor molecule 1 mRNA and protein) showed widespread activation. Other rats received i.v. IL-1beta (6 microg/kg). Their brains showed induction of Ikappa(B)alpha and COX-2 mRNAs in the vasculature at 2 h but none of the other sequelae. In summary, our data indicate that IL-1beta in the cerebrospinal fluid reaches its target receptors on the endothelia via perivascular volume transmission, up-regulates ICAM-1, and triggers a targeted leukocyte emigration and widespread glial activation stimulated perhaps by pro-inflammatory molecules expressed by leukocytes. The dramatic difference between i.c.v. and i.v. routes of administration underscores the potency of IL-1beta within the brain to dynamically affect the cellular trafficking component of 'immune privilege'.
Collapse
Affiliation(s)
- M G Proescholdt
- Section on Functional Neuroanatomy, National Institute of Mental Health, Bethesda, MD 20892-4070, USA
| | | | | | | | | | | |
Collapse
|
633
|
García-Ovejero D, Veiga S, García-Segura LM, Doncarlos LL. Glial expression of estrogen and androgen receptors after rat brain injury. J Comp Neurol 2002; 450:256-71. [PMID: 12209854 DOI: 10.1002/cne.10325] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Estrogens and androgens can protect neurons from death caused by injury to the central nervous system. Astrocytes and microglia are major players in events triggered by neural lesions. To determine whether glia are direct targets of estrogens or androgens after neural insults, steroid receptor expression in glial cells was assessed in two different lesion models. An excitotoxic injury to the hippocampus or a stab wound to the parietal cortex and hippocampus was performed in male rats, and the resultant expression of steroid receptors in glial cells was assessed using double-label immunohistochemistry. Both lesions induced the expression of estrogen receptors (ERs) and androgen receptors (ARs) in glial cells. ERalpha was expressed in astrocytes immunoreactive (ERalpha-ir) for glial fibrillary acidic protein or vimentin. AR immunoreactivity colocalized with microglial markers, such as Griffonia simplicifolia lectin-1 or OX-6. The time course of ER and AR expression in glia was studied in the stab wound model. ERalpha-ir astrocytes and AR-ir microglia were observed 3 days after lesion. The number of ERalpha-ir and AR-ir glial cells reached a maximum 7 days after lesion and returned to low levels by 28 days postinjury. The studies of ERbeta expression in glia were inconclusive; different results were obtained with different antibodies. In sum, these results suggest that reactive astrocytes and reactive microglia are a direct target for estrogens and androgens, respectively.
Collapse
Affiliation(s)
- Daniel García-Ovejero
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | | | | | | |
Collapse
|
634
|
Probst-Cousin S, Kowolik D, Kuchelmeister K, Kayser C, Neundörfer B, Heuss D. Expression of annexin-1 in multiple sclerosis plaques. Neuropathol Appl Neurobiol 2002; 28:292-300. [PMID: 12175341 DOI: 10.1046/j.1365-2990.2002.00396.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This study describes the distribution and identity of annexin-1 positive cells in the central nervous system in patients with multiple sclerosis (MS). Glucocorticoid-inducible, anti-inflammatory properties have been ascribed to annexin-1, a member of a family of calcium-binding proteins that are referred to collectively as annexins. We have found annexin-1 to be spatially associated with active MS lesions and demonstrated a stage-dependent expression of annexin-1 in MS plaques. All of the most important pathogenetically involved cells of MS lesions showed a strong annexin-1 reactivity. Both correlation analysis and double staining procedures suggested annexin-1 expression in macrophages and perivascular lymphocytes, where a cytoplasmic reactivity was displayed, whereas in activated, gemistocytic astrocytes it was also concentrated close to the plasma membrane. Although the exact roles of annexin-1 in this setting are still to be determined, a possible contribution to anti-inflammatory processes might be suggested.
