501
|
Karten B, Hayashi H, Francis G, Campenot R, Vance D, Vance J. Generation and function of astroglial lipoproteins from Niemann-Pick type C1-deficient mice. Biochem J 2005; 387:779-88. [PMID: 15544574 PMCID: PMC1135009 DOI: 10.1042/bj20041694] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Revised: 11/01/2004] [Accepted: 11/15/2004] [Indexed: 11/17/2022]
Abstract
NPC (Niemann-Pick type C) disease is a progressive neurological disorder characterized by defects in intracellular cholesterol trafficking, accumulation of cholesterol in the endosomal system and impaired cholesterol homoeostasis. Although these alterations appear to occur in all NPC1-deficient cell types, the consequences are most profound in the nervous system. Since glial cells are important mediators of brain cholesterol homoeostasis, we proposed that defective generation and/or function of lipoproteins released by glia might contribute to the neurological abnormalities associated with NPC disease. We found that, as in other cell types, Npc1-/- glia accumulate cholesterol intracellularly. We hypothesized that this sequestration of cholesterol in glia might restrict the availability of cholesterol for lipoprotein production. Cerebellar astroglia were cultured from a murine model of NPC disease to compare the lipoproteins generated by these cells and wild-type glia. The experiments demonstrate that the amount of cholesterol in glia-conditioned medium is not reduced by NPC1 deficiency. Similarly, cholesterol efflux to apo (apolipoprotein) A1 or glial expression of the transporter ATP-binding-cassette transporter A1 was not decreased by NPC1 deficiency. In addition, the ratio of apo E:cholesterol and the density distribution of lipoproteins in Npc1-/- and Npc1+/+ glia-conditioned medium are indistinguishable. Importantly, in a functional assay, apo E-containing lipoproteins generated by Npc1-/- and Npc1+/+ glia each stimulate axonal elongation of neurons by approx. 35%. On the basis of these observations, we speculate that the neuropathology characteristic of NPC disease can quite probably be ascribed to impaired processes within neurons in the brain rather than defective lipoprotein production by astroglia.
Collapse
Key Words
- apo a1
- apo e
- astroglia
- cholesterol secretion
- glial lipoprotein
- niemann–pick type c disease
- abc transporter, atp-binding-cassette transporter
- apo, apolipoprotein
- cns, central nervous system
- csf, cerebrospinal fluid
- dmem, dulbecco's modified eagle's medium
- fbs, fetal bovine serum
- gfap, glial fibrillary acidic protein
- hdl, high-density lipoproteins
- ldl, low-density lipoprotein
- vldl, very low density lipoprotein
- npc, niemann–pick type c
- rgc, retinal ganglion cells
Collapse
Affiliation(s)
- Barbara Karten
- *Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
- †Department of Medicine, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Hideki Hayashi
- *Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
- †Department of Medicine, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Gordon A. Francis
- *Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
- †Department of Medicine, University of Alberta, Edmonton, AB, Canada T6G 2S2
- ‡Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
| | - Robert B. Campenot
- §Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
| | - Dennis E. Vance
- *Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
- ‡Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
| | - Jean E. Vance
- *Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
- †Department of Medicine, University of Alberta, Edmonton, AB, Canada T6G 2S2
| |
Collapse
|
502
|
Shimamura M, Garcia JM, Prough DS, Dewitt DS, Uchida T, Shah SA, Avila MAA, Hellmich HL. Analysis of long-term gene expression in neurons of the hippocampal subfields following traumatic brain injury in rats. Neuroscience 2005; 131:87-97. [PMID: 15680694 DOI: 10.1016/j.neuroscience.2004.10.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2004] [Indexed: 01/19/2023]
Abstract
After experimental traumatic brain injury (TBI), widespread neuronal loss is progressive and continues in selectively vulnerable brain regions, such as the hippocampus, for months to years after the initial insult. To clarify the molecular mechanisms underlying secondary or delayed cell death in hippocampal neurons after TBI, we compared long-term changes in gene expression in the CA1, CA3 and dentate gyrus (DG) subfields of the rat hippocampus at 24 h and 3, 6, and 12 months after TBI with changes in gene expression in sham-operated rats. We used laser capture microdissection to collect several hundred hippocampal neurons from the CA1, CA3, and DG subfields and linearly amplified the nanogram samples of neuronal RNA with T7 RNA polymerase. Subsequent quantitative analysis of gene expression using ribonuclease protection assay revealed that mRNA expression of the anti-apoptotic gene, Bcl-2, and the chaperone heat shock protein 70 was significantly downregulated at 3, 6 (Bcl-2 only), and 12 months after TBI. Interestingly, the expression of the pro-apoptotic genes caspase-3 and caspase-9 was also significantly decreased at 3, 6 (caspase-9 only), and 12 months after TBI, suggesting that long-term neuronal loss after TBI is not mediated by increased expression of pro-apoptotic genes. The expression of two aging-related genes, p21 and integrin beta3 (ITbeta3), transiently increased 24 h after TBI, returned to baseline levels at 3 months and significantly decreased below sham levels at 12 months (ITbeta3 only). Expression of the gene for the antioxidant glutathione peroxidase-1 also significantly increased 6 months after TBI. These results suggest that decreased levels of neuroprotective genes may contribute to long-term neurodegeneration in animals and human patients after TBI. Conversely, long-term increases in antioxidant gene expression after TBI may be an endogenous neuroprotective response that compensates for the decrease in expression of other neuroprotective genes.
Collapse
Affiliation(s)
- M Shimamura
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0830, USA
| | | | | | | | | | | | | | | |
Collapse
|
503
|
Ghorpade A, Bruch L, Persidsky Y, Chin B, Brown WHC, Borgmann K, Persidsky R, Wu L, Holter S, Cotter R, Faraci J, Heilman D, Meyer V, Potter JF, Swindells S, Gendelman HE. Development of a rapid autopsy program for studies of brain immunity. J Neuroimmunol 2005; 163:135-44. [PMID: 15885316 DOI: 10.1016/j.jneuroim.2005.01.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2004] [Revised: 01/12/2005] [Accepted: 01/25/2005] [Indexed: 11/19/2022]
Abstract
Human glia are essential cellular models used for studies of neurodegenerative diseases. Fetal neuroglia are commonly used, as they can be recovered in large quantities and sustained for long periods in culture. However, fetal neuroglia may have limitations in reflecting adult diseases and additionally can pose ethical issues in translating products of abortion for research use. To address these concerns, we developed a rapid autopsy program to procure age- and disease-specific neuroglia from adult brain tissues within hours of death. The challenges in developing this initiative, reflecting experiences from 69 autopsies over 4 years, are presented.
Collapse
Affiliation(s)
- Anuja Ghorpade
- Laboratory of Cellular Neuroimmunology, 985215 Nebraska Medical Center, Omaha, NE, 68198-5215, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
504
|
Foote AK, Blakemore WF. Repopulation of oligodendrocyte progenitor cell depleted tissue in a model of chronic demyelination. Neuropathol Appl Neurobiol 2005; 31:105-14. [PMID: 15771704 DOI: 10.1111/j.1365-2990.2004.00634.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Some, but not all chronically demyelinated MS lesions are depleted of oligodendrocyte progenitor cells (OPCs) suggesting that OPCs are destroyed during the process of demyelination and some factor impedes OPC repopulation of the depleted tissue. The chronically demyelinated axons in MS lie in an astrocytic environment and it has been proposed that this might impede entry of OPCs into such regions. By depleting a short length of spinal cord of its OPCs using 40 Gy of X-irradiation in both normal rats and rats with progressive myelin loss accompanied by an astrocytosis (taiep rats), we investigated whether such changes affect the ability of OPCs to repopulate OPC-depleted tissue. In both taiep and normal rats, the rate of repopulation decreases with age, but no difference was detected in the rate at which OPCs repopulated normally myelinated and chronically demyelinated and astrocytosed tissue. This indicates that, if the astrocytic environment of the taiep CNS is comparable to that found in MS lesions, then the presence of chronically demyelinated axons and astrocytosis in chronic MS lesions does not represent a barrier to repopulation of the tissue by OPCs. However, similar to the situation in the normal adult rodent CNS, the rate of repopulation by endogenous OPCs in aged taiep rats is very slow, approximately 0.2 mm per week.
Collapse
Affiliation(s)
- A K Foote
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
505
|
|
506
|
Bolin LM, Zhaung A, Strychkarska-Orczyk I, Nelson E, Huang I, Malit M, Nguyen Q. Differential inflammatory activation of IL-6 (−/−) astrocytes. Cytokine 2005; 30:47-55. [PMID: 15804595 DOI: 10.1016/j.cyto.2004.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Revised: 11/22/2004] [Accepted: 11/29/2004] [Indexed: 10/25/2022]
Abstract
IL-6 is a major immunomodulatory cytokine with neuroprotective activity. The absence of interleukin-6 (IL-6) results in increased vulnerability of dopaminergic neurons to the neurotoxicant, MPTP, and a compromised reactive microgliosis. To determine how astrogliosis may contribute to nigrostriatal degeneration in IL-6 (-/-) mice, the inflammatory profiles of astrocytes of IL-6 genotype were compared. Fourteen cytokines and four chemokines were simultaneously assayed in the supernatants of LPS-stimulated primary astrocyte cultures. In a time course of 6, 18 and 48 h and LPS stimulations of 0, 0.1, 1, 10 and 100 ng/ml, IL-6 (-/-) astrocytes secreted significantly greater amounts of the pro-inflammatory cytokines IL-1alpha, IL-1beta and TNFalpha than did IL-6 (+/+) cells. Elevated levels of IL-10 and IL-12p40 were only detected at 48 h post-stimulation with greater IL-10 in IL-6 (-/-) supernatants and greater IL-12p40 in IL-6 (+/+) supernatants. IL-6 (+/+) astrocytes produced more G-CSF and GM-CSF when compared with IL-6 (-/-) astrocytes. Chemokine levels were greater in supernatants of IL-6 (+/+) astrocytes than IL-6 (-/-) cells prior to 48 h post-stimulation. At that time, higher levels of MIP-1alpha were maintained in IL-6 (+/+) supernatant, while similar levels of MCP-1 in supernatants of both IL-6 (+/+) and IL-6 (-/-) cells were measured. Additionally, LPS (100 ng/ml) resulted in greater levels of KC and Rantes in IL-6 (-/-) astrocyte supernatants compared with IL-6 (+/+) supernatants at that time. These results suggest that the autocrine modulatory activities of IL-6 affect multiple cytokine secretory pathways, which could participate in neurodegenerative processes.
Collapse
Affiliation(s)
- L M Bolin
- The Parkinson's Institute, 1170 Morse Avenue, Sunnyvale, CA 94089-1605, USA.
| | | | | | | | | | | | | |
Collapse
|
507
|
Lee HK, Choi SS, Han EJ, Lee JY, Kwon MS, Shim EJ, Seo YJ, Suh HW. Role of nicotinic acetylcholine receptors in the regulation of kainic acid-induced hippocampal cell death in mice. Brain Res Bull 2005; 64:309-17. [PMID: 15561465 DOI: 10.1016/j.brainresbull.2004.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Revised: 07/05/2004] [Accepted: 08/06/2004] [Indexed: 10/26/2022]
Abstract
Kainic acid (KA) is a well-known excitatory, neurotoxic substance. In mice, morphological damage of hippocampus induced by KA administered intracerebroventricularly (i.c.v.) was markedly concentrated on the CA3 pyramidal neurons. In the present study, the possible role of nicotinic acetylcholine receptors (nAchRs) in hippocampal cell death induced by KA (0.1 microg) administered i.c.v. was examined. Methyllycaconitine (MC; nAchRs antagonist, 20 microg) attenuated KA-induced CA3 pyramidal cell death. KA increased immunoreactivities (IRs) of phorylated extracellular signal-regulated kinase (p-ERK; at 30 min), p-CaMK II (at 30 min), c-Fos (at 2 h), c-Jun (at 2 h), glial fibrillary acidic protein (GFAP at 1 day), and the complement receptor type 3 (OX-42; at 1 day) in hippocampal area. MC attenuated selectively KA-induced p-CaMK II, GFAP and OX-42 IR in the hippocampal CA3 region. Our results suggest that p-CaMK II may play as an important regulator responsible for the hippocampal cell death induced by KA administered i.c.v. in mice. Reactive astrocytes, which was meant by GFAP IR, and activated microglia, which was meant by OX-42 IR, may be a good indicator for measuring the cell death in hippocampal regions by KA-induced excitotoxicity. Furthermore, it is implicated that niconitic receptors appear to be involved in hippocampal CA3 pyramidal cell death induced by KA administered i.c.v. in mice.
