401
|
Santos-Silva A, Fairless R, Frame MC, Montague P, Smith GM, Toft A, Riddell JS, Barnett SC. FGF/heparin differentially regulates Schwann cell and olfactory ensheathing cell interactions with astrocytes: a role in astrocytosis. J Neurosci 2007; 27:7154-67. [PMID: 17611269 PMCID: PMC6794582 DOI: 10.1523/jneurosci.1184-07.2007] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
After injury, the CNS undergoes an astrocyte stress response characterized by reactive astrocytosis/proliferation, boundary formation, and increased glial fibrillary acidic protein (GFAP) and chondroitin sulfate proteoglycan (CSPG) expression. Previously, we showed that in vitro astrocytes exhibit this stress response when in contact with Schwann cells but not olfactory ensheathing cells (OECs). In this study, we confirm this finding in vivo by demonstrating that astrocytes mingle with OECs but not Schwann cells after injection into normal spinal cord. We show that Schwann cell-conditioned media (SCM) induces proliferation in monocultures of astrocytes and increases CSPG expression in a fibroblast growth factor receptor 1 (FGFR1)-independent manner. However, SCM added to OEC/astrocyte cocultures induces reactive astrocytosis and boundary formation, which, although sensitive to FGFR1 inhibition, was not induced by FGF2 alone. Addition of heparin to OEC/astrocyte cultures induces boundary formation, whereas heparinase or chlorate treatment of Schwann cell/astrocyte cultures reduces it, suggesting that heparan sulfate proteoglycans (HSPGs) are modulating this activity. In vivo, FGF2 and FGFR1 immunoreactivity was increased over grafted OECs and Schwann cells compared with the surrounding tissue, and HSPG immunoreactivity is increased over reactive astrocytes bordering the Schwann cell graft. These data suggest that components of the astrocyte stress response, including boundary formation, astrocyte hypertrophy, and GFAP expression, are mediated by an FGF family member, whereas proliferation and CSPG expression are not. Furthermore, after cell transplantation, HSPGs may be important for mediating the stress response in astrocytes via FGF2. Identification of factors secreted by Schwann cells that induce this negative response in astrocytes would further our ability to manipulate the inhibitory environment induced after injury to promote regeneration.
Collapse
Affiliation(s)
- Alessandra Santos-Silva
- Division of Clinical Neuroscience, Beatson Institute, University of Glasgow, Glasgow G61 1BD, United Kingdom
| | - Richard Fairless
- Division of Clinical Neuroscience, Beatson Institute, University of Glasgow, Glasgow G61 1BD, United Kingdom
| | - Margaret C. Frame
- Beatson Institute, Cancer Research UK, Glasgow G61 1BD, United Kingdom
| | - Paul Montague
- Division of Clinical Neuroscience, Beatson Institute, University of Glasgow, Glasgow G61 1BD, United Kingdom
| | - George M. Smith
- Department of Physiology and Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40509
| | - Andrew Toft
- Division of Neuroscience and Biomedical Systems, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, and
| | - John S. Riddell
- Division of Neuroscience and Biomedical Systems, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom, and
| | - Susan C. Barnett
- Division of Clinical Neuroscience, Beatson Institute, University of Glasgow, Glasgow G61 1BD, United Kingdom
| |
Collapse
|
402
|
Monnet-Tschudi F, Zurich MG, Honegger P. Neurotoxicant-induced inflammatory response in three-dimensional brain cell cultures. Hum Exp Toxicol 2007; 26:339-46. [PMID: 17615115 DOI: 10.1177/0960327107074589] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Brain inflammatory response is triggered by the activation of microglial cells and astrocytes in response to various types of CNS injury, including neurotoxic insults. Its outcome is determined by cellular interactions, inflammatory mediators, as well as trophic and/or cytotoxic signals, and depends on many additional factors such as the intensity and duration of the insult, the extent of both the primary neuronal damage and glial reactivity and the developmental stage of the brain. Depending on particular circumstances, the brain inflammatory response can promote neuroprotection, regeneration or neurodegeneration. Glial reactivity, regarded as the central phenomenon of brain inflammation, has also been used as an early marker of neurotoxicity. To study the mechanisms underlying the glial reactivity, serum-free aggregating brain cell cultures were used as an in vitro model to test the effects of conventional neurotoxicants such as organophosphate pesticides, heavy metals, excitotoxins and mycotoxins. This approach was found to be relevant and justified by the complex cell-cell interactions involved in the brain inflammatory response, the variability of the glial reactions and the multitude of mediators involved. All these variables need to be considered for the elucidation of the specific cellular and molecular reactions and their consequences caused by a given chemical insult.
Collapse
Affiliation(s)
- F Monnet-Tschudi
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.
| | | | | |
Collapse
|
403
|
Favero CB, Mandell JW. A pharmacological activator of AMP-activated protein kinase (AMPK) induces astrocyte stellation. Brain Res 2007; 1168:1-10. [PMID: 17706943 PMCID: PMC2000700 DOI: 10.1016/j.brainres.2007.06.087] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 06/19/2007] [Accepted: 06/20/2007] [Indexed: 11/20/2022]
Abstract
AMP-activated protein kinase (AMPK) represents a key energy-sensing molecule in many cell types. Because astrocytes are key mediators of metabolic signaling in the brain, we have initiated studies on the expression and activation of AMPK in these cells. Treatment of cultured rat cortical astrocytes with a pharmacological AMPK activator, AICA-riboside (AICAR) resulted in a time- and concentration-dependent increase in phosphorylation of AMPK and acetyl-CoA carboxylase (ACC), a direct substrate. AICAR treatment also induced a transition from epithelioid to stellate morphology in a time- and concentration-dependent manner. As stellation is indicative of actin cytoskeletal reorganization, the formation of stress fibers and focal adhesions in response to AICAR was assessed. AICAR-induced stellation correlated with F-actin disassembly and focal adhesion dispersal. Furthermore, transient transfection of an activated RhoA construct prevented AICAR-induced stellation, indicating a mechanism upstream of RhoA. Use of pharmacological inhibitor compound C prevented AICAR-induced stellation demonstrating necessity of AMPK activity for the response. Our findings suggest that AMPK mediates morphological alterations of astrocytes in response to energy depletion.