Collapse
Affiliation(s)
- S Probst-Cousin
- Department of Neurology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany.
| | | | | | | | | | | |
Collapse
|
635
|
Lyons SA, O'Neal J, Sontheimer H. Chlorotoxin, a scorpion-derived peptide, specifically binds to gliomas and tumors of neuroectodermal origin. Glia 2002; 39:162-73. [PMID: 12112367 DOI: 10.1002/glia.10083] [Citation(s) in RCA: 199] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Highly migratory neuroectodermal cells share a common embryonic origin with cells of the central nervous system (CNS). They include enteric, parasympathetic, sympathoadrenal, and sensory neurons of the peripheral nervous system, Schwann cells, melanocytes, endocrine cells, and cells forming connective tissue of the face and neck. Because of their common embryologic origin, these cells and the tumors that derive from them can share genetic and antigenic phenotypes with gliomas, tumors derived from CNS glia. We recently discovered that chlorotoxin (ClTx), a 4-kD peptide purified from Leiurus quinquestriatus scorpion, is a highly specific marker for glioma cells in biopsy tissues (Soroceanu et al. Cancer Res 58:4871-4879, 1998) that can target tumors in animal models. We report on the specificity of ClTx as a marker for tumors of neuroectodermal origin that include peripheral neuroectodermal tumors (PNET) and gliomas. Specifically, we histochemically stained frozen and paraffin tissue sections of human biopsy tissues from 262 patients with a synthetically manufactured and biologically active ClTx bearing an N-terminal biotin. The vast majority (74 of 79) of primary human brain tumors investigated showed abundant binding of ClTx with greater than 90% ClTx-positive cells in each section. By comparison, 32 biopsies of uninvolved brain used for comparison were largely ClTx-negative, with only a few isolated reactive astrocytes showing some ClTx binding. However, as with gliomas, the vast majority of PNETs examined showed specific ClTx binding (31 of 34). These include medulloblastomas (4 of 4), neuroblastomas (6 of 7), ganglioneuromas (4 of 4), melanomas (7 of 7), adrenal pheochromocytomas (5 of 6), primitive PNET (1), small cell lung carcinoma (2 of 3), and Ewing's sarcoma (2 of 2). Under identical staining conditions, normal tissues from brain, skin, kidney, and lung were consistently negative for ClTx. These results suggest that chlorotoxin is a reliable and specific histopathological marker for tumors of neuroectodermal origin and that chlorotoxin derivatives with cytolytic activity may have therapeutic potential for these cancers.
Collapse
Affiliation(s)
- Susan A Lyons
- TransMolecular, Inc., Birmingham, Alabama 35294, USA
| | | | | |
Collapse
|
636
|
Hoyaux D, Boom A, Van den Bosch L, Belot N, Martin JJ, Heizmann CW, Kiss R, Pochet R. S100A6 overexpression within astrocytes associated with impaired axons from both ALS mouse model and human patients. J Neuropathol Exp Neurol 2002; 61:736-44. [PMID: 12152788 DOI: 10.1093/jnen/61.8.736] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Astrogliosis is one of the earliest pathological changes observed in neurodegenerative diseases in general and in amyotrophic lateral sclerosis (ALS) in particular. ALS is characterized by selective degeneration of motoneurons. There are 2 forms of the disease: sporadic ALS (SALS), comprising 90%-95% of cases, and familial ALS (FALS), comprising 5%-10% of cases. FALS is an age-dependent autosomal dominant disorder in which mutations in the homodimeric enzyme Cu/ Zn superoxide dismutase 1 (SOD1) is linked to the disease. The animal model for this disease is a transgenic mouse expressing the mutated human SOD1(G93A) gene. Here we show by immunohistochemistry and double immunofluorescence that astrocytes located near impaired axons of motoneurons that were selectively programmed to die overexpressed S100A6, a Ca2+/Zn2+ binding protein able to translocate into the nucleus. Transgenic mice overexpressing the mutated human SOD1 gene and patients suffering from SALS showed this selective astrocytic S100A6 expression. For instance, the pyramidal tract could be macroscopically detected on S100A6-labeled spinal cord and brainstem sections from SALS patients. Transgenic mice overexpressing the non-mutated SOD1 gene did not overexpress S100A6, although glial fibrillary associated protein astrogliosis was seen. Although these results do not give any clue about the beneficial or detrimental role played by S100A6, its induction may be assumed to appropriately serve some function(s).