Collapse
Affiliation(s)
- Han-Kyu Lee
- Department of Pharmacology and Institute of Natural Medicine, College of Medicine, Hallym University, 1 Okchun-Dong, Chunchon, Kangwon-Do 200-702, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
508
|
Mares V, Malík R, Lisá V, Sedo A. Up-regulation of gamma-glutamyl transpeptidase (GGT) activity in growth perturbed C6 astrocytes. ACTA ACUST UNITED AC 2005; 136:75-80. [PMID: 15893589 DOI: 10.1016/j.molbrainres.2005.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2004] [Revised: 12/31/2004] [Accepted: 01/08/2005] [Indexed: 11/28/2022]
Abstract
Activity of gamma-glutamyl transpeptidase (GGT) was studied in astrocyte-like C6 glial cells modulated in growth and maturation by different concentration of serum and dibutyryl cyclic AMP (Db-cAMP) supplement in culture medium. After reduction of serum concentration from 10% to 0.1%, the number of GGT positive cells determined histochemically increased 3.1 times and the GGT activity/mg protein in whole cell lysates was 5.1 times higher. In cultures with 0.1% serum + Db-cAMP, the histochemically and biochemically assayed GGT activity exceeded 5.1 and 7.9 times the values measured in control 10% serum cultures, respectively. The up-regulation of GGT was accompanied by an inhibition of proliferation, enhanced differentiation and hypertrophy of cells. In addition, the process of metabolic perturbation and/or cellular stress was revealed in these cultures by the (i) growth-support release followed by shrinkage and death of a small number of cells and (ii) higher oxidation of 2'7'dichlorofluorescein diacetate to its fluorescent form in the adherent/viable cells. The observed up-regulation of GGT is considered to primarily reflect increased metabolism of glutathione and/or the maintenance of the redox potential in cells stressed by sub-optimal concentration of serum and Db-cAMP supplement. The concomitant cellular hypertrophy and differentiation and their relationship to increased activity of GGT await further investigation. The study suggests that up-regulation of GGT can contribute to adaptation of astrocytic cells to metabolic and/or oxidative perturbances occurring under various pathological conditions, including radiation- and drug-induced toxicity.
Collapse
Affiliation(s)
- V Mares
- Joint Laboratory of Cancer Cell Biology of the First Faculty of Medicine, Charles University and Institute of Physiology, Academy of Sciences, Videńská 1083, CZ-14200 Prague, Czech Republic.
| | | | | | | |
Collapse
|
509
|
Kim SY, Li J, Bentsman G, Brooks AI, Volsky DJ. Microarray analysis of changes in cellular gene expression induced by productive infection of primary human astrocytes: implications for HAD. J Neuroimmunol 2005; 157:17-26. [PMID: 15579276 DOI: 10.1016/j.jneuroim.2004.08.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2004] [Indexed: 12/11/2022]
Abstract
The role of astrocytes in HIV-1 associated dementia (HAD) is not well understood. HIV-1 binds efficiently to astrocytes but infects only a small fraction of the cells in vitro and in vivo. To gain insight into the biology of HIV-1-expressing astrocytes, we productively infected human fetal astrocytes with pseudotyped HIV-1 and employed Affymetrix oligonucleotide microarrays to determine global changes in cellular gene expression at the peak of virus production. With a twofold change as a cutoff, HIV-1 increased transcription of 266 genes in astrocytes and suppressed expression of 468. The functions of highly expressed genes included interferon-mediated antiviral responses (OAS1, IFIT1), intercellular contacts (SH3, glia-derived nexin), cell homing/adhesion (matrix metalloproteinases), and cell-cell signaling (neuropilin 1 and 2). Surprisingly, genes involved in innate immune responses of astrocytes were largely unaffected. The single most significant effect of HIV-1, however, was down-modulation of at least 55 genes involved in control of cell cycle, DNA replication, and cell proliferation, which were overrepresented in these categories with probability scores of 10(-10)-10(-26). Our data suggest that HIV-1 expression in astrocytes profoundly alters host cell biology, with potential consequences for the physiological function of astrocytes during HIV-1 infection in the brain.
Collapse
Affiliation(s)
- Seon-Young Kim
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center, 432 West 58th Street, Antenucci Building, Room 709, New York, NY 10019, USA
| | | | | | | | | |
Collapse
|
510
|
Fairless R, Frame MC, Barnett SC. N-cadherin differentially determines Schwann cell and olfactory ensheathing cell adhesion and migration responses upon contact with astrocytes. Mol Cell Neurosci 2005; 28:253-63. [PMID: 15691707 DOI: 10.1016/j.mcn.2004.09.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2004] [Revised: 09/09/2004] [Accepted: 09/16/2004] [Indexed: 10/26/2022] Open
Abstract
Olfactory ensheathing cells (OECs) and Schwann cells provide a cellular environment that promotes axonal outgrowth in several models of CNS injury. However, they exhibit different properties when in contact with astrocytes. Schwann cells, but not OECs, induce characteristics that typify hypertrophy in astrocytes and exhibit a poor capacity to migrate within astrocyte-rich areas, making them less favourable for transplant-mediated repair. N-cadherin has been implicated in the adhesion of Schwann cells to astrocytes. Despite indistinguishable expression of N-cadherin, Schwann cells adhered more strongly to an astrocyte monolayer and migrated more slowly on astrocytes when compared to OECs. We have examined the role of N-cadherin in mediating these cellular interactions using RNA interference and found differing effects. In Schwann cells, suppression of N-cadherin reduced heterotypic and homotypic adhesion and they gained adhesion properties more akin to OECs. In contrast, suppression of N-cadherin in OECs had no effect. These findings imply that N-cadherin is differentially regulated in OECs and Schwann cells.
Collapse
Affiliation(s)
- Richard Fairless
- Division of Clinical Neuroscience, University of Glasgow, Beatson Institute, Glasgow G61 1BD, UK
| | | | | |
Collapse
|
511
|
Colodner KJ, Montana RA, Anthony DC, Folkerth RD, De Girolami U, Feany MB. Proliferative Potential of Human Astrocytes. J Neuropathol Exp Neurol 2005; 64:163-9. [PMID: 15751231 DOI: 10.1093/jnen/64.2.163] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Although a number of studies have demonstrated proliferation of nonneoplastic astrocytes in experimental animal models, the proliferative potential of human astrocytes has not been well defined. Using double-label immunohistochemistry, we identified proliferating cells with the proliferation marker MIB-1 and astrocytes with glial fibrillary acidic protein staining in human biopsy and autopsy tissue. MIB-1 labeling of astrocytes was monitored in a variety of conditions containing significant numbers of reactive astrocytes, including infections, arteriovenous malformations, demyelinating lesions, metastatic tumors, and long-standing gliosis. Twenty-nine of a total of 54 cases showed no evidence of astrocyte-specific MIB-1 labeling despite prominent reactive changes. An average proliferation rate of 0.9% was present in the remaining 25 cases. Labeling indices were highest in infectious conditions and acute demyelinating lesions. We also examined astrocyte proliferation in 5 cases of progressive multifocal leukoencephalopathy. Astrocytic labeling indices were notably elevated in these cases, with an average labeling index of 5.8%. We conclude that low, but appreciable, astrocytic proliferation may occur in nonneoplastic human astrocytes. These findings have implications for astrocyte function in the normal and disease states and for the diagnostic distinction between reactive lesions and low-grade astrocytic neoplasms.
Collapse
Affiliation(s)
- Kenneth J Colodner
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
512
|
Milenkovic I, Nedeljkovic N, Filipovic R, Pekovic S, Culic M, Rakic L, Stojiljkovic M. Pattern of Glial Fibrillary Acidic Protein Expression Following Kainate-Induced Cerebellar Lesion in Rats. Neurochem Res 2005; 30:207-13. [PMID: 15895824 DOI: 10.1007/s11064-004-2443-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In the present study glial fibrillary acidic protein (GFAP) expression was assessed following intravermian injection of kainic acid (KA) or physiological saline to adult rat cerebellum. After 2- to 30-day recovery period, free-floating sections cut with a microtome were obtained and were proccessed for immunocytochemistry against GFAP. Injection of both kainate and physiological saline elicited significant astrogliotic reaction, i.e. in the area around the lesion thick GFAP-positive Bergmann fibers with typical orientation appeared in the molecular and hypertrophied astrocytes abundantly appeared in the granular layer. However, following kainate intoxication lesion was not surrounded by typical demarcation glial scar during 30-day recovery period in contrast to the appearance of usual glial scar in the group injected with physiological saline, as early as 7-day postlesion. Preserved spatial organization of Bergmann fibers and the absence of typical demarcating glial scar after kainate-induced cerebellar lesion suggest distinct pattern of astrogliosis that presents an interesting model system to study the importance of glial scar in the recovery after ischemic brain insults.
Collapse
Affiliation(s)
- I Milenkovic
- Department of Neurobiology and Neurochemistry, Institute of Biological Research Sinisa Stankovic, Serbia and Montenegro
| | | | | | | | | | | | | |
Collapse
|
513
|
Zhou BY, Liu Y, Kim BO, Xiao Y, He JJ. Astrocyte activation and dysfunction and neuron death by HIV-1 Tat expression in astrocytes. Mol Cell Neurosci 2005; 27:296-305. [PMID: 15519244 DOI: 10.1016/j.mcn.2004.07.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Revised: 07/01/2004] [Accepted: 07/09/2004] [Indexed: 12/11/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) Tat protein plays an important role in HIV-associated neuropathogenesis. Astrocytosis and neuron death are two hallmarks of HIV-1 infection of the central nervous system (CNS). However, whether there is a direct link between Tat expression, astrocytosis and subsequent neuron death is not known. In this study, we expressed Tat in astrocytes and examined Tat effects on astrocyte function and subsequent neuronal survival. The results showed that Tat expression resulted in a significant increase in glial fibrillary acidic protein (GFAP) expression, a cellular marker of astrocyte activation or astrocytosis. The GFAP promoter-driven reporter gene assay showed that Tat transactivated GFAP expression at the transcriptional level. Furthermore, Tat expression markedly impaired glutamate uptake by astrocytes. Importantly, cell culture supernatants from Tat-expressing astrocytes induced dramatic neuron death. Taken together, these data provide evidence for the first time to directly link Tat expression in astrocytes to astrocytosis, astrocyte dysfunction, and subsequent neuron death. In addition, these data suggest that astrocyte dysfunction contributes, at least in part, to Tat neurotoxicity and subsequently HIV-associated neuropathogenesis.