Collapse
Affiliation(s)
- Carlita B Favero
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
404
|
Lee TS, Mane S, Eid T, Zhao H, Lin A, Guan Z, Kim JH, Schweitzer J, King-Stevens D, Weber P, Spencer SS, Spencer DD, de Lanerolle NC. Gene expression in temporal lobe epilepsy is consistent with increased release of glutamate by astrocytes. Mol Med 2007; 13:1-13. [PMID: 17515952 PMCID: PMC1869627 DOI: 10.2119/2006-00079.lee] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 12/07/2006] [Indexed: 11/06/2022] Open
Abstract
Patients with temporal lobe epilepsy (TLE) often have a shrunken hippocampus that is known to be the location in which seizures originate. The role of the sclerotic hippocampus in the causation and maintenance of seizures in temporal lobe epilepsy (TLE) has remained incompletely understood despite extensive neuropathological investigations of this substrate. To gain new insights and develop new testable hypotheses on the role of sclerosis in the pathophysiology of TLE, the differential gene expression profile was studied. To this end, DNA microarray analysis was used to compare gene expression profiles in sclerotic and non-sclerotic hippocampi surgically removed from TLE patients. Sclerotic hippocampi had transcriptional signatures that were different from non-sclerotic hippocampi. The differentially expressed gene set in sclerotic hippocampi revealed changes in several molecular signaling pathways, which included the increased expression of genes associated with astrocyte structure (glial fibrillary acidic protein, ezrin-moesin-radixin, palladin), calcium regulation (S100 calcium binding protein beta, chemokine (C-X-C motif) receptor 4) and blood-brain barrier function (Aquaaporin 4, Chemokine (C-C- motif) ligand 2, Chemokine (C-C- motif) ligand 3, Plectin 1, intermediate filament binding protein 55kDa) and inflammatory responses. Immunohistochemical localization studies show that there is altered distribution of the gene-associated proteins in astrocytes from sclerotic foci compared with non-sclerotic foci. It is hypothesized that the astrocytes in sclerotic tissue have activated molecular pathways that could lead to enhanced release of glutamate by these cells. Such glutamate release may excite surrounding neurons and elicit seizure activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhong Guan
- Epidemiology and Public Health
- Department of Mathematical Sciences, Indiana University, South Bend, Indiana
| | | | - Jeffrey Schweitzer
- Department of Neurosurgery, Kaiser Permanente Medical Center, Los Angeles, California
| | | | | | | | | | - Nihal C de Lanerolle
- Neurosurgery
- Address correspondence and reprint requests to Nihal C. de Lanerolle, Department of Neurosurgery FMB414, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8082. Phone: 203-785-3258; Fax: 203-737-2159; E-mail:
| |
Collapse
|
405
|
Widestrand A, Faijerson J, Wilhelmsson U, Smith PLP, Li L, Sihlbom C, Eriksson PS, Pekny M. Increased neurogenesis and astrogenesis from neural progenitor cells grafted in the hippocampus of GFAP-/- Vim-/- mice. Stem Cells 2007; 25:2619-27. [PMID: 17628017 DOI: 10.1634/stemcells.2007-0122] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
After neurotrauma, ischemia, or neurodegenerative disease, astrocytes upregulate their expression of the intermediate filament proteins glial fibrillary acidic protein (GFAP), vimentin (Vim), and nestin. This response, reactive gliosis, is attenuated in GFAP(-/-)Vim(-/-) mice, resulting in the promotion of synaptic regeneration after neurotrauma and improved integration of retinal grafts. Here we assessed whether GFAP(-/-)Vim(-/-) astrocytes affect the differentiation of neural progenitor cells. In coculture with GFAP(-/-)Vim(-/-) astrocytes, neural progenitor cells increased neurogenesis by 65% and astrogenesis by 124%. At 35 days after transplantation of neural progenitor cells into the hippocampus, adult GFAP(-/-)Vim(-/-) mice had more transplant-derived neurons and astrocytes than wild-type controls, as well as increased branching of neurite-like processes on transplanted cells. Wnt3 immunoreactivity was readily detected in hippocampal astrocytes in wild-type but not in GFAP(-/-)Vim(-/-) mice. These findings suggest that GFAP(-/-)Vim(-/-) astrocytes allow more neural progenitor cell-derived neurons and astrocytes to survive weeks after transplantation. Thus, reactive gliosis may adversely affect the integration of transplanted neural progenitor cells in the brain. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Asa Widestrand
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Göteborg University, Medicinaregatan 9A, SE-413 90 Göteborg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
406
|
Blanco S, Castro L, Hernández R, Del Moral ML, Pedrosa JA, Martínez-Lara E, Siles E, Peinado MA. Age modulates the nitric oxide system response in the ischemic cerebellum. Brain Res 2007; 1157:66-73. [PMID: 17544383 DOI: 10.1016/j.brainres.2007.01.141] [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: 10/23/2006] [Revised: 01/19/2007] [Accepted: 01/23/2007] [Indexed: 10/23/2022]
Abstract
To determine whether age influences the nitric oxide system response to ischemia in the cerebellum, we have analyzed the levels of nitrogen oxides (NOx) and the expression of the different nitric oxide synthase isoforms (NOS) in mature adult (4-5 months old) and aged rats (24-27 months old) subjected to a transient global ischemia/reperfusion (I/R) model. We also analyzed the nitrated proteins and the glial fibrillary acidic protein (GFAP) expression. NOx concentration in adult rats, which more than doubled the values found in the aged rats, decreased after the ischemia and reperfusion. However, in the aged animals, these NOx levels did not significantly change after I/R. Constitutive isoforms were first down-regulated in the ischemic period, in both adult and aged animals. However, after 6 h of reperfusion, these isoforms were up-regulated, but only in aged rats. After I/R, iNOS was up-regulated in adults but down-regulated in the aged rats. Hence, after an episode of transient global ischemia and reperfusion, the aged cerebellum maintains a balanced NO production, silencing the iNOS isoform and inducing a weak expression of nNOS and eNOS; this allows NO physiological functions while avoiding possible undesirable effects such as the nitrative damage or astrocyte activation.
Collapse
Affiliation(s)
- Santos Blanco
- Department of Experimental Biology, University of Jaén, Paraje Las Lagunillas s/n, 23071, Jaén, Spain
| | | | | | | | | | | | | | | |
Collapse
|
407
|
Suzuki A, Yamaguchi Y, Hoshi T, Hayashi A, Baba H. Differential inductions of small heat shock protein 27 and 1-Cys peroxiredoxin in reactive astrocytes in sulfatide-deficient mouse spinal cord. J Neurosci Res 2007; 85:1921-32. [PMID: 17510974 DOI: 10.1002/jnr.21338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In myelinated fibers, various interactions among axons, oligodendrocytes, and astrocytes are present, particularly around the node of Ranvier. In the present study, we examined the protein composition of cerebroside sulfotransferase knockout (CST KO) mouse spinal cord by two-dimensional gel electrophoresis to examine the molecular changes resulting from the disruption of paranodal junctions in addition to the sulfatide-deficient condition. Interestingly, heat shock protein 27 (Hsp27) and 1-cys peroxiredoxin (1-Cys Prx) were both elevated in CST KO mice. Hsp27 was increased specifically in reactive astrocytes in the white matter, and the elevation was well correlated to the progression of neurologic symptoms. In contrast, 1-Cys Prx was elevated both in white and gray matter astrocytes in CST KO mice. These results suggest that astrocytes do not always respond stereotypically, as they display differences in their activation in these two regions. To determine whether these changes are specific to the sulfatide-deficient condition, spinal cords from CST KO mice and the hypomyelinating mutant shiverer mice were compared. The same distribution patterns of Hsp27 and 1-Cys Prx were found in reactive astrocytes in both CST KO and shiverer mice, suggesting that paranodal disruption with progressive nodal changes may underlie the similar reaction of white matter astrocytes. In contrast, CST KO and shiverer mice showed distinctly different localization patterns of connexin 43 and connexin 47, suggesting that intercellular communication between astrocytes and oligodendrocytes was different in these mutants. These results suggest that astrocytes may respond differentially to individual white matter abnormalities and may modulate specific axonal functions.
Collapse
Affiliation(s)
- Ayaka Suzuki
- Department of Molecular Neurobiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Japan
| | | | | | | | | |
Collapse
|
408
|
Chakrabarty K, Serchov T, Mann SA, Dietzel ID, Heumann R. Enhancement of dopaminergic properties and protection mediated by neuronal activation of Ras in mouse ventral mesencephalic neurones. Eur J Neurosci 2007; 25:1971-81. [PMID: 17439485 DOI: 10.1111/j.1460-9568.2007.05457.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The poor differentiation and survival of dopaminergic neurones are practical constraints in their therapeutic applications. Here we explored the role of neuronally activated Ras in ventral mesencephalon-derived neurospheres generated from synRas mouse embryos. The expression of Val12 Ha-Ras transgene and enhanced Ras activity was evident after differentiation of the neurospheres with a corresponding activating phosphorylation of mitogen-activated protein kinase. Phosphorylation of Akt/PKB, the target kinase of phosphoinositide 3-kinase, along with phosphorylation of Bad and CREB were enhanced in synRas-derived differentiated neurosphere cultures. Furthermore, increased Nurr1 expression was associated with elevated numbers of dopaminergic neurones in synRas-derived cultures compared with the wild-type. Correspondingly, tyrosine hydroxylase promoter assays revealed enhanced transcriptional activation of the promoter in synRas-derived cultures. synRas-derived dopaminergic neurones were greatly resistant to degeneration induced by various noxious stimuli. Consistently, the transgenic expression of activated Ras attenuated the adverse 6-hydroxydopamine effects on dopaminergic neurones. Dopaminergic neurones derived from both wild-type and synRas cultures expressed voltage-gated potassium and sodium currents, fired action potentials and exhibited electrical network activity. Thus, expression of the transgene promotes survival and enhances differentiation towards a dopaminergic cell fate without altering their basic electrical properties. Our results suggest that intracellular cell therapy mimicking trophic signalling may offer potential benefit in models of human disease associated with dopamine neurone dysfunction.