Collapse
Affiliation(s)
- Daphné Hoyaux
- Laboratory of Histopathology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
637
|
Abstract
Inflammation is a defense reaction against diverse insults, designed to remove noxious agents and to inhibit their detrimental effects. It consists of a dazzling array of molecular and cellular mechanisms and an intricate network of controls to keep them in check. In neurodegenerative diseases, inflammation may be triggered by the accumulation of proteins with abnormal conformations or by signals emanating from injured neurons. Given the multiple functions of many inflammatory factors, it has been difficult to pinpoint their roles in specific (patho)physiological situations. Studies of genetically modified mice and of molecular pathways in activated glia are beginning to shed light on this issue. Altered expression of different inflammatory factors can either promote or counteract neurodegenerative processes. Since many inflammatory responses are beneficial, directing and instructing the inflammatory machinery may be a better therapeutic objective than suppressing it.
Collapse
Affiliation(s)
- Tony Wyss-Coray
- Gladstone Institute of Neurological Disease and Department of Neurology, University of California-San Francisco, San Francisco, CA 94141, USA.
| | | |
Collapse
|
638
|
Zou L, Yotnda P, Zhao T, Yuan X, Long Y, Zhou H, Yang K. Reduced inflammatory reactions to the inoculation of helper-dependent adenoviral vectors in traumatically injured rat brain. J Cereb Blood Flow Metab 2002; 22:959-70. [PMID: 12172381 DOI: 10.1097/00004647-200208000-00007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Traumatic brain injury (TBI) causes delayed neuronal deficits that in principle could be prevented by timely intervention with therapeutic genes. However, appropriate vectors for gene transfer to the brain with TBI remain to be developed. First-generation adenoviruses (fgAd) are usually associated with inflammatory and toxic effects when inoculated into brains, despite their high efficiency of gene transfer to these tissues. In this study the authors attempted to determine whether a less immunogenic gene-transfer protocol can be established in the traumatically injured rat brain using helper-dependent adenoviruses (hdAd), a novel adenoviral construct with full deletion of viral coding sequences. Their results show that transgene expression from intrahippocampally inoculated hdAd is maintained for at least 2 months after TBI, in contrast to the much shorter duration of fgAd-mediated gene expression. There was only minimal secretion of proinflammatory IL-1beta and TNF-alpha after inoculation of hdAd. Furthermore, the hdAd-mediated gene expression was associated with less microglial proliferation, astrocytic activation, and macrophage infiltration than observed in fgAd-inoculated brains. There was no additional tissue loss after hdAd inoculation compared with PBS injection. Although both anti-adenoviral and neutralizing antibodies were found in serum after brain inoculation of hdAd, they did not appear to affect transgene expression. The results suggest that hdAd are less immunogenic vectors than conventional adenoviral vectors, and offer improved vehicles for long-term therapeutic transgene transfer to traumatically injured brains.