Collapse
Affiliation(s)
- Betty Y Zhou
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
514
|
Ang ET, Wong PTH, Moochhala S, Ng YK. Cytokine changes in the horizontal diagonal band of Broca in the septum after running and stroke: a correlation to glial activation. Neuroscience 2005; 129:337-47. [PMID: 15501591 DOI: 10.1016/j.neuroscience.2004.06.087] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2004] [Indexed: 01/11/2023]
Abstract
The relationship between running, glial cell activation and pro-inflammatory cytokines was studied in the context of neuroprotection against ischemic stroke induced by middle cerebral artery occlusion (MCAO). This was investigated in four groups of rats, namely, (1) nonrunner, (2) runner after 12 weeks of treadmill running, (3) nonrunner with MCAO and (4) runner with MCAO. The horizontal diagonal band of Broca (HDB) in the septum was scrutinized for qualitative cum quantitative changes in the microglia and astrocytes. Reverse transcription-polymerase chain reaction and immunoblot work were carried out in the forebrain homogenate to determine, respectively, the gene and protein expression of several pro-inflammatory cytokines. Our results indicated that the runner exhibited less immunoreactivity and reduced numbers of glial cells within the HDB compared with the nonrunner. Interestingly, the mRNA and protein levels of tumor necrosis factor-alpha, interleukin (IL)-1beta, IL-6 and interferon-gamma, were significantly downregulated in the runner. Our data also suggest albeit with some inconsistency that the runner/MCAO rats had benefited from running. These observations suggest that running can result in changes to the microenvironment, in which the microglia and astrocytes exist in a state of quiescence concomitant with a reduced expression of pro-inflammatory cytokines, that may lead to beneficial effects seen in ischemic stroke induced by MCAO.
Collapse
Affiliation(s)
- E T Ang
- Department of Anatomy, Faculty of Medicine, National University of Singapore, Defence Medical and Environmental Research Institute, MD10, 4 Medical Drive, Singapore 117597
| | | | | | | |
Collapse
|
515
|
Ciriza I, Carrero P, Azcoitia I, Lundeen SG, Garcia-Segura LM. Selective estrogen receptor modulators protect hippocampal neurons from kainic acid excitotoxicity: differences with the effect of estradiol. ACTA ACUST UNITED AC 2005; 61:209-21. [PMID: 15389604 DOI: 10.1002/neu.20043] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neuroprotective effects of estradiol are well characterized in animal experimental models. However, in humans, the outcome of estrogen treatment for cognitive function and neurological diseases is very controversial. Selective estrogen receptor modulators (SERMs) may represent an alternative to estrogen for the treatment or the prevention of neurodegenerative disorders. SERMs interact with the estrogen receptors and have tissue-specific effects distinct from those of estradiol, acting as estrogen agonists in some tissues and as antagonists in others. In this study we have assessed the effect of tamoxifen, raloxifene, lasofoxifene (CP-336,156), bazedoxifene (TSE-424), and 17beta-estradiol on the hippocampus of adult ovariectomized rats, after the administration of the excitotoxin kainic acid. Administration of kainic acid induced the expression of vimentin in reactive astroglia and a significant neuronal loss in the hilus. SERMs did not affect vimentin immunoreactivity in the hilus, while 17beta-estradiol significantly reduced the surface density of vimentin immunoreactive profiles. Estradiol, tamoxifen (0.4-2 mg/kg), raloxifene (0.4-2 mg/kg), and bazedoxifene (2 mg/kg) prevented neuronal loss in the hilus after the administration of kainic acid. Lasofoxifene (0.4-2 mg/kg) was not neuroprotective. These findings indicate that SERMs present different dose-dependent neuroprotective effects. Furthermore, the mechanisms of neuroprotection by SERMs and estradiol are not identical, because SERMs do not significantly affect reactive gliosis while neuroprotection by estradiol is associated with a strong down-regulation of reactive astroglia.
Collapse
|
516
|
Dittmann J, Fung SJ, Vickers JC, Chuah MI, Chung RS, West AK. Metallothionein biology in the ageing and neurodegenerative brain. Neurotox Res 2005; 7:87-93. [PMID: 15639801 DOI: 10.1007/bf03033779] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In recent years metallothionein (MT) biology has moved from investigation of its ability to protect against environmental heavy metals to a wider appreciation of its role in responding to cellular stress, whether as a consequence of normal function, or following injury and disease. This is exemplified by recent investigation of MT in the mammalian brain where plausible roles for MT action have been described, including zinc metabolism, free radical scavenging, and protection and regeneration following neurological injury. Along with other laboratories we have used several models of central nervous system (CNS) injury to investigate possible parallels between injury-dependent changes in MT expression and those observed in the ageing and/or degenerating brain. Therefore, this brief review aims to summarise existing information on MT expression during CNS ageing, and to examine the possible involvement of this protein in the course of human neurodegenerative disease, as exemplified by Alzheimer's disease.
Collapse
Affiliation(s)
- J Dittmann
- NeuroRepair Group, School of Medicine, University of Tasmania, Tasmania 7001 Australia
| | | | | | | | | | | |
Collapse
|
517
|
Poulsen CB, Penkowa M, Borup R, Nielsen FC, Cáceres M, Quintana A, Molinero A, Carrasco J, Giralt M, Hidalgo J. Brain response to traumatic brain injury in wild-type and interleukin-6 knockout mice: a microarray analysis. J Neurochem 2005; 92:417-32. [PMID: 15663489 DOI: 10.1111/j.1471-4159.2004.02877.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traumatic injury to the brain is one of the leading causes of injury-related death or disability. Brain response to injury is orchestrated by cytokines, such as interleukin (IL)-6, but the full repertoire of responses involved is not well known. We here report the results obtained with microarrays in wild-type and IL-6 knockout mice subjected to a cryolesion of the somatosensorial cortex and killed at 0, 1, 4, 8 and 16 days post-lesion. Overall gene expression was analyzed by using Affymetrix genechips/oligonucleotide arrays with approximately 12,400 probe sets corresponding to approximately 10,000 different murine genes (MG_U74Av2). A robust, conventional statistical method (two-way anova) was employed to select the genes significantly affected. An orderly pattern of gene responses was clearly detected, with genes being up- or down-regulated at specific timings consistent with the processes involved in the initial tissue injury and later regeneration of the parenchyma. IL-6 deficiency showed a dramatic effect in the expression of many genes, especially in the 1 day post-lesion timing, which presumably underlies the poor capacity of IL-6 knockout mice to cope with brain damage. The results highlight the importance of IL-6 controlling the response of the brain to injury as well as the suitability of microarrays for identifying specific targets worthy of further study.
Collapse
Affiliation(s)
- Christian Bjørn Poulsen
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
518
|
Holley JE, Gveric D, Whatmore JL, Gutowski NJ. Tenascin C induces a quiescent phenotype in cultured adult human astrocytes. Glia 2005; 52:53-8. [PMID: 15892123 DOI: 10.1002/glia.20231] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Astrocytic scar formation occurs subsequent to brain and spinal cord injury and impedes repair. The exact mechanisms of scar formation have yet to be elucidated but it is known that astrocytes within the scar have a different antigenic phenotype from normal or reactive astrocytes. Astrocyte cell culture offers a suitable system to identify factors that induce the scar phenotype as well as factors that reverse this process and that may help identify therapeutic strategies to treat astrogliosis. However, when placed in standard culture conditions, astrocytes become activated/reactive and express molecules characteristic of scar tissue in vivo. In the present study, we made use of this phenomenon to identify culture conditions that change the activated phenotype of cultured astrocytes into one characteristic of normal quiescent astrocytes. In particular, we examined the effect of extracellular matrix (ECM) proteins found in the human brain, on the phenotype of human adult astrocytes. Significantly fewer astrocytes expressed scar properties when grown on tenascin-C (TN-C) than those cultured on other ECM proteins or poly-L-lysine-coated dishes. TN-C also significantly reduced the proliferation rate of the astrocytes in vitro. In addition, further manipulation of culture conditions induced partial astrocyte reactivation. Our findings suggest that astrocytes grown on TN-C revert to a quiescent, nonactivated state that is partially reversible. This raises the possibility that therapeutic strategies aimed at manipulating TN-C levels during CNS injury may help reduce astrocytic scarring.
Collapse
Affiliation(s)
- Janet E Holley
- Institute of Biomedical and Clinical Sciences, Peninsula Medical School (Exeter), Department of Neurology, Royal Devon and Exeter Hospital, Exeter, United Kingdom.
| | | | | | | |
Collapse
|
519
|
Slamon ND, Mead C, Morgan C, Mitchell A, Pentreath VW. The involvement of calcium in the protective and toxic (nonlinear) responses of rodent and human astroglial cells. NONLINEARITY IN BIOLOGY, TOXICOLOGY, MEDICINE 2005; 3:79-95. [PMID: 19330156 PMCID: PMC2657843 DOI: 10.2201/nonlin.003.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The involvement of [Ca(2+)](i) in the reactive changes of astrocytes which accompany exposure to different chemicals were studied in cultures of C6 and 1321N1 cells. Cells were exposed to up to three serial pulses of the differentiating agent dBcAMP, which induces activation-type changes in the cells. Other cells, with or without the dBcAMP treatments, were treated with a range of concentrations of the antidepressants amitriptyline and fluoxetine and the glial toxicants acrylamide and chloroquine. In some experiments the L-type voltage calcium channel blocker Nifedipine was employed. [Ca(2+)](i) was measured in populations of the cells using Fura-2AM and a charge coupled device (CCD) camera attached to a fluorescence microscope. dBcAMP induced both dose- and time-dependent changes in [ Ca(2+)](i) with increases in both the [Ca(2+)](i) oscillations and mean [Ca(2+)](i) (e.g. in C6 cells at 18 min mean [Ca(2+)](i) was 318 +/- 20nM following the single differentiating dBcAMP pulses, 489 +/- 17nM (p < 0.001) following two serial pulses, and 275 +/- 30nM (not significant) following three pulses). Therapeutic doses of fluoxetine and amitriptyline caused increases in the calcium oscillations and the mean calcium concentrations ( maximum recorded mean increase was in the C6 cells at 10min by 0.02 muM fluoxetine when [Ca(2+)](i) was 411 +/- 35nM c.f. control 254 +/- 25nM, p = 0.01). Higher (non-therapeutic) doses of both antidepressants caused significant reductions. Chloroquine and acrylamide also caused dose-dependent bi-phasic types of alterations in [Ca(2+)](i), with significant reductions at lower, sub-cytotoxic doses followed by significant increases at higher concentrations, approaching those which cause cell damage. Nifedipine treatment caused some reductions in the dBcAMP, antidepressant or toxicant-induced calcium changes, but this substance also initiated cytotoxic alterations. The findings show that both the activation-type changes (which are frequently associated with increased protective capacities) and toxic responses of C6 and 1321N1 cells to different chemical agents are associated with dose-dependent alterations in [Ca(2+)](i).