Collapse
Affiliation(s)
- Koushik Chakrabarty
- Department of Molecular Neurobiochemistry, NC7/174, Ruhr University, 44780 Bochum, Germany
| | | | | | | | | |
Collapse
|
409
|
Klaver CL, Caplan MR. Bioactive surface for neural electrodes: decreasing astrocyte proliferation via transforming growth factor-beta1. J Biomed Mater Res A 2007; 81:1011-6. [PMID: 17265435 DOI: 10.1002/jbm.a.31153] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Implantation of deep-brain recording devices is a traumatic event, which inevitably elicits reactive gliosis. The ensuing glial scar encapsulating the implanted device impedes the long-term functional recording capability of the microelectrode. In this work, a bioactive surface is prepared by conjugation of transforming growth factor-beta one (TGF-beta1) and laminin to dextran, which is in turn conjugated to a biomaterial substrate. Poly-L-lysine coated surfaces are treated with oxidized dextran, and the dextran is re-oxidized with sodium metaperiodate to generate hemiacetal structures to which TGF-beta1 and laminin are covalently bound. Covalent conjugation of the ligand is confirmed by enzyme-linked immunosorbent assay. A primary cell line of astrocytes is incubated on a surface conjugated with laminin and TGF-beta1 and a surface only conjugated with laminin. Proliferation on the laminin plus TGF-beta1 surface is 57% less (p < 0.002) than the control surface (laminin alone). The results demonstrate that conjugated TGF-beta1 retains its efficacy toward astrocyte proliferation and represents a potential strategy for reducing glial scar formation in vivo.
Collapse
Affiliation(s)
- Christopher L Klaver
- Department of Chemical and Materials Engineering, Arizona State University, P.O. Box 876006, Tempe, Arizona 85287-6006, USA
| | | |
Collapse
|
410
|
Lim JH, Gibbons HM, O'Carroll SJ, Narayan PJ, Faull RLM, Dragunow M. Extracellular signal-regulated kinase involvement in human astrocyte migration. Brain Res 2007; 1164:1-13. [PMID: 17644078 DOI: 10.1016/j.brainres.2007.06.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 06/07/2007] [Accepted: 06/13/2007] [Indexed: 01/09/2023]
Abstract
Glial scar formation occurs after virtually any injury to the brain. The migration of astrocytes into regions of brain injury underlies the formation of the glial scar. The exact role of the glial scar has yet to be elucidated, although it is likely to impair brain recovery. Understanding astrocyte migration is fundamental to understanding the formation of the glial scar. We have used human astrocytes (NT2A cells), derived from human NT2/D1 precursor cells to study astrocyte migration using an in vitro scratch wound model. Time-lapse microscopy and bromodeoxyuridine labeling revealed that the astrocytes migrated rather than proliferated across the scratch. Time course immunocytochemical studies showed that scratching human astrocytes induced the activation (phosphorylation) of ERK 1/2 at 10 min after scratch. The MEK 1/2 inhibitor U0126 inhibited both the ERK 1/2 phosphorylation and the migration of the astrocytes across the wound after scratch. Thus, the migration of human astrocytes after injury is partly initiated by activation of the MEK-ERK signalling pathway.
Collapse
Affiliation(s)
- Joanne H Lim
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | | | | | | | | | | |
Collapse
|
411
|
Galoyan AA, Sarkissian DS, Chavushyan VA, Meliksetyan IB, Avakyan ZE, Sulkhanyan RM, Poghosyan MV, Avetisyan ZA. Studies of the protective effect of the hypothalamic peptide PRP-3 on spinal cord neurons at different periods after lateral hemisection. NEUROCHEM J+ 2007. [DOI: 10.1134/s1819712407020092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
412
|
Brillaud E, Piotrowski A, de Seze R. Effect of an acute 900MHz GSM exposure on glia in the rat brain: a time-dependent study. Toxicology 2007; 238:23-33. [PMID: 17624651 DOI: 10.1016/j.tox.2007.05.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 05/10/2007] [Accepted: 05/14/2007] [Indexed: 11/25/2022]
Abstract
Because of the increasing use of mobile phones, the possible risks of radio frequency electromagnetic fields adverse effects on the human brain has to be evaluated. In this work we measured GFAP expression, to evaluate glial evolution 2, 3, 6 and 10 days after a single GSM exposure (15min, brain averaged SAR=6W/kg, 900MHz signal) in the rat brain. A statistically significant increase of GFAP stained surface area was observed 2 days after exposure in the frontal cortex and the caudate putamen. A smaller statistically significant increase was noted 3 days after exposure in the same areas and in the cerebellum cortex. Our results confirm the Mausset-Bonnefont et al. study [Mausset-Bonnefont, A.L., Hirbec, H., Bonnefont, X., Privat, A., Vignon, J., de Seze, R., 2004. Acute exposure to GSM 900MHz electromagnetic fields induces glial reactivity and biochemical modifications in the rat brain. Neurobiol. Dis. 17, 445-454], showing the existence of glial reactivity after a 15min GSM acute exposure at a brain averaged SAR of 6W/kg. We conclude to a temporary effect, probably due to a hypertrophy of glial cells, with a temporal and a spatial modulation of the effect. Whether this effect could be harmful remains to be studied.
Collapse
Affiliation(s)
- Elsa Brillaud
- INERIS, Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550 Verneuil-en-Halatte, France.
| | | | | |
Collapse
|
413
|
Kimura N, Ishii Y, Suzaki S, Negishi T, Kyuwa S, Yoshikawa Y. Abeta upregulates and colocalizes with LGI3 in cultured rat astrocytes. Cell Mol Neurobiol 2007; 27:335-50. [PMID: 17387609 PMCID: PMC11881812 DOI: 10.1007/s10571-006-9127-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Accepted: 10/16/2006] [Indexed: 11/26/2022]
Abstract
1. The leucine-rich glioma inactivated (LGI) family of genes encodes a leucine-rich repeat (LRR) protein, proteins that are thought to be specifically involved in protein-protein and protein-matrix interactions. Since amyloid beta peptide (Abeta) has been previously shown to induce the expression of another LRR-encoding gene in neural cells, we assessed how Abeta affects LGI gene expression in rat primary cerebral cortical cultures and astrocyte cultures. Both RT-PCR and Western Blotting analyses revealed that Abeta robustly induced the expression of LGI3 in rat astrocyte cultures.2. Western Blotting analyses also showed that both glial fibrillary acidic protein (GFAP) and apolipoprotein E (ApoE) significantly increased coincidentally with the Abeta-induced upregulation of LGI3. Immunocytochemistry showed that LGI3 colocalized with Abeta at plasma membranes and also with internalized Abeta in astrocytes. These findings suggest that activated LGI3 may be involved in the astroglial response against Abeta.
Collapse
Affiliation(s)
- Nobuyuki Kimura
- Laboratory of Disease Control, Tsukuba Primate Research Center, National Institute of Biomedical Innovation, 1-1 Hachimandai, Tsukuba-shi, Ibaraki 35-0843, Japan.
| | | | | | | | | | | |
Collapse
|
414
|
Hinman JD, Abraham CR. What's behind the decline? The role of white matter in brain aging. Neurochem Res 2007; 32:2023-31. [PMID: 17447140 DOI: 10.1007/s11064-007-9341-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 03/23/2007] [Indexed: 02/05/2023]
Abstract
The specific molecular events that underlie the age-related loss of cognitive function are poorly understood. Although not experimentally substantiated, age-dependent neuronal loss has long been considered central to age-related cognitive decline. More recently, age-related changes in brain white matter have taken precedence in explaining the steady decline in cognitive domains seen in non-diseased elderly. Characteristic alterations in the ultrastructure of myelin coupled with evidence of inflammatory processes present in the white matter of several different species suggest that specific molecular events within brain white matter may better explain observed pathological changes and cognitive deficits. This review focuses on recent evidence highlighting the importance of white matter in deciphering the course of "normal" brain aging.