Collapse
Affiliation(s)
- Linglong Zou
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
639
|
Ahlemeyer B, Kölker S, Zhu Y, Hoffmann GF, Krieglstein J. Increase in glutamate-induced neurotoxicity by activated astrocytes involves stimulation of protein kinase C. J Neurochem 2002; 82:504-15. [PMID: 12153475 DOI: 10.1046/j.1471-4159.2002.00994.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Activation of astrocytes is a common feature of neurological disorders, but the importance of this phenomenon for neuronal outcome is not fully understood. Treatment of mixed hippocampal cultures of neurones and astrocytes from day 2-4 in vitro (DIV 2-4) with 1 micro m cytosine arabinofuranoside (AraC) caused an activation of astrocytes as detected by a stellate morphology and a 10-fold increase in glial fibrillary acidic protein (GFAP) level compared with vehicle-treated cultures. After DIV 12, we determined 43% and 97% damaged neurones 18 h after the exposure to glutamate (1 mm, 1 h) in cultures treated with vehicle and AraC, respectively. Dose-response curves were different with a higher sensitivity to glutamate in cultures treated with AraC (EC50 = 0.01 mm) than with vehicle (EC50 = 0.12 mm). The susceptibility of neurones to 1 mm glutamate did not correlate with the percentage of astrocytes and was insensitive to an inhibition of glutamate uptake. In cultures treated with vehicle and AraC, glutamate-induced neurotoxicity was mediated through stimulation of the NR1-NR2B subtype of NMDA receptors, because it was blocked by the NMDA receptor antagonist MK-801 and the NR1-NR2B selective receptor antagonist ifenprodil. Protein levels of the NR2A and NR2B subunits of NMDA receptor were similar in cultures treated with vehicle or AraC. AraC-induced changes in glutamate-induced neurotoxicity were mimicked by activation of protein kinase C (PKC), whereas neuronal susceptibility to glutamate was reduced in cultures depleted of PKC and treated with AraC suggesting that the increase in glutamate toxicity by activated astrocytes involves activation of PKC.
Collapse
Affiliation(s)
- Barbara Ahlemeyer
- Institut für Pharmakologie und Toxikologie, Fachbereich Pharmazie der Philipps-Universität Marburg, Germany.
| | | | | | | | | |
Collapse
|
640
|
Teunissen CE, de Vente J, Steinbusch HWM, De Bruijn C. Biochemical markers related to Alzheimer's dementia in serum and cerebrospinal fluid. Neurobiol Aging 2002; 23:485-508. [PMID: 12009495 DOI: 10.1016/s0197-4580(01)00328-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The diagnosis of Alzheimer's disease (AD) is currently based on clinical and neuropsychological examination. To date there is no blood test available that can discriminate dementia patients from healthy individuals. In the present paper, an overview of the current state of knowledge on biologic markers in serum (plasma) and CSF is presented. The combination of characteristic plaque markers tau and amyloid bèta may constitute a specific and sensitive CSF marker for AD. Glial fibrillary acidic protein and antibodies in CSF may be a marker for severe neurodegeneration. CSF concentrations of the oxidative stress markers 3-nitrotyrosine, 8-hydroxy-2'-deoxyguanosine and isoprostanes are increased in AD patients. Serum 24S-OH-cholesterol may be an early whereas glial fibrillary acidic protein autoantibody level may be a late marker for neurodegeneration. To date, serum alpha(1)-Antichymotripsin concentration is the most convincing marker for CNS inflammation. Increased serum homocysteine concentrations have also been consistently reported in AD. In summary, a large overlap in mean concentrations has been observed in studies comparing AD patients with healthy controls for single markers. These studies together support the theory of testing several serum markers in combination for the diagnosis of AD.
Collapse
Affiliation(s)
- C E Teunissen
- European Graduate School of Neuroscience (Euron), Universiteit Maastricht, Department of Psychiatry and Neuropsychology, P.O. Box 616, 6200 MD, The Netherlands.
| | | | | | | |
Collapse
|
641
|
Baltrons MA, Pedraza CE, Heneka MT, García A. Beta-amyloid peptides decrease soluble guanylyl cyclase expression in astroglial cells. Neurobiol Dis 2002; 10:139-49. [PMID: 12127152 DOI: 10.1006/nbdi.2002.0492] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In astroglial cells beta-amyloid peptides (betaA) induce a reactive phenotype and increase expression of NO synthase. Here we show that treatment of rat brain astrocytes with betaA decreases their capacity to accumulate cyclic GMP (cGMP) in response to NO as a result of a decreased expression of soluble guanylyl cyclase (sGC) at the protein and mRNA levels. Potentiation of betaA-induced NO formation by interferon-gamma did not result in a larger decrease in cGMP formation and inhibition of NO synthase failed to reverse down-regulation of sGC, indicating that NO is not involved. The betaA effect was prevented by the protein synthesis inhibitor cycloheximide. Intracerebral betaA injection also decreased sGC beta1 subunit mRNA levels in adult rat hippocampus and cerebellum. A loss of sGC in reactive astrocytes surrounding beta-amyloid plaques could be a mechanism to prevent excess signalling via cGMP at sites of high NO production.