Collapse
Affiliation(s)
- N Debbie Slamon
- Division of Biosciences, University of Salford, Salford, U.K
| | | | | | | | | |
Collapse
|
520
|
Grimpe B, Pressman Y, Lupa MD, Horn KP, Bunge MB, Silver J. The role of proteoglycans in Schwann cell/astrocyte interactions and in regeneration failure at PNS/CNS interfaces. Mol Cell Neurosci 2005; 28:18-29. [PMID: 15607938 DOI: 10.1016/j.mcn.2004.06.010] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2004] [Revised: 06/15/2004] [Accepted: 06/15/2004] [Indexed: 11/19/2022] Open
Abstract
In the dorsal root entry zone (DREZ) peripheral sensory axons fail to regenerate past the peripheral nervous system/central nervous system (PNS/CNS) interface. Additionally, in the spinal cord, central fibers that regenerate into Schwann cell (SC) bridges can enter but do not exit at the distal Schwann cell/astrocyte (AC) boundary. At both interfaces where limited mixing of the two cell types occurs, one can observe an up-regulation of inhibitory chondroitin sulfate proteoglycans (CSPGs). We treated confrontation Schwann cell/astrocyte cultures with the following: (1) a deoxyribonucleic acid (DNA) enzyme against the glycosaminoglycan (GAG)-chain-initiating enzyme, xylosyltransferase-1 (XT-1), (2) a control DNA enzyme, and (3) chondroitinase ABC (Ch'ase ABC) to degrade the GAG chains. Both techniques for reducing CSPGs allowed Schwann cells to penetrate deeply into the territory of the astrocytes. After adding sensory neurons to the assay, the axons showed different growth behaviors depending upon the glial cell type that they first encountered during regeneration. Our results help to explain why regeneration fails at PNS/CNS glial boundaries.
Collapse
Affiliation(s)
- Barbara Grimpe
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
521
|
Barbeito LH, Pehar M, Cassina P, Vargas MR, Peluffo H, Viera L, Estévez AG, Beckman JS. A role for astrocytes in motor neuron loss in amyotrophic lateral sclerosis. ACTA ACUST UNITED AC 2004; 47:263-74. [PMID: 15572176 DOI: 10.1016/j.brainresrev.2004.05.003] [Citation(s) in RCA: 243] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2004] [Indexed: 12/31/2022]
Abstract
A strong glial reaction typically surrounds the affected upper and lower motor neurons and degenerating descending tracts of ALS patients. Reactive astrocytes in ALS contain protein inclusions, express inflammatory makers such as the inducible forms of nitric oxide synthase (iNOS) and cyclooxygenase (COX-2), display nitrotyrosine immunoreactivity and downregulate the glutamate transporter EAAT2. In this review, we discuss the evidence sustaining an active role for astrocytes in the induction and propagation of motor neuron loss in ALS. Available evidence supports the view that glial activation could be initiated by proinflammatory mediators secreted by motor neurons in response to injury, axotomy or muscular pathology. In turn, reactive astrocytes produce nitric oxide and peroxynitrite, which cause mitochondrial damage in cultured neurons and trigger apoptosis in motor neurons. Astrocytes may also contribute to the excitotoxic damage of motor neurons by decreasing glutamate transport or actively releasing the excitotoxic amino acid. In addition, reactive astrocytes secrete pro-apoptotic mediators, such as nerve growth factor (NGF) or Fas-ligand, a mechanism that may serve to eliminate vulnerable motor neurons. The comprehensive understanding of the interactions between motor neurons and glia in ALS may lead to a more accurate theory of the pathogenesis of the disease.
Collapse
Affiliation(s)
- Luis H Barbeito
- Departamento de Neurobiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable, Avenida Italia 3318-CP 11600, Montevideo, Uruguay.
| | | | | | | | | | | | | | | |
Collapse
|
522
|
Crestini A, Zona C, Sebastiani P, Pieri M, Caracciolo V, Malvezzi-Campeggi L, Confaloni A, Di Loreto S. Effects of simulated microgravity on the development and maturation of dissociated cortical neurons. In Vitro Cell Dev Biol Anim 2004; 40:159-65. [PMID: 15479120 DOI: 10.1290/1543-706x(2004)40<159:eosmot>2.0.co;2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although a wealth of evidence supports the hypothesis that some functions of the nervous system may be altered during exposure to microgravity, the possible changes in basic neuronal physiology are not easy to assess. Indeed, few studies have examined whether microgravity affects the development of neurons in culture. In the present study, a suspension of dissociated cortical cells from rat embryos were exposed to 24 h of simulated microgravity before plating in a normal adherent culture system. Both preexposed and control cells were used after a period of 7-10 d in vitro. The vitality and the level of reactive oxygen species of cultures previously exposed did not differ from those of normal cultures. Cellular characterization by immunostaining with a specific antibody displayed normal neuronal phenotype in control cells, whereas pretreatment in simulated microgravity revealed an increase of glial fibrillary acidic protein fluorescence in the elongated stellate glial cells. Electrophysiological recording indicated that the electrical properties of neurons preexposed were comparable with those of controls. Overall, our results indicate that a short time of simulated microgravity preexposure does not affect dramatically the ability of dissociated neural cells to develop and differentiate in an adherent culture system.
Collapse
Affiliation(s)
- Alessio Crestini
- Department of Cellular Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
523
|
Hampton DW, Rhodes KE, Zhao C, Franklin RJM, Fawcett JW. The responses of oligodendrocyte precursor cells, astrocytes and microglia to a cortical stab injury, in the brain. Neuroscience 2004; 127:813-20. [PMID: 15312894 DOI: 10.1016/j.neuroscience.2004.05.028] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2004] [Indexed: 10/26/2022]
Abstract
The cortical stab injury has been widely used for biochemical analysis of molecular changes following CNS injury. However, the cellular responses to this injury have not been accurately quantified. In order to provide a baseline for biochemical studies and future experiments on the manipulation of the CNS injury response we have undertaken a quantitative analysis of this injury. The proliferative and reactive responses of oligodendrocyte precursor cells, astrocytes and microglia were measured, using antibodies to NG2, glial fibrillary acidic protein (GFAP) and the cd11-b clone OX-42 to characterise these cell types at 2, 4, 7 and 14 days post-injury. Oligodendrocyte precursors and microglia proliferated rapidly during the first week, mostly within 0.3 mm of the lesion. Of the dividing cells over 60% were oligodendrocyte precursor cells with microglia making up the balance of the dividing cells. Minimal numbers of astrocytes divided in response to the lesion. Large cells with one or two short processes that were both NG2 and OX-42 positive were identified very close to the lesion at 2 and 4 days post-lesion but not thereafter. They are likely to be blood-derived cells that express NG2 or have ingested it. NG2 immunohistochemistry and platelet-derived growth factor alpha receptor (PDGFalpha-R) in situ hybridisation on neighbouring sections was performed. In the lesioned area only 12% of NG2 positive (+ive) cells were PDGFalpha-R +ive (a ratio of 1:8 for PDGFalpha-R +ive cells: NG2 +ive cells) compared with 33% in the unlesioned cortex and an almost 100% overlap in the spinal cord.
Collapse
Affiliation(s)
- D W Hampton
- Cambridge Centre for Brain Repair, E. D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 2PY, UK.
| | | | | | | | | |
Collapse
|
524
|
Burbach GJ, Dehn D, Nagel B, Del Turco D, Deller T. Laser microdissection of immunolabeled astrocytes allows quantification of astrocytic gene expression. J Neurosci Methods 2004; 138:141-8. [PMID: 15325122 DOI: 10.1016/j.jneumeth.2004.03.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2004] [Revised: 03/29/2004] [Accepted: 03/30/2004] [Indexed: 10/26/2022]
Abstract
Astrocytes represent the major glial cell population within the central nervous system. In order to elucidate the function of astrocytes under physiological conditions and during the course of neurological disease, astrocytic gene expression profiling is necessary. However, since astrocytes form an intimately connected network with neurons and other cell types in the brain, gene expression analysis of astrocytes with a sufficient degree of cellular specificity is difficult. Here we are presenting a rapid and, thus, RNA preserving immunostaining protocol for the detection of astrocytes in rodent brain. This protocol can readily be combined with laser microdissection (Leica AS LMD platform) and quantitative RT-PCR (qPCR). Employing this method, we studied changes in glial fibrillary acidic protein (GFAP) expression in astrocytes of mouse entorhinal cortex following entorhinal cortex lesion. Using laser microdissection, astrocytes (n = 60) were collected in the tissue surrounding the lesion, the entorhinal cortex contralateral to the lesion, and in unlesioned control animals. Changes in GFAP mRNA were quantified using qPCR. GFAP mRNA levels were 82-fold higher in astrocytes of lesioned animals at the site of the lesion compared to GFAP mRNA levels in entorhinal cortex astrocytes of control mice. GFAP mRNA levels were only slightly elevated at the contralateral side (lesioned animals). This optimized protocol for immunolabeling and laser microdissection of astrocytes followed by qPCR allows quantification of astrocytic gene expression levels with a high degree of cellular specificity. It may similarly be employed in different settings where other cell types need to be identified and microdissected for gene expression profiling.
Collapse
Affiliation(s)
- Guido J Burbach
- Institute of Clinical Neuroanatomy, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
525
|
Dervan AG, Meshul CK, Beales M, McBean GJ, Moore C, Totterdell S, Snyder AK, Meredith GE. Astroglial plasticity and glutamate function in a chronic mouse model of Parkinson's disease. Exp Neurol 2004; 190:145-56. [PMID: 15473988 DOI: 10.1016/j.expneurol.2004.07.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2004] [Revised: 06/25/2004] [Accepted: 07/08/2004] [Indexed: 10/26/2022]
Abstract
Astrocytes play a major role in maintaining low levels of synaptically released glutamate, and in many neurodegenerative diseases, astrocytes become reactive and lose their ability to regulate glutamate levels, through a malfunction of the glial glutamate transporter-1. However, in Parkinson's disease, there are few data on these glial cells or their regulation of glutamate transport although glutamate cytotoxicity has been blamed for the morphological and functional decline of striatal neurons. In the present study, we use a chronic mouse model of Parkinson's disease to investigate astrocytes and their relationship to glutamate, its extracellular level, synaptic localization, and transport. C57/bl mice were treated chronically with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and probenecid (MPTP/p). From 4 to 8 weeks after treatment, these mice show a significant loss of dopaminergic terminals in the striatum and a significant increase in the size and number of GFAP-immunopositive astrocytes. However, no change in extracellular glutamate, its synaptic localization, or transport kinetics was detected. Nevertheless, the density of transporters per astrocyte is significantly reduced in the MPTP/p-treated mice when compared to controls. These results support reactive gliosis as a means of striatal compensation for dopamine loss. The reduction in transporter complement on individual cells, however, suggests that astrocytic function may be compromised. Although reactive astrocytes are important for maintaining homeostasis, changes in their ability to regulate glutamate and its associated synaptic functions could be important for the progressive nature of the pathophysiology associated with Parkinson's disease.
Collapse
Affiliation(s)
- Adrian G Dervan
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | | | | | | | | | | | | | | |
Collapse
|
526
|
Aronica E, Ozbas-Gerçeker F, Redeker S, Ramkema M, Spliet WGM, van Rijen PC, Leenstra S, Gorter JA, Troost D. Expression and cellular distribution of high- and low-affinity neurotrophin receptors in malformations of cortical development. Acta Neuropathol 2004; 108:422-34. [PMID: 15375667 DOI: 10.1007/s00401-004-0906-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2004] [Revised: 06/28/2004] [Accepted: 06/28/2004] [Indexed: 12/01/2022]
Abstract
An increasing number of observations suggests an important and complex role for both high- (tyrosine kinase receptor, trk) and low- (p75) affinity neurotrophin receptors (NTRs) during development in human brain. In the present study, the cell-specific distribution of NTRs was studied in different developmental lesions, including focal cortical dysplasia (FCD, n = 15), ganglioglioma (GG, n = 15) and dysembryoplastic neuroepithelial tumors, (DNT, n = 10), from patients with medically intractable epilepsy. Lesional, perilesional, as well as normal brain regions were examined for the expression of trkA, trkB, trkC and p75(NTR) by immunocytochemistry. In normal postmortem human cortex, immunoreactivity (IR) for trk and p75(NTR) was mainly observed in pyramidal neurons, whereas no notable glial IR was found within the white matter. All three trk receptors were encountered in high levels in the neuronal component of the majority of FCD, GG and DNT specimens. Strong trkA, trkB and trkC IR was found in neurons of different size, including large dysplastic neurons and balloon cells in FCD cases. In contrast, p75(NTR) IR was observed in only a small number of neuronal cells, which also contain trk receptors. Glial cells with astrocytic morphology showed predominantly IR for trkA in FCD and GG specimens, whereas oligodendroglial-like cells in DNT showed predominently IR for trkB. P75(NTR) IR was observed in a population of cells of the microglial/macrophage lineage in both FCD and glioneuronal tumors. Taken together, our findings indicate that the neuronal and the glial components of malformations of cortical development express both high- and low-affinity NTRs. Further research is necessary to investigate how activation of these specific receptors could contribute to the development and the epileptogenicity of these developmental disorders.