Collapse
Affiliation(s)
- Jason D Hinman
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | | |
Collapse
|
415
|
He Y, Li HL, Xie WY, Yang CZ, Yu ACH, Wang Y. The presence of active Cdk5 associated with p35 in astrocytes and its important role in process elongation of scratched astrocyte. Glia 2007; 55:573-83. [PMID: 17295212 DOI: 10.1002/glia.20485] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a unique member of the Cdk family; its kinase activity requires association with its activator, p35 or p39. p35 is the strongest and best characterized activator. Previous studies showed that p35 is a neuron-specific protein that restricts Cdk5 activity in neurons. However, a high expression level of Cdk5 is found in astrocytes, which raises the possibility that astrocytic Cdk5 is functional. Here we show the presence of functional Cdk5 associated with p35 in astrocytes and demonstrate its important role in process elongation of scratched astrocytes. We found that p35 and glial fibrillary acidic protein (GFAP) were co-localized in primary cultured and acute isolated brain cells. Cdk5 could form an immunocomplex with p35 and its activity was shown in pure primary cultured astrocytes. p35 was upregulated in astrocytes injured by scratching, concomitantly with upregulation of Cdk5 kinase activity. Pretreatment of the scratched astrocytes with a Cdk5 inhibitor, roscovitine, could delay wound healing by inhibiting the reorganization of tubulin, GFAP, and the extension of hypertrophic processes. Moreover, overexpression of dominant negative Cdk5 could shorten the length of extending protrusion of reactive astrocytes. Thus, our findings demonstrated that functional Cdk5, associated with p35, was expressed in astrocytes and its activity could be upregulated in reactive astrocytes, a new role of Cdk5 that has never been reported in the nervous system. The present study may provide new insight for understanding the multifunctional protein complex Cdk5/p35 in the nervous system.
Collapse
Affiliation(s)
- Yi He
- Neuroscience Research Institute, Department of Neurobiology, the Key Laboratory for Neuroscience, Peking University, Beijing 100083, People's Republic of China
| | | | | | | | | | | |
Collapse
|
416
|
Bourguignon LYW, Gilad E, Peyrollier K, Brightman A, Swanson RA. Hyaluronan-CD44 interaction stimulates Rac1 signaling and PKN gamma kinase activation leading to cytoskeleton function and cell migration in astrocytes. J Neurochem 2007; 101:1002-17. [PMID: 17403031 DOI: 10.1111/j.1471-4159.2007.04485.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Both hyaluronan [HA, the major glycosaminoglycans in the extracellular matrix (ECM)] and CD44 (a primary HA receptor) are associated with astrocyte activation and tissue repair following central nervous system (CNS) injury. In this study we investigated the question of whether HA-CD44 interaction influences astrocyte signaling and migration. Our data indicated that HA binding to the cultured astrocytes stimulated Rac1 signaling and cytoskeleton-mediated migration. To determine the cellular and molecular basis of these events, we focused on PKN gamma, a Rac1-activated serine/threonine kinase in astrocytes. We determined that HA binding to astrocytes stimulated Rac1-dependent PKN gamma kinase activity which, in turn, up-regulated the phosphorylation of the cytoskeletal protein, cortactin, and attenuated the ability of cortactin to cross-link F-actin. Further analyses indicated that the N-terminal antiparallel coiled-coil (ACC) domains of PKN gamma interacted with Rac1, and transfection of astrocytes with PKN gamma-ACCcDNA inhibited PKN gamma activity. Over-expression of the PKN gamma-ACC domain also functions as a dominant-negative mutant to block HA/CD44-mediated PKN gamma activation of cortactin and astrocyte migration. Taken together, these findings strongly suggest that hyaluronan/CD44 interaction with Rac1-PKN gamma plays a pivotal role in cytoskeleton activation and astrocyte migration. These newly discovered HA/CD44-induced astrocyte function may provide important insight into novel therapeutic treatments for tissue repair following CNS injury.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- Department of Medicine, University of California San Francisco, Endocrine Unit (111 N), San Francisco VA Medical Center, San Francisco, CA 94121, USA.
| | | | | | | | | |
Collapse
|
417
|
Blechingberg J, Holm IE, Nielsen KB, Jensen TH, Jørgensen AL, Nielsen AL. Identification and characterization of GFAPkappa, a novel glial fibrillary acidic protein isoform. Glia 2007; 55:497-507. [PMID: 17203480 DOI: 10.1002/glia.20475] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is the principal component of the intermediary filaments in mature astrocytes of the central nervous system (CNS). The protein consists of three domains: the head, the coiled-coil, and the tail. Here, we describe the isolation of an evolutionary conserved novel GFAP isoform, GFAPkappa, produced by alternative splicing and polyadenylation of the 3'-region of the human GFAP pre-mRNA. As a consequence, the resulting human GFAPkappa protein harbors a nonconserved C-terminal tail sequence distinct from the tails of GFAPalpha, the predominant GFAP isoform, and GFAPepsilon, an isoform which also results from alternative splicing. The head and coiled-coil rod domains are identical between the three GFAP isoforms. Interestingly, GFAPkappa is incapable of forming homomeric filaments, and increasing GFAPkappa expression levels causes a collapse of intermediate filaments formed by GFAPalpha. In searching for a biological relevance of GFAPkappa, we noticed that mRNA expression levels of GFAPalpha, GFAPepsilon, and GFAPkappa are gradually increased during development of the embryonic pig brain. However, whereas the GFAPalpha/GFAPepsilon ratio is constant, the GFAPkappa/GFAPepsilon ratio decreases during brain development. Furthermore, in glioblastoma tumors, an increased GFAPkappa/GFAPepsilon ratio is detected. Our results suggest that the relative expression level of the GFAPkappa isoform could modulate the properties of GFAP intermediate filaments and perhaps thereby influencing the motility of GFAP positive astrocytes and progenitor cells within the CNS.
Collapse
Affiliation(s)
- Jenny Blechingberg
- Institute of Human Genetics, The Bartholin Building, University of Aarhus, Aarhus C DK-8000, Denmark
| | | | | | | | | | | |
Collapse
|
418
|
Fawcett JW. The Glial Response to Injury and Its Role in the Inhibition of CNS Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:11-24. [PMID: 16955702 DOI: 10.1007/0-387-30128-3_2] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
419
|
|
420
|
Calandrella N, Scarsella G, Pescosolido N, Risuleo G. Degenerative and apoptotic events at retinal and optic nerve level after experimental induction of ocular hypertension. Mol Cell Biochem 2007; 301:155-63. [PMID: 17242991 DOI: 10.1007/s11010-006-9407-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Accepted: 12/20/2006] [Indexed: 11/28/2022]
Abstract
Ocular hypertension is a symptom of a glaucomatous condition characterized by a severe vision decrease. Blindness caused by the apoptotic death of the retinal ganglion cells and of the astrocytes of the optic nerve may eventually result. Experimental hypertension was induced by inoculation of methylcellulose in the anterior chamber. Chromatin staining, TUNEL assay, and inter-nucleosomal DNA fragmentation observed in retina and optic nerve strongly suggest that hypertension causes apoptosis. Immunolocalization of the fibrillary acidic glial protein, specific of cell stress, and caspase-3 in the same tissues, further support this mode of cell death. Activation of the ubiquitin dependent proteolytic system was also observed. Protection from apoptosis exerted by administration of the peroxide scavenger trolox, suggests that the apoptotic pathway is activated by an oxidative stress. The data presented here show that the experimental hypertensive insult induces degenerative and apoptotic events comparable to those observed in human glaucoma.