Collapse
Affiliation(s)
- María Antonia Baltrons
- Instituto de Biotecnología y Biomedicina V. Villar Palasi, Departamento de Bioquímica Biología Molecular, Universidad Autónoma de Barcelona, 08193, Bellaterra, Spain
| | | | | | | |
Collapse
|
642
|
Uhlmann EJ, Apicelli AJ, Baldwin RL, Burke SP, Bajenaru ML, Onda H, Kwiatkowski D, Gutmann DH. Heterozygosity for the tuberous sclerosis complex (TSC) gene products results in increased astrocyte numbers and decreased p27-Kip1 expression in TSC2+/- cells. Oncogene 2002; 21:4050-9. [PMID: 12037687 DOI: 10.1038/sj.onc.1205435] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2001] [Revised: 11/15/2001] [Accepted: 11/26/2001] [Indexed: 11/09/2022]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant tumor predisposition syndrome characterized by benign proliferations (hamartomas). In the brain, individuals with TSC develop autism, mental retardation and seizures associated with focal cortical dysplasias, subependymal nodules, and subependymal giant cell astrocytomas (SEGAs). We hypothesize that dysregulated astrocyte function due to mutations in the tumor suppressor genes, TSC1 and TSC2, may contribute to the pathogenesis of these brain abnormalities. In this report, we demonstrate that mice heterozygous for a targeted defect in either the Tsc1 or Tsc2 genes(Tsc1+/- and Tsc2+/- mice) exhibit a 1.5-fold increase in the number of astrocytes in vivo. Whereas increased astrocyte numbers in vivo were suggestive of a proliferative advantage, Tsc2+/- primary astrocyte cultures did not show a cell-autonomous growth advantage, anchorage-independent growth, increased saturation density, or increased fluid-phase endocytosis compared to wild type astrocytes. Tsc2 null mouse embryonic fibroblasts (MEFs) however, did exhibit increased saturation density compared to Tsc2 wild type controls. In both Tsc2+/- astrocytes and Tsc2 null mouse embryonic fibroblasts, p27-Kip1 expression was decreased compared to wild type cells, and was reversed by tuberin re-expression in Tsc2-/- MEFs. In contrast, no change in endocytosis was observed upon tuberin re-expression in Tsc2-/- MEFs. Collectively, these results suggest Tsc heterozygosity may provide a non-cell-autonomous growth advantage for astrocytes that may involve p27-Kip1 expression.
Collapse
Affiliation(s)
- Erik J Uhlmann
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
643
|
Tateishi N, Mori T, Kagamiishi Y, Satoh S, Katsube N, Morikawa E, Morimoto T, Matsui T, Asano T. Astrocytic activation and delayed infarct expansion after permanent focal ischemia in rats. Part II: suppression of astrocytic activation by a novel agent (R)-(-)-2-propyloctanoic acid (ONO-2506) leads to mitigation of delayed infarct expansion and early improvement of neurologic deficits. J Cereb Blood Flow Metab 2002; 22:723-34. [PMID: 12045671 DOI: 10.1097/00004647-200206000-00011] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A novel agent, (R)-(-)-2-propyloctanoic acid (ONO-2506), has a unique property in that it modulates functions of activated cultured astrocytes, including pronounced inhibition of S-100beta synthesis. The present study examined whether administration of this agent would mitigate the delayed expansion of infarct volume and the neurologic deficits after permanent middle cerebral artery occlusion (pMCAO) in rats. Daily intravenous administration of ONO-2506 (10 mg/kg) abolished the delayed infarct expansion between 24 and 168 hours after pMCAO, whereas the acute infarct expansion until 24 hours was unaffected. The agent significantly reduced the expression of S-100beta and glial fibrillary acidic protein in the activated astrocytes and the number of terminal deoxynucleotidyl transferase-mediated 2;-deoxyuridine 5;-triphosphate-biotin nick end labeling-positive cells in the periinfarct area. The neurologic deficits were significantly improved, compared with the vehicle-treated groups, as early as 24 hours after the initial administration of ONO-2506. The agent had a wide therapeutic time window of 0 to 48 hours after pMCAO. These results indicate that because of the pharmacologic modulation of astrocytic activation induced by ONO-2506, symptoms can regress whereas delayed expansion of the lesion is arrested. Pharmacologic modulation of astrocytic activation may confer a novel therapeutic strategy against stroke.