Collapse
Affiliation(s)
- Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
527
|
Srinivasan J, Schmidt WJ. Treatment with alpha2-adrenoceptor antagonist, 2-methoxy idazoxan, protects 6-hydroxydopamine-induced Parkinsonian symptoms in rats: neurochemical and behavioral evidence. Behav Brain Res 2004; 154:353-63. [PMID: 15313023 DOI: 10.1016/j.bbr.2004.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2004] [Revised: 03/02/2004] [Accepted: 03/04/2004] [Indexed: 11/22/2022]
Abstract
Noradrenaline, not only functions as a synaptic transmitter, but also promotes neural differentiation and regenerative processes. In Parkinson's disease, besides the dopaminergic degeneration, noradrenergic neurons of locus coeruleus origin degenerate as well. Drugs enhancing noradrenergic transmission in the locus coeruleus (e.g. alpha2-adrenoceptor antagonists) have been shown to be neuroprotective against Huntington's and ischemic animal models. However, in Parkinsonian animal models, most of the studies evaluated the worsening of experimental nigral neurodegeneration after locus coeruleus lesions. Here, it has been tested, whether treatment with the selective alpha2-adrenoceptor antagonist, 2-methoxy idazoxan (2.5 mg/kg i.p., twice daily for 5 days), before an experimental lesion to nigra, protects dopaminergic neurodegeneration. Dopaminergic degeneration was produced by 6-hydroxydopamine lesion in the median forebrain bundle. The concentrations of dopamine, 5-hydroxytryptamine and its metabolites were analysed in the various regions of the basal ganglia. The concentrations of noradrenaline and dopamine were measured in the regions innervated by locus coeruleus neurons and in the basal ganglia respectively, after 2-methoxy idazoxan treatment. The Parkinsonian behavior was assessed by catalepsy and activity test. 2-Methoxy idazoxan specifically increased the concentration of noradrenaline in the brain regions, innervated by locus coeruleus neurons. 6-OHDA lesion strongly depleted the concentration of dopamine and its metabolites in the striatum and SN, producing catalepsy and hypoactivity. Multiple treatments with 2-methoxy idazoxan reduced some of the observed neurochemical and behavioral indices of 6-hydroxydopamine-induced Parkinsonism, indicating neuroprotection. Although the mechanism underlying the neuroprotective property remains elusive, the therapeutic usage of alpha2-antagonists might be helpful in slowing the neuronal death and progression of Parkinson's disease.
Collapse
Affiliation(s)
- J Srinivasan
- Neuropharmacology, Zoological Institute, University of Tuebingen, Auf der Morgenstelle 28E, 72076 Tuebingen, Germany
| | | |
Collapse
|
528
|
Konwinski RR, Haddad R, Chun JA, Klenow S, Larson SC, Haab BB, Furge LL. Oltipraz, 3H-1,2-dithiole-3-thione, and sulforaphane induce overlapping and protective antioxidant responses in murine microglial cells. Toxicol Lett 2004; 153:343-55. [PMID: 15454310 DOI: 10.1016/j.toxlet.2004.06.006] [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] [Received: 04/20/2004] [Revised: 06/12/2004] [Accepted: 06/14/2004] [Indexed: 11/24/2022]
Abstract
Oltipraz (OPZ) is a known inducer of glutathione S-transferases and a mechanism-based inhibitor of cytochrome P450 1A2. Given the detoxification characteristics of this compound, the transcriptional effects of OPZ, along with the related naturally occurring compounds 3H-1,2-dithiole-3-thione (D3T) and sulforaphane (SF), were examined by gene expression profiling in murine BV-2 microglial cells, a neuronal macrophage cell type that mediates inflammatory responses in the brain. We show that the three compounds generate largely overlapping transcriptional changes in genes that are associated with detoxification and antioxidant responses. In addition, induction of an antioxidant/detoxification response in the microglial cells by OPZ, D3T, or SF was also able to protect cells from H2O2 -induced toxicity and to attenuate the production of reactive oxygen species in response to lipopolysaccharide treatment of cells. These results show that OPZ, D3T, and SF activate overlapping changes in gene expression and that they can regulate detoxification/antioxidant responses in multiple cells types, including cell types known to have a role in the production of oxidative stress.
Collapse
Affiliation(s)
- Ryan R Konwinski
- Department of Chemistry, Kalamazoo College, 1200 Academy Street, Kalamazoo, MI 49006, USA
| | | | | | | | | | | | | |
Collapse
|
529
|
Ostrow LW, Sachs F. Mechanosensation and endothelin in astrocytes--hypothetical roles in CNS pathophysiology. ACTA ACUST UNITED AC 2004; 48:488-508. [PMID: 15914254 DOI: 10.1016/j.brainresrev.2004.09.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2003] [Revised: 08/19/2004] [Accepted: 09/09/2004] [Indexed: 01/23/2023]
Abstract
Endothelin (ET) is a potent autocrine mitogen produced by reactive and neoplastic astrocytes. ET has been implicated in the induction of astrocyte proliferation and other transformations engendered by brain pathology, and in promoting the malignant behavior of astrocytomas. Reactive astrocytes containing ET are found in the periphery/penumbra of a wide array of CNS pathologies. Virtually all brain pathology deforms the surrounding parenchyma, either by direct mass effect or edema. Mechanical stress is a well established stimulus for ET production and release by other cell types, but has not been well studied in the brain. However, numerous studies have illustrated that astrocytes can sense mechanical stress and translate it into chemical messages. Furthermore, the ubiquitous reticular meshwork formed by interconnected astrocytes provides an ideal morphology for sensing and responding to mechanical disturbances. We have recently demonstrated stretch-induced ET production by astrocytes in vitro. Inspired by this finding, the purpose of this article is to review the literature on (1) astrocyte mechanosensation, and (2) the endothelin system in astrocytes, and to consider the hypothesis that mechanical induction of the ET system may influence astrocyte functioning in CNS pathophysiology. We conclude by discussing evidence supporting future investigations to determine whether specific inhibition of stretch-activated ion channels may represent a novel strategy for treating or preventing CNS disturbances, as well as the relevance to astrocyte-derived tumors.
Collapse
Affiliation(s)
- Lyle W Ostrow
- Department of Physiology and Biophysics, S.U.N.Y. at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | | |
Collapse
|
530
|
Stanic D, Tripanichkul W, Drago J, Finkelstein DI, Horne MK. Glial responses associated with dopaminergic striatal reinnervation following lesions of the rat substantia nigra. Brain Res 2004; 1023:83-91. [PMID: 15364022 DOI: 10.1016/j.brainres.2004.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2004] [Indexed: 11/22/2022]
Abstract
Lesioning of dopaminergic substantia nigra pars compacta (SNpc) neurons leads to depletion of dopamine (DA) and dopaminergic axons in the dorsal striatum, followed by subsequent compensatory sprouting of dopaminergic fibers and striatal reinnervation. In this study, the response of striatal glia (microglia and astroglia) was compared with the degeneration and regeneration of dopaminergic axons following SNpc lesions. Following partial SNpc lesions, density of dopamine transporter (DAT) immunoreactive (-ir) terminals in the dorsal striatum returned to normal within 16 weeks of injury, suggesting that dopaminergic reinnervation of the striatum was complete. In conjunction, the glial responses in the dorsal striatum consisted of two peaks. The first peak in glial density occurred immediately after lesioning, peaking at 7 days, implying that it was likely to be associated with removal of debris from degenerating terminals. The second glial response commenced 8 weeks after lesioning and peaked some time after 16 weeks. The time of onset of the second peak suggests that it may be associated with the establishment of synapses rather than with axonal guidance.
Collapse
Affiliation(s)
- Davor Stanic
- Department of Medicine, Monash University, Monash Medical Centre, Block E, Level 5, 246 Clayton Rd, Clayton 3168, Australia
| | | | | | | | | |
Collapse
|
531
|
Facchinetti F, Del Giudice E, Furegato S, Passarotto M, Arcidiacono D, Leon A. Dopamine inhibits responses of astroglia-enriched cultures to lipopolysaccharide via a beta-adrenoreceptor-mediated mechanism. J Neuroimmunol 2004; 150:29-36. [PMID: 15081246 DOI: 10.1016/j.jneuroim.2004.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2003] [Revised: 12/23/2003] [Accepted: 01/12/2004] [Indexed: 11/21/2022]
Abstract
We here investigated the effect of the catecholaminergic neurotransmitter dopamine (DA), on the release of two major inflammatory effectors, TNF-alpha and nitric oxide, in rat astroglia-enriched cultures stimulated with the bacterial endotoxin lipopolysaccharide (LPS). Upon LPS challenge, we observed a dramatic increase in the culture medium of the TNF-alpha protein, an effect thereafter followed by an increase of nitric oxide synthase type 2 (NOS2) mRNA and, at later times, of nitrite accumulation, an index of nitric oxide (NO) production. DA substantially inhibited the release of TNF-alpha and NO evoked by LPS, an effect not mimicked by selective agonists nor prevented by selective antagonists of the DA receptors. The inhibitory effects of DA were mimicked by noradrenalin and isoproterenol and fully reverted by propranolol, a selective antagonist of the beta-adrenergic receptors. In addition, selective antagonists of beta-adrenergic receptor type 1 (metoprolol) and type 2 (ICI-118,551) counteracted the inhibitory effects of DA on LPS-induced TNF-alpha and NO release. Accordingly, agents capable of elevating intracellular cyclic 3',5'-adenosine monophosphate (cAMP), such as forskolin and dibutyryl-cAMP, mimicked DA inhibitory effects on LPS-evoked accumulation of TNF-alpha and nitrite. These data, consistent with a role of DA as local modulator of glial inflammatory responses, uncover the existence of an interaction between DA and heterologous beta-adrenergic receptors in astroglial cells.
Collapse
Affiliation(s)
- Fabrizio Facchinetti
- Neurobiology Unit, Research and Innovation Company, via Svizzera 16, 35127 Padova, Italy.
| | | | | | | | | | | |
Collapse
|
532
|
Ahmed MM, Hoshino H, Chikuma T, Yamada M, Kato T. Effect of memantine on the levels of glial cells, neuropeptides, and peptide-degrading enzymes in rat brain regions of ibotenic acid-treated alzheimer's disease model. Neuroscience 2004; 126:639-49. [PMID: 15183513 DOI: 10.1016/j.neuroscience.2004.04.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2004] [Indexed: 02/06/2023]
Abstract
It has been implicated that glia activation plays a critical role in the progression of Alzheimer's disease (AD). However, the precise mechanism of glia activation is not clearly understood yet. In our present studies, we confirmed our previous results where change the levels of neuropeptides and peptidases in ibotenic acid (IBO) infusion into the rat nucleus basalis magnocellularis, an animal model of AD. Furthermore, we extended our study to investigate a possible protection effect of co-administration on the changes of neuropeptides, and neuronal and glial cells in IBO-infused rat brain by memantine treatment. The levels of substance P and somatostatin were decreased in the striatum and frontal cortex 1 week after IBO infusion, and recovered to the control level by memantine treatment, indicating the involvement of neuropeptides in AD pathology. Furthermore, the immunohistochemical and enzymatic studies of GFAP and CD 11b, and peptidylarginine deiminase, markers of glia, in the striatum and frontal cortex showed the increase in IBO-treated rat brain as compared with controls, while co-administration of memantine and IBO no increase of astrocytes and microglia activation was observed. The present biochemical and immunohistochemical results suggest that glia activation might play an important role to the pathology of AD, and correlate with the changes of neuropeptide levels in AD brain that is recovered by memantine treatment.