Collapse
Affiliation(s)
- Nicola Calandrella
- Dipartimento di Biologia Cellulare e dello Sviluppo, Università di Roma La Sapienza, Piazzale Aldo Moro, Rome, Italy
| | | | | | | |
Collapse
|
421
|
Chesik D, De Keyser J, Glazenburg L, Wilczak N. Insulin-like growth factor binding proteins: regulation in chronic active plaques in multiple sclerosis and functional analysis of glial cells. Eur J Neurosci 2007; 24:1645-52. [PMID: 17004928 DOI: 10.1111/j.1460-9568.2006.05034.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Studies in experimental allergic encephalomyelitis, an animal model of multiple sclerosis (MS), suggest that astrocyte-secreted insulin-like growth factor binding protein-2 (IGFBP-2) helps target IGF-1 to IGF-1 receptor-expressing oligodendrocytes and promote remyelination. We examined the presence of IGFBPs 1-6 in astrocytes in normal post-mortem human brain tissue and lesions of MS by means of immunohistochemistry. Under normal conditions all six IGFBPs were detected. Compared to controls, hypertrophic astrocytes at the borders of chronic active MS lesions displayed increased immunoreactivity for IGFBP-2 and IGFBP-4. In vitro studies were performed to analyse the effects of IGFBPs on cellular proliferation of neonatal rat glial cells. Treatment of astrocytes with IGF-1 and -2 enhanced proliferation whereas IGFBP-2 and -4 inhibited cellular growth. Interestingly, combined treatment with IGFBP-2 and IGF-1 potentiated effects on cellular proliferation whereas combined treatment with IGFBP-2 and IGF-2 inhibited growth. Unlike IGFBP-2, IGFBP-4 inhibited proliferation in combined treatment with IGF-1. In contrast, combined treatment with IGFBP-2 and IGF-1 resulted in decreased cell survival of oligodendrocyte precursor cells. Our results suggest that the up-regulation of IGFBP-2 in reactive astrocytes in MS lesions may primarily serve to enhance the IGF-1-mediated mitogenic stimulus for astrocytes rather than supporting oligodendrocyte survival.
Collapse
Affiliation(s)
- Daniel Chesik
- Department of Neurology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | | | | | | |
Collapse
|
422
|
Chesik D, Wilczak N, De Keyser J. The insulin-like growth factor system in multiple sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 79:203-26. [PMID: 17531843 DOI: 10.1016/s0074-7742(07)79009-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a chronic disorder of the central nervous system characterized by inflammation, demyelination, and axonal degeneration. Present therapeutic strategies for MS reduce inflammation and its destructive consequences, but are not effective in the progressive phase of the disease. There is a need for neuroprotective and restorative therapies in MS. Insulin-like growth factor-1 (IGF-1) is of considerable interest because it is not only a potent neuroprotective trophic factor but also a survival factor for cells of the oligodendrocyte lineage and possesses a potent myelinogenic capacity. However, the IGF system is complex and includes not only IGF-1 and IGF-2 and their receptors but also modulating IGF-binding proteins (IGFBPs), of which six have been identified. This chapter provides an overview of the role of the IGF system in the pathophysiology of MS, relevant findings in preclinical models, and discusses the possible use of IGF-1 as a therapeutic agent for MS.
Collapse
Affiliation(s)
- Daniel Chesik
- Department of Neurology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | | | | |
Collapse
|
423
|
Britschgi M, Wyss-Coray T. Systemic and acquired immune responses in Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 82:205-33. [PMID: 17678963 DOI: 10.1016/s0074-7742(07)82011-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized clinically by a progressive cognitive decline and dementia. AD brains are marked by amyloid plaques and neurofibrillary tangles, neuronal cell loss, and a prominent activation of glial cells, and innate immune responses. A growing number of studies in AD have also reported alterations in systemic immune responses including changes in lymphocyte and macrophage distribution and activation, the presence of autoantibodies, or abnormal cytokine production. Studies in animal models for AD support the notion that immune cells infiltrate the brain and may modulate the disease. Here we will review evidence for systemic alterations in immune responses and a role for acquired immunity in AD and discuss their potential contribution to the disease.
Collapse
Affiliation(s)
- Markus Britschgi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | | |
Collapse
|
424
|
|
425
|
Pekny M, Wilhelmsson U, Bogestål YR, Pekna M. The role of astrocytes and complement system in neural plasticity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 82:95-111. [PMID: 17678957 DOI: 10.1016/s0074-7742(07)82005-8] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In neurotrauma, brain ischemia or neurodegenerative diseases, astrocytes become reactive (which is known as reactive gliosis) and this is accompanied by an altered expression of many genes. Two cellular hallmarks of reactive gliosis are hypertrophy of astrocyte processes and the upregulation of the part of the cytoskeleton known as intermediate filaments, which are composed of nestin, vimentin, and GFAP. Our aim has been to better understand the function of reactive astrocytes in CNS diseases. Using mice deficient for astrocyte intermediate filaments (GFAP(-/-)Vim(-/-)), we were able to attenuate reactive gliosis and slow down the healing process after neurotrauma. We demonstrated the key role of reactive astrocytes in neurotrauma-at an early stage after neurotrauma, reactive astrocytes have a neuroprotective effect; at a later stage, they facilitate the formation of posttraumatic glial scars and inhibit CNS regeneration, specifically, they seem to compromise neural graft survival and integration, reduce the extent of synaptic regeneration, inhibit neurogenesis in the old age, and inhibit regeneration of severed CNS axons. We propose that reactive astrocytes are the future target for the therapeutic strategies promoting regeneration and plasticity in the brain and spinal cord in various disease conditions. Through its involvement in inflammation, opsonization, and cytolysis, complement protects against infectious agents. Although most of the complement proteins are synthesized in CNS, the role of the complement system in the normal or ischemic CNS remains unclear. Complement activiation in the CNS has been generally considered as contributing to tissue damage. However, growing body of evidence suggests that complement may be a physiological neuroprotective mechanism as well as it may participate in maintenance and repair of the adult brain.
Collapse
Affiliation(s)
- Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute for Neuroscience and Physiology at Sahlgrenska Academy Göteborg University, 405 30 Göteborg, Sweden
| | | | | | | |
Collapse
|
426
|
Zhu Z, Zhang Q, Yu Z, Zhang L, Tian D, Zhu S, Bu B, Xie M, Wang W. Inhibiting cell cycle progression reduces reactive astrogliosis initiated by scratch injury in vitro and by cerebral ischemia in vivo. Glia 2007; 55:546-58. [PMID: 17243097 DOI: 10.1002/glia.20476] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Astrogliosis occurs in a variety of neuropathological disorders and injuries, and excessive astrogliosis can be devastating to the recovery of neuronal function. In this study, we asked whether reactive astrogliosis can be suppressed in the lesion area by cell cycle inhibition and thus have therapeutic benefits. Reactive astrogliosis induced in either cultured astrocytes by hypoxia or scratch injury, or in a middle cerebral artery occlusion (MCAO) ischemia model were combined to address this issue. In the cultured astrocytes, hypoxia induced a cell cycle activation that was associated with upregulation of the proliferating cell nuclear marker (PCNA). Significantly, the cell cycle inhibitor, olomoucine, inhibited hypoxia-induced cell cycle activation by arresting the cells at G1/S and G2/M in a dose-dependent manner and also reversed hypoxia-induced upregulation of PCNA. Also in the cultured astrocytes, scratch injury induced reactive astrogliosis, such as hypertrophy and an increase in BrdU(+) astrocytes, both of which were ameliorated by olomoucine. In the MCAO ischemia mouse model, dense reactive glial fibrillary acidic protein and PCNA immunoreactivity were evident at the boundary zone of focal cerebral ischemia at days 7 and 30 after MCAO. We found that intraperitoneal olomoucine administration significantly inhibited these astrogliosis-associated changes. To demonstrate further that cell cycle regulation impacts on astrogliosis, cyclin D1 gene knockout mice (cyclin D1(-/-)) were subjected to ischemia, and we found that the percentage of Ki67-positive astrocytes in these mice was markedly reduced in the boundary zone. The number of apoptotic neurons and the lesion volume in cyclin D1(-/-) mice also decreased as compared to cyclin D1(+/+) and cyclin D1(+/-) mice at days 3, 7, and 30 after local cerebral ischemia. Together, these in vitro and in vivo results strongly suggest that astrogliosis can be significantly affected by cell cycle inhibition, which therefore emerges as a promising intervention to attenuate reactive glia-related damage to neuronal function in brain pathology.