Collapse
Affiliation(s)
- Narito Tateishi
- Minase Research Institute, Ono Pharmaceutical Co. Ltd., Shimamoto, Mishima, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
644
|
|
645
|
Matsui T, Mori T, Tateishi N, Kagamiishi Y, Satoh S, Katsube N, Morikawa E, Morimoto T, Ikuta F, Asano T. Astrocytic activation and delayed infarct expansion after permanent focal ischemia in rats. Part I: enhanced astrocytic synthesis of s-100beta in the periinfarct area precedes delayed infarct expansion. J Cereb Blood Flow Metab 2002; 22:711-22. [PMID: 12045670 DOI: 10.1097/00004647-200206000-00010] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
An astrocytic protein S-100beta enhances the expression of inducible nitric oxide synthase in cultured astrocytes at micromolar concentrations, leading to nitric oxide-mediated death of cocultured neurons. The present study examined whether S-100beta production by reactive astrocytes accumulating within the periinfarct area was related to delayed expansion of infarct volume after permanent middle cerebral artery occlusion in the rat. After rapid increases during the initial 24 hours, the increase of infarct volume then decelerated while maintaining the increasing tendency until 168 hours in this model, attaining a significant difference compared with that at 24 hours. In the periinfarct area, the number of reactive astrocytes expressing both S-100 and glial fibrillary acidic protein, the tissue level of S-100beta as measured by the sandwich enzyme-linked immunosolvent assay method using anti-S-100beta monoclonal antibody, and the number of terminal deoxynucleotidyl transferase-mediated 2;-deoxyuridine 5;-triphosphate-biotin nick end labeling-positive cells were significantly increased preceding the delayed expansion of infarct volume. The CSF concentration of S-100beta showed a biphasic increase, presumably reflecting the immediate release from astrocytes within the ischemic core and the subsequent production in reactive astrocytes within the periinfarct area. These results show for the first time that the enhanced synthesis of S-100beta by reactive astrocytes participates in the inflammatory responses within the periinfarct area, which may be related to the occurrence of delayed infarct expansion as a major component of the cytokine network.
Collapse
Affiliation(s)
- Toru Matsui
- Department of Neurosurgery and Institute of Laboratory Animal Science, Saitama Medical Center/School, Kawagoe, Saitama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
646
|
Girvin AM, Gordon KB, Welsh CJ, Clipstone NA, Miller SD. Differential abilities of central nervous system resident endothelial cells and astrocytes to serve as inducible antigen-presenting cells. Blood 2002; 99:3692-701. [PMID: 11986225 DOI: 10.1182/blood-2001-12-0229] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Microglial cells and astrocytes are capable of processing and presenting antigens for efficient activation of T cells. However, the antigen-presenting function and role of cerebrovascular endothelial cells (CVEs) in central nervous system inflammatory responses remain controversial. We compared the expression of necessary accessory molecules and the functional antigen-presenting capacity of cloned SJL/J CVEs and primary astrocytes in response to the pro-inflammatory cytokines interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). Astrocytes and CVEs up-regulated major histocompatibility complex (MHC) class II, and primarily B7-1 as opposed to B7-2, in response to IFN-gamma. TNF-alpha inhibited the IFN-gamma-induced up-regulation of MHC class II on CVEs correlating to a decrease in the mRNA for the class II transactivator (CIITA), whereas CIITA expression in astrocytes was unaffected. Unlike astrocytes, CVEs did not elicit significant MHC class II-restricted T-cell responses. Furthermore, we have found that CVE monolayers are altered following T-cell contact, implicating CVE/T-cell contact in the breakdown of the blood-brain barrier during neuro-inflammatory responses.