Collapse
Affiliation(s)
- M M Ahmed
- Laboratory of Natural Information Science, Graduate School of Integrated Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | | | | | | | | |
Collapse
|
533
|
Chesik D, Kühl NM, Wilczak N, De Keyser J. Enhanced production and proteolytic degradation of insulin-like growth factor binding protein-2 in proliferating rat astrocytes. J Neurosci Res 2004; 77:354-62. [PMID: 15248291 DOI: 10.1002/jnr.20172] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Insulin-like growth factors (IGFs) protect neurons, are important for oligodendrocyte survival and myelin production, and stimulate the proliferation of astrocytes. The effects of IGFs are regulated by a family of IGF binding proteins (IGFBPs). Astrocytes express predominantly IGFBP-2. In the present study, primary neonatal rat astrocytes were cultivated in a chemically defined medium to initiate a differentiated cell status. After stimulation with fetal calf serum, astrocytes became hypertrophic and increased proliferation. Western blot analysis of cell lysate of proliferating astrocytes displayed an increased expression of IGFBP-2. This finding was supported by immunocytochemical images. Semiquantitative polymerase chain reaction analysis demonstrated equal mRNA levels in both differentiated and proliferating astrocytes, suggesting that the increase in IGFBP-2 production in proliferating astrocytes was exerted at the translational level. Concentrated medium of proliferating cells, however, displayed lower levels of IGFBP-2 than differentiated cells. When recombinant IGFBP-2 was incubated with culture media, we found degradation in the medium of proliferating cells, but not in medium of differentiated cells. This degradation could be inhibited with protease inhibitors, indicating that lower levels of IGFBP-2 in the medium of proliferating astrocytes are due to the presence of proteases. Our results suggest that, in proliferating astrocytes, IGFBP-2 may help target IGFs to IGF-1 receptors, and IGFBP-2 proteases may play a role in enhancing the availability of IGFs.
Collapse
Affiliation(s)
- Daniel Chesik
- Department of Neurology, University Hospital Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
534
|
Yasuda Y, Tateishi N, Shimoda T, Satoh S, Ogitani E, Fujita S. Relationship between S100β and GFAP expression in astrocytes during infarction and glial scar formation after mild transient ischemia. Brain Res 2004; 1021:20-31. [PMID: 15328028 DOI: 10.1016/j.brainres.2004.06.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2004] [Indexed: 11/28/2022]
Abstract
The expression of astrocyte marker proteins (S100beta and GFAP) during infarction and glial scar formation after transient middle cerebral artery (MCA) occlusion was examined using double immunostaining. S100beta immunoreactivity markedly decreased in the core of the injured area when observed immediately after reperfusion and did not increase again. In the periphery, however, S100beta expression increased, showing that S100beta synthesis was up-regulated. S100beta+/iNOS+ astrocytes in the periphery were observed from day 1, when small infarct areas were detectable, up to day 5, when infarct expansion had almost ended. TUNEL+ cells in the periphery were present from days 1 to 5. S100beta+/TUNEL+ cells were observed centrally and around the periphery of the injured area, predicting that cell death contributes to the increase of S100beta concentration in the injured area. Our results suggest that (1) higher concentration of S100beta in the extracellular space due to S100beta leakage from damaged astrocytes leads to up-regulation of S100beta synthesis and induction of inducible nitric oxide synthase (iNOS) synthesis in astrocytes, contributing to infarct expansion that results in DNA damage or cell death via NO and ROS production, and (2) GFAP, but not S100beta, is a main contributor to glial scar formation. On day 1 postreperfusion, the microdiascopic images of the injured areas from the unstained thick sections or the areas detected by S100beta immunoreactivity were larger than those of the infarct areas detected by hematoxylin--eosin (HE)-staining. The difference between these sizes might be useful to predict infarct expansion.
Collapse
Affiliation(s)
- Yuko Yasuda
- Cell Biology Section, Division of Basic Research, Louis Pasteur Center for Medical Research, 103-5 Sakyo, Tanaka, Monzen-cho, Kyoto 606-8225, Japan.
| | | | | | | | | | | |
Collapse
|
535
|
Raivich G, Bohatschek M, Da Costa C, Iwata O, Galiano M, Hristova M, Nateri AS, Makwana M, Riera-Sans L, Wolfer DP, Lipp HP, Aguzzi A, Wagner EF, Behrens A. The AP-1 transcription factor c-Jun is required for efficient axonal regeneration. Neuron 2004; 43:57-67. [PMID: 15233917 DOI: 10.1016/j.neuron.2004.06.005] [Citation(s) in RCA: 357] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Revised: 04/27/2004] [Accepted: 06/04/2004] [Indexed: 01/06/2023]
Abstract
Nerve injury triggers numerous changes in the injured neurons and surrounding nonneuronal cells that ultimately result in successful target reinnervation or cell death. c-Jun is a component of the heterodimeric AP-1 transcription factor, and c-Jun is highly expressed in response to neuronal trauma. Here we have investigated the role of c-jun during axonal regeneration using mice lacking c-jun in the central nervous system. After transection of the facial nerve, the absence of c-Jun caused severe defects in several aspects of the axonal response, including perineuronal sprouting, lymphocyte recruitment, and microglial activation. c-Jun-deficient motorneurons were atrophic, resistant to axotomy-induced cell death, and showed reduced target muscle reinnervation. Expression of CD44, galanin, and alpha7beta1 integrin, molecules known to be involved in regeneration, was greatly impaired, suggesting a mechanism for c-Jun-mediated axonal growth. Taken together, our results identify c-Jun as an important regulator of axonal regeneration in the injured central nervous system.
Collapse
Affiliation(s)
- Gennadij Raivich
- Perinatal Brain Repair Group, Department of Obstetrics and Gynaecology, University College London, 86-96 Chenies Mews, London WC1E 6HX, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
536
|
Teismann P, Schulz JB. Cellular pathology of Parkinson?s disease: astrocytes, microglia and inflammation. Cell Tissue Res 2004; 318:149-61. [PMID: 15338271 DOI: 10.1007/s00441-004-0944-0] [Citation(s) in RCA: 246] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Accepted: 06/22/2004] [Indexed: 10/26/2022]
Abstract
Parkinson's disease (PD) is a frequent neurological disorder of the basal ganglia, which is characterized by the progressive loss of dopaminergic neurons mainly in the substantia nigra pars compacta (SNpc). Inflammatory processes have been shown to be associated with the pathogenesis of PD. Activated microglia, as well as to a lesser extent reactive astrocytes, are found in the area associated with cell loss, possibly contributing to the inflammatory process by the release of pro-inflammatory prostaglandins or cytokines. Further deleterious factors released by activated microglia or astrocytes are reactive oxygen species. On the other hand, they may mediate neuroprotective properties by the release of trophic factors or the uptake of glutamate. In this review, we will discuss the different aspects of activated glial cells and potential mechanisms that mediate or protect against cell loss in PD.
Collapse
Affiliation(s)
- Peter Teismann
- Neurodegeneration Laboratory, Department of General Neurology, Center of Neurology and Hertie Institute for Clinical Brain Research, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany.
| | | |
Collapse
|
537
|
Falsig J, Pörzgen P, Lotharius J, Leist M. Specific Modulation of Astrocyte Inflammation by Inhibition of Mixed Lineage Kinases with CEP-1347. THE JOURNAL OF IMMUNOLOGY 2004; 173:2762-70. [PMID: 15294995 DOI: 10.4049/jimmunol.173.4.2762] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Inflammatory conversion of murine astrocytes correlates with the activation of various MAPK, and inhibition of terminal MAPKs like JNK or p38 dampens the inflammatory reaction. Mixed lineage kinases (MLKs), a family of MAPK kinase kinases, may therefore be involved in astrocyte inflammation. In this study, we explored the effect of the MLK inhibitors CEP-1347 and CEP-11004 on the activation of murine astrocytes by either TNF plus IL-1 or by a complete cytokine mix containing additional IFN-gamma. The compounds blocked NO-, PG-, and IL-6 release with a median inhibitory concentration of approximately 100 nM. This activity correlated with a block of the JNK and the p38 pathways activated in complete cytokine mix-treated astrocytes. Although CEP-1347 did not affect the activation of NF-kappaB, it blocked the expression of cyclooxygenase-2 and inducible NO synthase at the transcriptional level. Quantitative transcript profiling of 17 inflammation-linked genes revealed a specific modulation pattern of astrocyte activation by MLK inhibition, for instance, characterized by up-regulation of the anti-stress factors inhibitor of apoptosis protein-2 and activated transcription factor 4, no effect on manganese superoxide dismutase and caspase-11, and down-regulation of major inflammatory players like TNF, GM-CSF, urokinase-type plasminogen activator, and IL-6. In conclusion, MLK inhibitors like CEP-1347 are highly potent astrocyte immune modulators with a novel spectrum of activity.
Collapse
|
538
|
Norenberg MD, Smith J, Marcillo A. The pathology of human spinal cord injury: defining the problems. J Neurotrauma 2004; 21:429-40. [PMID: 15115592 DOI: 10.1089/089771504323004575] [Citation(s) in RCA: 442] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This article reviews the pathology of human spinal cord injury (SCI), focusing on potential differences between humans and experimental animals, as well as on aspects that may have mechanistic or therapeutic relevance. Importance is placed on astrocyte and microglial reactions. These cells carry out a myriad of functions and we review the evidence that supports their beneficial or detrimental effects. Likewise, vascular responses and the role of inflammation and demyelination in the mechanism of SCI are reviewed. Lastly, schwannosis is discussed, highlighting its high frequency and potential role when designing therapeutic interventions. We anticipate that a better understanding of the pathological responses in the human will be useful to investigators in their studies on the pathogenesis and therapy of SCI.