Collapse
Affiliation(s)
- Zhou Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
427
|
Abstract
Patients with Alzheimer's disease or other neurodegenerative disorders show remarkable fluctuations in neurological functions, even during the same day. These fluctuations cannot be caused by sudden loss or gain of nerve cells. Instead, it is likely that they reflect variations in the activity of neural networks and, perhaps, chronic intoxication by abnormal proteins that the brain is temporarily able to overcome. These ideas have far-reaching therapeutic implications.
Collapse
Affiliation(s)
- Jorge J Palop
- Gladstone Institute of Neurological Disease and Department of Neurology, University of California, San Francisco, California 94158, USA
| | | | | |
Collapse
|
428
|
Wilhelmsson U, Bushong EA, Price DL, Smarr BL, Phung V, Terada M, Ellisman MH, Pekny M. Redefining the concept of reactive astrocytes as cells that remain within their unique domains upon reaction to injury. Proc Natl Acad Sci U S A 2006; 103:17513-8. [PMID: 17090684 PMCID: PMC1859960 DOI: 10.1073/pnas.0602841103] [Citation(s) in RCA: 451] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Reactive astrocytes in neurotrauma, stroke, or neurodegeneration are thought to undergo cellular hypertrophy, based on their morphological appearance revealed by immunohistochemical detection of glial fibrillary acidic protein, vimentin, or nestin, all of them forming intermediate filaments, a part of the cytoskeleton. Here, we used a recently established dye-filling method to reveal the full three-dimensional shape of astrocytes assessing the morphology of reactive astrocytes in two neurotrauma models. Both in the denervated hippocampal region and the lesioned cerebral cortex, reactive astrocytes increased the thickness of their main cellular processes but did not extend to occupy a greater volume of tissue than nonreactive astrocytes. Despite this hypertrophy of glial fibrillary acidic protein-containing cellular processes, interdigitation between adjacent hippocampal astrocytes remained minimal. This work helps to redefine the century-old concept of hypertrophy of reactive astrocytes.
Collapse
Affiliation(s)
- Ulrika Wilhelmsson
- *Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Göteborg University, SE-405 30 Göteborg, Sweden; and
| | - Eric A. Bushong
- National Center for Microscopy and Imaging Research, University of California at San Diego, La Jolla, CA 92093-0608
| | - Diana L. Price
- National Center for Microscopy and Imaging Research, University of California at San Diego, La Jolla, CA 92093-0608
| | - Benjamin L. Smarr
- National Center for Microscopy and Imaging Research, University of California at San Diego, La Jolla, CA 92093-0608
| | - Van Phung
- National Center for Microscopy and Imaging Research, University of California at San Diego, La Jolla, CA 92093-0608
| | - Masako Terada
- National Center for Microscopy and Imaging Research, University of California at San Diego, La Jolla, CA 92093-0608
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, University of California at San Diego, La Jolla, CA 92093-0608
| | - Milos Pekny
- *Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Göteborg University, SE-405 30 Göteborg, Sweden; and
- To whom correspondence should be addressed at:
Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, Göteborg University, Box 440, SE-405 30 Göteborg, Sweden. E-mail:
| |
Collapse
|
429
|
Hamby ME, Hewett JA, Hewett SJ. TGF-beta1 potentiates astrocytic nitric oxide production by expanding the population of astrocytes that express NOS-2. Glia 2006; 54:566-77. [PMID: 16921522 DOI: 10.1002/glia.20411] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Both transforming growth factor-beta1 (TGF-beta1) and nitric oxide synthase-2 (NOS-2) are upregulated under various neuropathological states. Evidence suggests that TGF-beta1 can either attenuate or augment NOS-2 expression, with the prevailing effect dependent on the experimental paradigm employed and the cell-type under study. The purpose of the present study was to determine the effect of TGF-beta1 on astrocytic NOS-2 expression. In purified astrocyte cultures, TGF-beta1 alone did not induce NOS-2 or NO production. However, NO production induced by lipopolysaccharide (LPS) plus IFNgamma was enhanced by TGF-beta1 in a concentration-dependent manner between 10 and 1,000 pg/mL. The presence of IFNgamma was not necessary for this effect to occur, as TGF-beta1 enhanced NO production induced by LPS in a similar fashion. In cultures stimulated with LPS plus IFNgamma, the enhancement of NO production by TGF-beta1 was associated with a corresponding increase in NOS-2 mRNA and protein expression. Interestingly, immunocytochemical assessment of NOS-2 protein expression demonstrated that TGF-beta1 augmented astrocytic NO production, specifically by increasing the pool of astrocytes capable of expressing NOS-2 induced by either LPS (approximately threefold) or LPS plus IFNgamma (approximately sevenfold). In a broader sense, our results suggest that TGF-beta1 recruits a latent population of astrocytes to respond to stimulation by pro-inflammatory mediators.
Collapse
Affiliation(s)
- Mary E Hamby
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | |
Collapse
|
430
|
Lechuga-Sancho AM, Arroba AI, Frago LM, García-Cáceres C, de Célix ADR, Argente J, Chowen JA. Reduction in the number of astrocytes and their projections is associated with increased synaptic protein density in the hypothalamus of poorly controlled diabetic rats. Endocrinology 2006; 147:5314-24. [PMID: 16873533 DOI: 10.1210/en.2006-0766] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Processes under hypothalamic control, such as thermogenesis, feeding behavior, and pituitary hormone secretion, are disrupted in poorly controlled diabetes, but the underlying mechanisms are poorly understood. Because glial cells regulate neurosecretory neurons through modulation of synaptic inputs and function, we investigated the changes in hypothalamic glia in rats with streptozotocin-induced diabetes mellitus. Hypothalamic glial fibrillary acidic protein (GFAP) levels decreased significantly 6 wk after diabetes onset. This was coincident with decreased GFAP immunoreactive surface area, astrocyte number, and the extension of GFAP immunoreactive processes/astrocyte in the arcuate nucleus. Cell death, analyzed by terminal deoxyuridine 5-triphosphate nick-end labeling and ELISA, increased significantly at 4 wk of diabetes. Proliferation, measured by Western blot for proliferating cell nuclear antigen and immunostaining for phosphorylated histone H-3, decreased in the hypothalamus of diabetic rats throughout the study, becoming significantly reduced by 8 wk. Both proliferation and death affected astroctyes because both phosphorylated histone H-3- and terminal deoxyuridine 5-triphosphate nick-end labeling-labeled cells were GFAP positive. Western blot analysis revealed that postsynaptic density protein 95 and the presynaptic proteins synapsin I and synaptotagmin increased significantly at 8 wk of diabetes, suggesting increased hypothalamic synaptic density. Thus, in poorly controlled diabetic rats, there is a decrease in the number of hypothalamic astrocytes that is correlated with modifications in synaptic proteins and possibly synaptic inputs. These morphological changes in the arcuate nucleus could be involved in neurosecretory and metabolic changes seen in diabetic animals.