Collapse
Affiliation(s)
- Ann M Girvin
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
647
|
Omori Y, Imai JI, Suzuki Y, Watanabe S, Tanigami A, Sugano S. OASIS is a transcriptional activator of CREB/ATF family with a transmembrane domain. Biochem Biophys Res Commun 2002; 293:470-7. [PMID: 12054625 DOI: 10.1016/s0006-291x(02)00253-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Murine OASIS is a putative CREB/ATF family transcription factor that is induced in gliosis, but its molecular role has not been determined. We have isolated the human OASIS gene and investigated the potential of OASIS protein as a transcriptional activator. We found that OASIS can activate transcription through box-B elements but not through the somatostatin CRE. OASIS contains a putative C-terminal hydrophobic transmembrane domain, a typical structural feature for the transcription factors activated by regulated intramembrane proteolysis. Truncation of the OASIS transmembrane domain resulted in a significant increase in transcriptional activity and altered its subcellular localization from the endoplasmic reticulum to the nucleus. Western blot analysis of transfected cells identified OASIS polypeptides of 82 and 66 kDa. These results suggest that the transmembrane domain plays an important role in the regulation of transcriptional activation by OASIS.
Collapse
Affiliation(s)
- Yoshihiro Omori
- Laboratory of Genome Structure Analysis Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokane-dai, Minato-ku, Tokyo 108-8639, Japan.
| | | | | | | | | | | |
Collapse
|
648
|
The critical role of basement membrane-independent laminin gamma 1 chain during axon regeneration in the CNS. J Neurosci 2002. [PMID: 11943817 DOI: 10.1523/jneurosci.22-08-03144.2002] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have addressed the question of whether a family of axon growth-promoting molecules known as the laminins may play a role during axon regeneration in the CNS. A narrow sickle-shaped region containing a basal lamina-independent form of laminin exists in and around the cell bodies and proximal portion of the apical dendrites of CA3 pyramidal neurons of the postnatal hippocampus. To understand the possible function of laminin in axon regeneration within this pathway, we have manipulated laminin synthesis at the mRNA level in a slice culture model of the lesioned mossy system. In this model early postnatal mossy fibers severed near the hilus can regenerate across the lesion and elongate rapidly within strata lucidum and pyramidale. In slice cultures of the postnatal day 4 hippocampus, 2 d before lesion and then continuing for 1-5 d after lesion, translation of the gamma1 chain product of laminin was reduced by using antisense oligodeoxyribonucleotides and DNA enzymes. In the setting of the lesioned organotypic hippocampal slice, astroglial repair of the lesion and overall glial patterning were unperturbed by the antisense or DNA enzyme treatments. However, unlike controls, in the treated, lesioned slices the vast majority of regenerating mossy fibers could not cross the lesion site; those that did were very much shorter than usual, and they took a meandering course. In a recovery experiment in which the DNA enzyme or antisense oligos were washed away, laminin immunoreactivity returned and mossy fiber regeneration resumed. These results demonstrate the critical role of laminin(s) in an axon regeneration model of the CNS.