Collapse
Affiliation(s)
- Michael D Norenberg
- Department of Pathology, University of Miami School of Medicine, The Miami Project to Cure Paralysis, and the Miami Veterans Affairs Medical Center Miami, Florida 33101, USA.
| | | | | |
Collapse
|
539
|
Ozbas-Gerçeker F, Gorter JA, Redeker S, Ramkema M, van der Valk P, Baayen JC, Ozgüç M, Saygi S, Soylemezoglu F, Akalin N, Troost D, Aronica E. Neurotrophin receptor immunoreactivity in the hippocampus of patients with mesial temporal lobe epilepsy. Neuropathol Appl Neurobiol 2004; 30:651-64. [PMID: 15541005 DOI: 10.1111/j.1365-2990.2004.00582.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Recent evidence supports a critical role of neurotrophins in the regulation of both neuronal survival and synaptic transmission during epileptogenesis. We have examined the immunohistochemical expression of high- (tyrosine kinase receptors, trk) and low-affinity (p75) neurotrophin receptors (NTRs) in the hippocampal specimens from 18 patients with chronic temporal lobe epilepsy [TLE; 14 patients with hippocampal sclerosis (HS) and four with focal lesions (tumours) not involving the hippocampus proper]. Nonepileptic autopsy brains (n = 6) and surgical specimens from tumour patients without epilepsy (n = 3) were used as controls. Immunoreactivity (IR) for the trk receptors (trkA, trkB, trkC) was detected in normal human brain within the pyramidal neurones of hippocampal cornus ammoni (CA) regions and in the dentate gyrus. There were no detectable differences in the neuronal trk IR patterns in the hippocampus between control and TLE cases with HS, except for a decrease in neuronal density in regions where cell death had occurred (CA1, CA3 and CA4). In contrast, a consistent increase in trkA IR was observed in reactive astrocytes in CA1 and dentate gyrus. The low-affinity p75 neurotrophin receptor (p75(NTR)) was expressed in low levels in postnatal normal hippocampus. In contrast, neuronal p75(NTR) IR was detected in 10/14 cases of HS in spared neurones within the CA and hilar regions of the hippocampus. Double labelling revealed that p75(NTR)-positive neurones also contain trk receptor IR. In six cases with prominent glial activation strong p75(NTR) IR was observed in microglial cells within the sclerotic hippocampus. The present results indicate that changes in NTR expression are still detectable in the hippocampus of patients with chronic TLE and involve both glial and neuronal cells. Reactive astrocytes were immunoreactive for trkA, whereas activated microglia cells were reactive for p75(NTR), suggesting different functions for specific NTRs in the development of reactive gliosis. Moreover, the increased expression of p75(NTR) in hippocampal neurones of TLE patients may critically influence the neuronal survival during the epileptogenic process.
Collapse
Affiliation(s)
- F Ozbas-Gerçeker
- Department of Medical Biology, Hacettepe University, Ankara, Turkey
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
540
|
Spach KM, Pedersen LB, Nashold FE, Kayo T, Yandell BS, Prolla TA, Hayes CE. Gene expression analysis suggests that 1,25-dihydroxyvitamin D3reverses experimental autoimmune encephalomyelitis by stimulating inflammatory cell apoptosis. Physiol Genomics 2004; 18:141-51. [PMID: 15138306 DOI: 10.1152/physiolgenomics.00003.2004] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating autoimmune disease of the central nervous system (CNS) that develops in genetically susceptible individuals who are exposed to undefined environmental risk factors. Epidemiological, genetic, and biological evidence suggests that insufficient vitamin D may be an MS risk factor. However, little is known about how vitamin D might be protective in MS. We hypothesized that 1,25-dihydroxyvitamin D3[1,25-(OH)2D3] might regulate gene expression patterns in a manner that would resolve inflammation. To test this hypothesis, experimental autoimmune encephalomyelitis (EAE) was induced in mice, 1,25-(OH)2D3or a placebo was administered, and 6 h later, DNA microarray hybridization was performed with spinal cord RNA to analyze the gene expression patterns. At this time, clinical, histopathological, and biological studies showed that the two groups did not differ in EAE disease, but changes in several 1,25-(OH)2D3-responsive genes indicated that the 1,25-(OH)2D3had reached the CNS. Compared with normal mice, placebo-treated mice with EAE showed increased expression of many immune system genes, confirming the acute inflammation. When 1,25-(OH)2D3was administered, several genes like glial fibrillary acidic protein and eukaryotic initiation factor 2α kinase 4, whose expression increased or decreased with EAE, returned to homeostatic levels. Also, two genes with pro-apoptotic functions, calpain-2 and caspase-8-associated protein, increased significantly. A terminal deoxynucleotidyl transferase-mediated dUTP nicked end labeling study detected increased nuclear fragmentation in the 1,25-(OH)2D3-treated samples, confirming increased apoptosis. Together, these results suggest that sensitization of inflammatory cells to apoptotic signals may be one mechanism by which the 1,25-(OH)2D3resolved EAE.
Collapse
Affiliation(s)
- Karen M Spach
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | | | | | | | | | | |
Collapse
|
541
|
Ma M, Wei P, Wei T, Ransohoff RM, Jakeman LB. Enhanced axonal growth into a spinal cord contusion injury site in a strain of mouse (129X1/SvJ) with a diminished inflammatory response. J Comp Neurol 2004; 474:469-86. [PMID: 15174067 DOI: 10.1002/cne.20149] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
After injury in the adult central nervous system, invading and intrinsic cells contribute to the formation of a lesion site that is refractory to axonal growth. To test the hypothesis that the inflammatory response to trauma dictates the extent of axonal growth after spinal cord injury, the time course of lesion evolution was compared in two mouse strains with contrasting cellular responses to peripheral inflammatory challenge. Adult C57Bl/6 and 129X1/SvJ mice received identical contusion injuries to the mid-thoracic spinal cord and were allowed to recover for 6 hours to 9 weeks. Both strains responded with a rapid, transient increase in chemokine expression, but the magnitude of this early response was slightly reduced in the 129X1/SvJ mice. Morphological indicators of inflammation were similar during the first week postinjury. After 7 days postinjury, however, the cellular responses differed between strains. The C57Bl/6 lesion core was chronically occupied by macrophages, devoid of astrocytes, and contained few axonal profiles. In contrast, as the macrophage density decreased a network of astrocytic processes and axons of central and peripheral origin invaded the center of the lesion site in 129X1Sv/J mice. Growth of axons in the 129X1Sv/J mice was accompanied by increased extravascular laminin in the lesion core and a reduced expression of chondroitin sulfate proteoglycan glycosaminoglycan sidechains in the periphery of the lesion. These results demonstrate that the diminished chronic inflammatory response in 129X1/SvJ mice is associated with enhanced cellular repair and increased axonal growth after spinal cord injury.
Collapse
Affiliation(s)
- Manhong Ma
- Department Physiology and Cell Biology, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
542
|
Buckwalter MS, Wyss-Coray T. Modelling neuroinflammatory phenotypes in vivo. J Neuroinflammation 2004; 1:10. [PMID: 15285805 PMCID: PMC500895 DOI: 10.1186/1742-2094-1-10] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2004] [Accepted: 07/01/2004] [Indexed: 11/25/2022] Open
Abstract
Inflammation of the central nervous system is an important but poorly understood part of neurological disease. After acute brain injury or infection there is a complex inflammatory response that involves activation of microglia and astrocytes and increased production of cytokines, chemokines, acute phase proteins, and complement factors. Antibodies and T lymphocytes may be involved in the response as well. In neurodegenerative disease, where injury is more subtle but consistent, the inflammatory response is continuous. The purpose of this prolonged response is unclear, but it is likely that some of its components are beneficial and others are harmful. Animal models of neurological disease can be used to dissect the specific role of individual mediators of the inflammatory response and assess their potential benefit. To illustrate this approach, we discuss how mutant mice expressing different levels of the cytokine transforming growth factor beta-1 (TGF-beta1), a major modulator of inflammation, produce important neuroinflammatory phenotypes. We then demonstrate how crosses of TGF-beta1 mutant mice with mouse models of Alzheimer's disease (AD) produced important new information on the role of inflammation in AD and on the expression of different neuropathological phenotypes that characterize this disease.
Collapse
Affiliation(s)
- Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, 94305-5235, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, 94305-5235, USA
- Geriatric Research and Education and Clinical Center, Palo Alto Veteran's Medical Center, Palo Alto, California, 94304, USA
| |
Collapse
|
543
|
Tanga FY, Raghavendra V, DeLeo JA. Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain. Neurochem Int 2004; 45:397-407. [PMID: 15145554 DOI: 10.1016/j.neuint.2003.06.002] [Citation(s) in RCA: 290] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2003] [Revised: 06/11/2003] [Accepted: 06/11/2003] [Indexed: 12/01/2022]
Abstract
Activated spinal glial cells have been strongly implicated in the development and maintenance of persistent pain states following a variety of stimuli including traumatic nerve injury. The present study was conducted to characterize the time course of surface markers indicative of microglial and astrocytic activation at the transcriptional level following an L5 nerve transection that results in behavioral hypersensitivity. Male Sprague-Dawley rats were divided into a normal group, a sham surgery group with an L5 spinal nerve exposure and an L5 spinal nerve transected group. Mechanical allodynia (heightened response to a non-noxious stimulus) of the ipsilateral hind paw was assessed throughout the study. Spinal lumbar mRNA levels of glial fibrillary acidic protein (GFAP), integrin alpha M (ITGAM), toll-like receptor 4 (TLR4) and cluster determinant 14 (CD14) were assayed using real-time reverse transcription polymerase chain reaction (RT-PCR) at 4 h, 1, 4, 7, 14 and 28 days post surgery. The spinal lumbar mRNA expression of ITGAM, TLR4, and CD14 was upregulated at 4 h post surgery, CD14 peaked 4 days after spinal nerve transection while ITGAM and TLR4 continued to increase until day 14 and returned to almost normal levels by postoperative day 28. In contrast, spinal GFAP mRNA did not significantly increase until postoperative day 4 and then continued to increase over the duration of the study. Our optimized real-time RT-PCR method was highly sensitive, specific and reproducible at a wide dynamic range. This study demonstrates that peripheral nerve injury induces an early spinal microglial activation that precedes astrocytic activation using mRNA for surface marker expression; the delayed but sustained expression of mRNA coding for GFAP implicates astrocytes in the maintenance phase of persistent pain states. In summary, these data demonstrate a distinct spinal glial response following nerve injury using real-time RT-PCR.
Collapse
Affiliation(s)
- F Y Tanga
- Department of Anesthesiology, Dartmouth-Hitchcock Medical Center, HB 7125, One Medical Center Drive, Lebanon, NH 03756, USA.
| | | | | |
Collapse
|
544
|
Mori T, Tateishi N, Kagamiishi Y, Shimoda T, Satoh S, Ono S, Katsube N, Asano T. Attenuation of a delayed increase in the extracellular glutamate level in the peri-infarct area following focal cerebral ischemia by a novel agent ONO-2506. Neurochem Int 2004; 45:381-7. [PMID: 15145552 DOI: 10.1016/j.neuint.2003.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2003] [Revised: 06/13/2003] [Accepted: 06/14/2003] [Indexed: 11/20/2022]
Abstract
A novel agent, ONO-2506 [(R)-(-)-2-propyloctanoic acid, ONO Pharmaceutical Co. Ltd.] was previously shown to mitigate delayed infarct expansion through inhibition of the enhanced production of S-100beta, while inducing a prompt symptomatic improvement that attained a significant level as early as 24h after drug administration. To elucidate the mechanism underlying the prompt symptomatic improvement, the present study aimed to examine whether ONO-2506 modulates the level of extracellular glutamate ([Glu]e) in the rat subjected to transient middle cerebral artery occlusion (tMCAO). In this model, it had been shown that ONO-2506 reduces the infarct volume, improves the neurological deficits, and enhances the mRNA expression of glial glutamate transporters (GLT-1 and GLAST). The [Glu]e levels in the ischemic cortices were continuously measured using intracerebral microdialysis. The alterations in the [Glu]e levels in the sham-operated and tMCAO-operated groups with or without drug administration were compared. In the tMCAO groups, the [Glu]e level increased during tMCAO to a similar extent, returned to normal on reperfusion, and increased again around 5h. In the saline-treated group, however, the [Glu]e level further increased from 15 h on to reach about 280% of the normal level at 24h. This secondary increase in the [Glu]e level in the late phase of reperfusion was prevented by ONO-2506. The intracerebral infusion of glutamate transporter inhibitor, l-trans-pyrrolidine-2,4-dicarboxylic acid, at 24h after tMCAO induced an increase in the [Glu]e level, which was marked in both the sham-operated and ONO-2506-treated groups, but much less pronounced in the saline-treated group. The above results suggest that functional modulation of activated astrocytes by pharmacological agents like ONO-2506 may inhibit the secondary rise of [Glu]e level in the late phase of reperfusion, leading to amelioration of delayed infarct expansion and neurological deficits.