Collapse
Affiliation(s)
- Alfonso M Lechuga-Sancho
- Hospital Infantil Universitario Niño Jesús, Departamento de Endocrinología, Avenida Menéndez Pelayo, 65, 28009 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
431
|
Ovanesov MV, Sauder C, Rubin SA, Richt J, Nath A, Carbone KM, Pletnikov MV. Activation of microglia by borna disease virus infection: in vitro study. J Virol 2006; 80:12141-8. [PMID: 17020949 PMCID: PMC1676289 DOI: 10.1128/jvi.01648-06] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neonatal Borna disease virus (BDV) infection of the rat brain is associated with microglial activation and damage to the certain neuronal populations. Since persistent BDV infection of neurons in vitro is noncytolytic and noncytopathic, activated microglia have been suggested to be responsible for neuronal cell death in vivo. However, the mechanisms of activation of microglia in neonatally BDV-infected rat brain have not been investigated. To address these issues, activation of primary rat microglial cells was studied following exposure to purified BDV or to persistently BDV-infected primary cortical neurons or after BDV infection of primary mixed neuron-glial cultures. Neither purified virus nor BDV-infected neurons alone activated primary microglia as assessed by the changes in cell shape or production of the proinflammatory cytokines. In contrast, in the BDV-infected primary mixed cultures, we observed proliferation of microglia cells that acquired the round morphology and expressed major histocompatibility complex molecules of classes I and II. These manifestations of microglia activation were observed in the absence of direct BDV infection of microglia or overt neuronal toxicity. In addition, compared to uninfected mixed cultures, activation of microglia in BDV-infected mixed cultures was associated with a significantly greater lipopolysaccharide-induced release of tumor necrosis factor alpha, interleukin 1beta, and interleukin 10. Taken together, the present data are the first in vitro evidence that persistent BDV infection of neurons and astrocytes rather than direct exposure to the virus or dying neurons is critical for activating microglia.
Collapse
Affiliation(s)
- Mikhail V Ovanesov
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, CMSC 8-121, Baltimore, MD 21287, USA
| | | | | | | | | | | | | |
Collapse
|
432
|
Dorta-Contreras AJ, Lewczuc P, Noris-García E, Interián-Morales MT, Magraner Tarrau ME, Padilla-Docal B, Escobar-Pérez X. sICAM-1 in meningoencephalitis due to Angiostrongylus cantonensis. ARQUIVOS DE NEURO-PSIQUIATRIA 2006; 64:589-91. [PMID: 17119798 DOI: 10.1590/s0004-282x2006000400011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2005] [Accepted: 03/17/2006] [Indexed: 11/21/2022]
Abstract
INTRODUCTION: Angiostrongylus cantonensis meningoencephalitis is an emergent disease in the Americas. METHOD: Twelve children suffering from eosinophilic meningoencephalitis due to this parasite aged between 6-10 years were studied. Cerebrospinal fluid (CSF) and serum samples were taken simultaneously in the first diagnostic puncture at admission. RESULTS: All cases showed typical findings on the routine CSF and serum analysis: increased CSF total protein, increased Q (CSF/serum) albumin accompanied by eosinophilia in CSF. No intrathecal synthesis of immunoglobulins was found. Mean serum and CSF sICAM-1 values were 337.4 and 3.97 ng/mL. Qalbumin and QsICAM-1 mean values were 4.1 and 6.2 respectively. In 50% of the patients an increased brain-derived fraction of sICAM-1 was found. CONCLUSION: It may be suggested that a dynamic of the sICAM-1 brain derived fraction is perhaps associated to the immune response in the evolution of the disease.sICAM-1 may be an agent in negative feedback for eosinophils passage through the blood-CSF barrier into the inflammatory brain response.
Collapse
|
433
|
Orihuela CJ, Fillon S, Smith-Sielicki SH, El Kasmi KC, Gao G, Soulis K, Patil A, Murray PJ, Tuomanen EI. Cell wall-mediated neuronal damage in early sepsis. Infect Immun 2006; 74:3783-9. [PMID: 16790750 PMCID: PMC1489725 DOI: 10.1128/iai.00022-06] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neuronal dysfunction can occur in the course of sepsis without meningitis. Sepsis-associated neuronal damage (SAND) was observed in the hippocampus within hours in experimental pneumococcal bacteremia. Intravascular challenge with purified bacterial cell wall recapitulated SAND. SAND persisted in PAFr(-/-) mice but was partially mitigated in mice lacking cell wall recognition proteins TLR2 and Nod2 and in mice overexpressing interleukin-10 (IL-10) in macrophages. Thus, cell wall drives SAND through IL-10-repressible inflammatory events. Treatment with CDP-choline ameliorated SAND, suggesting that it may be an effective adjunctive therapy to increase survival and reduce organ damage in sepsis.
Collapse
Affiliation(s)
- Carlos J Orihuela
- Infectious Diseases, St. Jude Children's Research Hospital, Mailstop 320 IRC 8057, 332 N. Lauderdale Rd., Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
434
|
Silva AMM, Silva AR, Pinheiro AM, Freitas SRVB, Silva VDA, Souza CS, Hughes JB, El-Bachá RS, Costa MFD, Velozo ES, Tardy M, Costa SL. Alkaloids from Prosopis juliflora leaves induce glial activation, cytotoxicity and stimulate NO production. Toxicon 2006; 49:601-14. [PMID: 17241650 DOI: 10.1016/j.toxicon.2006.07.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2006] [Revised: 07/21/2006] [Accepted: 07/25/2006] [Indexed: 11/19/2022]
Abstract
Prosopis juliflora is used for feeding cattle and humans. Intoxication with the plant has been reported, and is characterized by neuromuscular alterations and gliosis. Total alkaloidal extract (TAE) was obtained using acid/basic-modified extraction and was fractionated. TAE and seven alkaloidal fractions, at concentrations ranging 0.03-30 microg/ml, were tested for 24h on astrocyte primary cultures derived from the cortex of newborn Wistar rats. The MTT test and the measure of LDH activity on the culture medium, revealed that TAE and fractions F29/30, F31/33, F32 and F34/35 were cytotoxic to astrocytes. The EC(50) values for the most toxic compounds, TAE, F31/33 and F32 were 2.87 2.82 and 3.01 microg/ml, respectively. Morphological changes and glial cells activation were investigated through Rosenfeld's staining, by immunocytochemistry for the protein OX-42, specific of activated microglia, by immunocytochemistry and western immunoblot for GFAP, the marker of reactive and mature astrocytes, and by the production of nitric oxide (NO). We observed that astrocytes exposed to 3 microg/ml TAE, F29/30 or F31/33 developed compact cell body with many processes overexpressing GFAP. Treatment with 30 microg/ml TAE and fractions, induced cytotoxicity characterized by a strong cell body contraction, very thin and long processes and condensed chromatin. We also observed that when compared with the control (+/-1.34%), the proportion of OX-42 positive cells was increased in cultures treated with 30 microg/ml TAE or F29/30, F31/33, F32 and F34/35, with values raging from 7.27% to 28.74%. Moreover, incubation with 3 microg/ml F32, 30 microg/ml TAE, F29/30, F31/33 or F34/35 induced accumulation of nitrite in culture medium indicating induction of NO production. Taken together these results show that TAE and fractionated alkaloids from P. juliflora act directly on glial cells, inducing activation and/or cytotoxicity, stimulating NO production, and may have an impact on neuronal damages observed on intoxicated animals.