Collapse
|
649
|
McCarthy JB, Barker-Gibb AL, Alves SE, Milner TA. TrkA immunoreactive astrocytes in dendritic fields of the hippocampal formation across estrous. Glia 2002; 38:36-44. [PMID: 11921202 DOI: 10.1002/glia.10060] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neurotrophins are important modulators of structural synaptic plasticity. (Through trophic action (Jordan. J Neurobiol 40:434-445, 1999), astrocytes serve as permissive substrates to support axonal regrowth (Ridet et al. Trends Neurosci 20:570-571, 1997), and are involved in estrogen-induced synaptic structural plasticity (Garcia-Segura et al. Cell Mol Neurobiol 16:225-237, 1996). Previously, we reported that tyrosine kinase A receptor (TrkA) immunoreactivity was present both in presynaptic neuronal processes (axons and terminals) and in select astrocytes of the male rat hippocampal formation (Barker-Gibb et al. J Comp Neurol 430:182-199, 2001). We show that the number of TrkA-immunoreactive astrocytes in female rats fluctuates 16-fold across the estrous cycle in dendritic fields of the hippocampal formation, with the greatest number at estrus after the peak plasma estradiol concentration of proestrus. Few TrkA-labeled astrocytes were found in ovariectomized animals; after estrogen replacement, this number increased by 12-fold in the hippocampal formation, indicating estrogen-mediated induction. Dual-labeling studies showed that TrkA-labeled astrocytes were also immunoreactive for vimentin, a protein expressed by reactive astrocytes. Ultrastructural analysis of the dentate gyrus molecular layer demonstrated that TrkA immunoreactive astrocytes are positioned primarily next to dendrites and unmyelinated axons. Because nerve growth factor (NGF) has been reported to stimulate astrocytes to function as substrates for axon growth (Kawaja and Gage. Neuron 7:1019-1030, 1991), these findings are consistent with the theory that TrkA immunoreactive astrocytes serve a role in structural plasticity, axon guidance, and synaptic regeneration across the estrous cycle in the hippocampal formation.
Collapse
Affiliation(s)
- J B McCarthy
- Division of Neurobiology, Weill Medical College of Cornell University, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
650
|
Roch C, Leroy C, Nehlig A, Namer IJ. Magnetic resonance imaging in the study of the lithium-pilocarpine model of temporal lobe epilepsy in adult rats. Epilepsia 2002; 43:325-35. [PMID: 11952761 DOI: 10.1046/j.1528-1157.2002.11301.x] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE In temporal lobe epilepsy, it remains to be clarified whether hippocampal sclerosis is the cause or the consequence of epilepsy. We studied the temporal evolution of the lesions in the lithium-pilocarpine model of epilepsy in the rat with magnetic resonance imaging (MRI) to determine the progressive morphologic changes occurring before the appearance of chronic epilepsy. METHODS MRI was performed on an MR scanner operating at 4.7 T. We followed the evolution of lesions using T(2)- and T(1)-weighted sequences before and after the injection of gadolinium from 2 h to 9 weeks. RESULTS At 2 h after status epilepticus (SE), a blood-brain barrier breakdown could be observed only in the thalamus; it had disappeared by 6 h. At 24 h after SE, edema was present in the amygdala and the piriform and entorhinal cortices together with extensive neuronal loss; it disappeared progressively over a 5-day period. During the chronic phase, a cortical signal reappeared in all animals; this signal corresponded to gliosis, which appeared on glial fibrillary acidic protein (GFAP) immunohistochemically stained sections as hypertrophic astrocytes with thickened processes. In the hippocampus, the correlation between histopathology and T(2)-weighted signal underscored the progressive constitution of atrophy and sclerosis, starting 2 days after SE. CONCLUSIONS These data show the reactivity of the cortex that characterizes the initial step leading to the development of epilepsy and the late gliosis that could result from the spontaneous seizures. Moreover, it appears that hippocampal sclerosis progressively worsened and could be both the cause and the consequence of epileptic activity.
Collapse
Affiliation(s)
- Catherine Roch
- Institut de Physique Biologique (UMR 7004 ULP/CNRS/IFR37), Faculté de Médecine, 4 rue Kirschleger, 67085 Strasbourg Cedex, France.
| | | | | | | |
Collapse
|