Collapse
Affiliation(s)
- Takashi Mori
- Institute of Laboratory Animal Science, Saitama Medical Center/School, 1981 Kamoda, Kawagoe, Saitama 350-8550, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
545
|
Lundkvist A, Reichenbach A, Betsholtz C, Carmeliet P, Wolburg H, Pekny M. Under stress, the absence of intermediate filaments from Müller cells in the retina has structural and functional consequences. J Cell Sci 2004; 117:3481-8. [PMID: 15226376 DOI: 10.1242/jcs.01221] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In epithelial and muscle cells, intermediate filaments (IFs) are important for resistance to mechanical stress. The aim of this study was to elucidate whether IFs are also important for providing resistance to mechanical stress in the Müller cells of the retina and whether this has any pathophysiological consequences. We used mice deficient in IF proteins glial fibrillary acidic protein and/or vimentin (GFAP(-/-), Vim(-/-) and GFAP(-/-) Vim(-/-)), and stress on the retina was applied by excision of the eyes immediately post mortem (compared with in situ fixation) or by inducing a neovascular response to oxygen-induced retinopathy (OIR). The structure of unchallenged retinas was normal, but mechanical stress caused local separation of the inner limiting membrane (ILM) and adjacent tissue from the rest of the retina in GFAP(-/-) Vim(-/-) mice and, to a lesser extent, in Vim(-/-) mice. This detachment occurred within the endfeet of Müller cells, structures normally rich in IFs but IF-free in GFAP(-/-) Vim(-/-) mice. Hypoxia-induced neovascularization was comparable in all groups of mice with respect to the retinal surface area occupied by new vessels. However, the vessels traversed the ILM and penetrated the vitreous body less frequently than in wild-type retinas (31-55% in Vim(-/-), 66-79% in GFAP(-/-) Vim(-/-)). We conclude that IFs are important for maintaining the mechanical integrity of Müller-cell endfeet and the inner retinal layers under a mechanical challenge. Furthermore, the absence of IFs in Müller cells leads to an abnormal response of the vascular system to ischemia, specifically decreased ability of newly formed blood vessels to traverse the ILM.
Collapse
Affiliation(s)
- Andrea Lundkvist
- Department of Medical Biochemistry, The Sahlgrenska Academy at Göteborg University, Medicinaregatan 9C, SE-41390 Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
546
|
Marien MR, Colpaert FC, Rosenquist AC. Noradrenergic mechanisms in neurodegenerative diseases: a theory. ACTA ACUST UNITED AC 2004; 45:38-78. [PMID: 15063099 DOI: 10.1016/j.brainresrev.2004.02.002] [Citation(s) in RCA: 316] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2004] [Indexed: 11/26/2022]
Abstract
A deficiency in the noradrenergic system of the brain, originating largely from cells in the locus coeruleus (LC), is theorized to play a critical role in the progression of a family of neurodegenerative disorders that includes Parkinson's disease (PD) and Alzheimer's disease (AD). Consideration is given here to evidence that several neurodegenerative diseases and syndromes share common elements, including profound LC cell loss, and may in fact be different manifestations of a common pathophysiological process. Findings in animal models of PD indicate that the modification of LC-noradrenergic activity alters electrophysiological, neurochemical and behavioral indices of neurotransmission in the nigrostriatal dopaminergic system, and influences the response of this system to experimental lesions. In models related to AD, noradrenergic mechanisms appear to play important roles in modulating the activity of the basalocortical cholinergic system and its response to injury, and to modify cognitive functions including memory and attention. Mechanisms by which noradrenaline may protect or promote recovery from neural damage are reviewed, including effects on neuroplasticity, neurotrophic factors, neurogenesis, inflammation, cellular energy metabolism and excitotoxicity, and oxidative stress. Based on evidence for facilitatory effects on transmitter release, motor function, memory, neuroprotection and recovery of function after brain injury, a rationale for the potential of noradrenergic-based approaches, specifically alpha2-adrenoceptor antagonists, in the treatment of central neurodegenerative diseases is presented.
Collapse
Affiliation(s)
- Marc R Marien
- Centre de Recherche Pierre Fabre, Neurobiology I, 17 Avenue Jean Moulin, 81106 Castres Cedex, France.
| | | | | |
Collapse
|
547
|
Moxon KA, Kalkhoran NM, Markert M, Sambito MA, McKenzie JL, Webster JT. Nanostructured Surface Modification of Ceramic-Based Microelectrodes to Enhance Biocompatibility for a Direct Brain-Machine Interface. IEEE Trans Biomed Eng 2004; 51:881-9. [PMID: 15188854 DOI: 10.1109/tbme.2004.827465] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Many different types of microelectrodes have been developed for use as a direct Brain-Machine Interface (BMI) to chronically recording single neuron action potentials from ensembles of neurons. Unfortunately, the recordings from these microelectrode devices are not consistent and often last for only a few weeks. For most microelectrode types, the loss of these recordings is not due to failure of the electrodes but most likely due to damage to surrounding tissue that results in the formation of nonconductive glial-scar. Since the extracellular matrix consists of nanostructured microtubules, we have postulated that neurons may prefer a more complex surface structure than the smooth surface typical of thin-film microelectrodes. We, therefore, investigated the suitability of a nano-porous silicon surface layer to increase the biocompatibility of our thin film ceramic-insulated multisite electrodes. In-vitro testing demonstrated, for the first time, decreased adhesion of astrocytes and increased extension of neurites from pheochromocytoma cells on porous silicon surfaces compared to smooth silicon sufaces. Moreover, nano-porous surfaces were more biocompatible than macroporous surfaces. Collectively, these results support our hypothesis that nano-porous silicon may be an ideal material to improve biocompatibility of chronically implanted microelectrodes. We next developed a method to apply nano-porous surfaces to ceramic insulated, thin-film, microelectrodes and tested them in vivo. Chronic testing demonstrated that the nano-porous surface modification did not alter the electrical properties of the recording sites and did not interfere with proper functioning of the microelectrodes in vivo.
Collapse
Affiliation(s)
- Karen A Moxon
- School of Biomedical Engineering, Drexel University, 3141 Chestnut St, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
548
|
Mori T, Town T, Kobayashi M, Tan J, Fujita SC, Asano T. Augmented delayed infarct expansion and reactive astrocytosis after permanent focal ischemia in apolipoprotein E4 knock-in mice. J Cereb Blood Flow Metab 2004; 24:646-56. [PMID: 15181372 DOI: 10.1097/01.wcb.0000120787.53851.a4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Using homozygous human apolipoprotein E2 (apoE2) (2/2)-, apoE3 (3/3)-, or apoE4 (4/4)-knock-in (KI) mice, we aimed to examine whether an apoE isoform-specific exacerbation of delayed infarct expansion occurs after permanent middle cerebral artery occlusion (pMCAO). Compared with 2/2- or 3/3-KI mice, 4/4-KI mice exhibited significantly larger infarct volumes and worse neurologic deficits after pMCAO, with no significant differences between the latter two groups. Infarct volume in 4/4-KI mice was significantly increased from 1 to 5 days after pMCAO, whereas that in 2/2- or 3/3-KI mice was not significantly altered. DNA fragmentation in the peri-infarct area as detected by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphatenick end-labeling was increased to a similar degree in all of the KI mice by 5 days after pMCAO, with no significant differences among the mouse groups. At every time-point examined, human apoE was most markedly expressed in the peri-infarct area, with similar immunoreactivity among the three lines of KI mice. The glial fibrillary acidic protein immunoreactive burden in the peri-infarct area was progressively increased through 7 days in 4/4-KI mice, but not in 2/2- or 3/3-KI mice. Taken together, these data show that the apoE4 isoform acts to aggravate delayed infarct expansion and peri-infarct reactive astrocytosis during the subacute phase of pMCAO in genetically engineered apoE-KI mice.
Collapse
Affiliation(s)
- Takashi Mori
- Institute of Laboratory Animal Science, Saitama Medical Center/School, Kawagoe, Saitama, Japan.
| | | | | | | | | | | |
Collapse
|
549
|
Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci 2004; 24:2143-55. [PMID: 14999065 PMCID: PMC6730429 DOI: 10.1523/jneurosci.3547-03.2004] [Citation(s) in RCA: 1184] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Reactive astrocytes are prominent in the cellular response to spinal cord injury (SCI), but their roles are not well understood. We used a transgenic mouse model to study the consequences of selective and conditional ablation of reactive astrocytes after stab or crush SCI. Mice expressing a glial fibrillary acid protein-herpes simplex virus-thymidine kinase transgene were given mild or moderate SCI and treated with the antiviral agent ganciclovir (GCV) to ablate dividing, reactive, transgene-expressing astrocytes in the immediate vicinity of the SCI. Small stab injuries in control mice caused little tissue disruption, little demyelination, no obvious neuronal death, and mild, reversible functional impairments. Equivalent small stab injuries in transgenic mice given GCV to ablate reactive astrocytes caused failure of blood-brain barrier repair, leukocyte infiltration, local tissue disruption, severe demyelination, neuronal and oligodendrocyte death, and pronounced motor deficits. Moderate crush injuries in control mice caused focal tissue disruption and cellular degeneration, with moderate, primarily reversible motor impairments. Equivalent moderate crush injuries combined with ablation of reactive astrocytes caused widespread tissue disruption, pronounced cellular degeneration, and failure of wound contraction, with severe persisting motor deficits. These findings show that reactive astrocytes provide essential activities that protect tissue and preserve function after mild or moderate SCI. In nontransgenic animals, crush or contusion SCIs routinely exhibit regions of degenerated tissue that are devoid of astrocytes. Our findings suggest that identifying ways to preserve reactive astrocytes, to augment their protective functions, or both, may lead to novel approaches to reducing secondary tissue degeneration and improving functional outcome after SCI.
Collapse
Affiliation(s)
- Jill R Faulkner
- Department of Neurobiology, University of California, Los Angeles, California 90095-1763, USA
| | | | | | | | | | | |
Collapse
|
550
|
Wu K, Meyers CA, Bennett JA, King MA, Meyer EM, Hughes JA. Polyethylenimine-mediated NGF gene delivery protects transected septal cholinergic neurons. Brain Res 2004; 1008:284-7. [PMID: 15145767 DOI: 10.1016/j.brainres.2004.02.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2004] [Indexed: 01/18/2023]
Abstract
Polyethylenimine (PEI) is an effective vehicle for in vivo gene delivery in many tissues including brain. PEI mediates transgene expression in brain neurons and glia. To investigate whether PEI-mediated nerve growth factor (NGF) gene transfer protected axotomized septal cholinergic neurons, we injected linear PEI (in vivo jetPEI, Qbiogene) complexed with a plasmid encoding for mouse NGF (PEI/pNGF-W) into the rat septum. PEI complexed with a plasmid encoding for green fluorescent protein (PEI/pGFP) was used as the control. PEI-mediated gene expression was predominantly neuronal. Fimbria-fornix transections (FFTs), conducted 1 day after rats were injected with control vector, resulted in a 70% loss of septal cholinergic neurons. In contrast, PEI/pNGF-W injection prior to FFTs attenuated the loss of septal cholinergic neurons. This is the first study, to our knowledge, that shows the neuroprotective effects induced by PEI-mediated trophic factor gene transfer in brain.
Collapse
Affiliation(s)
- Ke Wu
- Department of Pharmacology and Therapeutics, ARB5-148, 1600 SW Archer Rd, Gainesville, FL 32610, USA.
| | | | | | | | | | | |
Collapse
|