Collapse
Affiliation(s)
- A M M Silva
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Biofunção, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, BA, 40.110-100, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
435
|
Koyama Y, Kiyo-oka M, Osakada M, Horiguchi N, Shintani N, Ago Y, Kakuda M, Baba A, Matsuda T. Expression of prokineticin receptors in mouse cultured astrocytes and involvement in cell proliferation. Brain Res 2006; 1112:65-9. [PMID: 16901473 DOI: 10.1016/j.brainres.2006.07.013] [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] [Received: 02/13/2006] [Revised: 06/30/2006] [Accepted: 07/04/2006] [Indexed: 11/30/2022]
Abstract
Effects of prokineticins (PKs), a novel family of bioactive peptides with a mitogenic action to endothelial cells of the endocrine gland and testis, on astrocytic functions were examined. Mouse cultured astrocytes expressed PK-R1 type PK receptors, while there was little expression of the PK-R2 type. PKs caused increases in astrocytic cytosolic Ca2+ levels and BrdU incorporation. Increases in Ca2+ levels by PK-2 were diminished by U73122 (a phospholipase C inhibitor). PK-induced BrdU incorporation was inhibited by U73122, GF109203 (a protein kinase C inhibitor) and PD98059 (a MEK/ERK inhibitor). These results indicate that PK receptors are expressed in astrocytes and regulate astrocytic proliferation.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-Oka 1-6 Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
436
|
Vidal L, Díaz F, Villena A, Moreno M, Campos JG, de Vargas IP. Nitric oxide synthase in retina and optic nerve head of rat with increased intraocular pressure and effect of timolol. Brain Res Bull 2006; 70:406-13. [PMID: 17027776 DOI: 10.1016/j.brainresbull.2006.07.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 06/23/2006] [Accepted: 07/14/2006] [Indexed: 11/16/2022]
Abstract
We investigated the expression of nitric oxide synthase (NOS) isoforms -1, -2 and -3 in the retina and optic nerve head (ONH) in an experimental rat model of elevated intraocular pressure (IOP) before and after treatment with timolol, to assess whether its neuroprotective action is associated with the activity of these enzymes. Episcleral vein cauterization in unilateral eyes of Wistar rats was performed to produce elevated IOP. Histological sections of retina and ONH from animals with normal IOP, with elevated IOP, and elevated IOP treated with timolol, were studied by immunohistochemistry with antibodies to NOS-1, NOS-2, and NOS-3. In the control rats, NOS-1 was localized to photoreceptor inner segments, amacrine cells and bipolar cells in the retina, and in astrocytes, pericytes and vascular nitrergic terminals in the ONH. NOS-3 immunostaining localized to the endothelial cells. The rats with elevated IOP showed increased expression of NOS-1 in the plexiform layers of the retina and reactive astrocytes in the ONH. These cells also showed NOS-2 positivity. The rats treated with timolol showed reduced expression of NOS-1 in the retina and ONH. NOS-2 was only detected in a few groups of astrocytes in the ONH. NOS-3 was unchanged in both elevated IOP and timolol-treated groups. These results show that excessive levels of NO synthesized by the NOS-1 and -2 isoforms, considered neurotoxic, might contribute to the progressive lesions of retinal ganglion cell axons. Their reduction after treatment suggests a possible neuroprotective effect of timolol in neurons exposed to excessive amounts of NO.
Collapse
Affiliation(s)
- Lourdes Vidal
- Department of Histology and Histopathology, School of Medicine, University of Malaga, Boulevard Louis Pasteur 32, 29071 Malaga, Spain.
| | | | | | | | | | | |
Collapse
|
437
|
Liu B, Chen H, Johns TG, Neufeld AH. Epidermal growth factor receptor activation: an upstream signal for transition of quiescent astrocytes into reactive astrocytes after neural injury. J Neurosci 2006; 26:7532-40. [PMID: 16837601 PMCID: PMC6674203 DOI: 10.1523/jneurosci.1004-06.2006] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Modulating the behaviors of reactive astrocytes is a potential therapeutic strategy for neurodegenerative diseases. We found that upregulation and activation of the epidermal growth factor receptor (EGFR) occur in astrocytes after different injuries in optic nerves in vivo. Activation of EGFR regulates genes and cellular processes representing most major markers of reactive astrocytes and genes related with glaucomatous optic neuropathy and other neural disorders. These results suggest that activation of EGFR is a common, regulatory pathway that triggers quiescent astrocytes into reactive astrocytes in response to neural injuries in the optic nerve, and perhaps other parts of the CNS. Targeting EGFR activation using an EGFR tyrosine kinase inhibitor prevents the loss of retinal ganglion cells in a model of glaucomatous optic neuropathy. Because these inhibitors are currently used clinically, our results present an approach to reactive astrocytes as a potential new target for the treatment of neurodegenerations.
Collapse
Affiliation(s)
- Bin Liu
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | |
Collapse
|
438
|
Ricci-Vitiani L, Casalbore P, Petrucci G, Lauretti L, Montano N, Larocca LM, Falchetti ML, Lombardi DG, Gerevini VDG, Cenciarelli C, D'Alessandris QG, Fernandez E, De Maria R, Maira G, Peschle C, Parati E, Pallini R. Influence of local environment on the differentiation of neural stem cells engrafted onto the injured spinal cord. Neurol Res 2006; 28:488-492. [PMID: 16808877 DOI: 10.1179/016164106x115134] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES In vitro, neural stem cells (NSCs) proliferate as undifferentiated spheroids and differentiate into neurons, astrocytes and oligodendrocytes. These features make NSCs suitable for spinal cord (SC) reconstruction. However, in vivo experiments have demonstrated that in the injured SC transplanted NSCs either remain undifferentiated or differentiate into the astrocytic phenotype. The microenvironment of the injured SC is believed to play a crucial role in driving the differentiation of the engrafted NSCs. Here, we tested the hypothesis that inflammatory cytokines (ICs) may be involved in the restricted differentiation of NSCs after grafting onto the injured SC. METHODS As the first step, we used immunohistochemistry to analyse the expression of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta and interferon (IFN)-gamma in the normal SC of mice and following traumatic injury. Then, we investigated whether a combination of TNF-alpha, IL-1beta and IFN-gamma may affect the phenotype of murine NSCs in vitro. RESULTS We found that TNF-alpha, IL-1beta and IFN-gamma, which are absent in the normal SC, are all expressed in the injured SC and the expression of these cytokines follows a timely tuned fashion with IFN-gamma being detectable as long as 4 weeks after injury. In culture, exposure of proliferating NSCs to a combination of TNF-alpha, IL-1beta and IFN-gamma was per se sufficient to induce the astrocytic differentiation of these cells even in the absence of serum. CONCLUSIONS In the traumatically injured SC, differentiation of engrafted NSCs is restricted towards the astrocytic lineage because of the inflammatory environment. ICs are likely to play a major role in differentiation of NSCs in the in vivo conditions.
Collapse
Affiliation(s)
- Lucia Ricci-Vitiani
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
439
|
Abstract
TNF-related apoptosis inducing ligand (TRAIL) expressions were studied in primary human brain astrocytes in response to pro-inflammatory cytokines. When astrocytes were treated with IL-1beta, TNF-alpha or IFN-gamma, TRAIL was induced in cultured fetal astrocytes. In particular, IFN-gamma induced the highest levels of TRAIL in cultured astrocytes. When astrocytes were pre-treated with IFN-gamma, they induced apoptosis in TRAIL-sensitive Peer cells. Our results suggest that IFN-gamma modulates the expression of TRAIL in astrocytes, which may enhance cytotoxic sensitivity of infiltrating immune cells or brain cells other than astrocytes during inflammation of brain.
Collapse
Affiliation(s)
- Jeonggi Lee
- Department of Microbiology, Institute for Immunology and Immunological Diseases and Brain Korea 21 Project for Medical Science, Seoul, Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases and Brain Korea 21 Project for Medical Science, Seoul, Korea
| | - In-Hong Choi
- Department of Microbiology, Institute for Immunology and Immunological Diseases and Brain Korea 21 Project for Medical Science, Seoul, Korea
| |
Collapse
|
440
|
Tran MD, Neary JT. Purinergic signaling induces thrombospondin-1 expression in astrocytes. Proc Natl Acad Sci U S A 2006; 103:9321-6. [PMID: 16754856 PMCID: PMC1482608 DOI: 10.1073/pnas.0603146103] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Thrombospondin (TSP)-1, a multidomain glycoprotein, is secreted from astrocytes and promotes synaptogenesis. However, little is known about the mechanisms regulating its expression and release. In this article, we report that purinergic signaling participates in the production and secretion of TSP-1. Treatment of primary cultures of rat cortical astrocytes with extracellular ATP caused an increase in TSP-1 expression in a time- and concentration-dependent manner and was inhibited by antagonists of P2 and P1 purinergic receptors. Agonist studies revealed that UTP, but not 2',3'-O-(4-benzoyl)benzoyl-ATP, 2-methylthio-ADP, adenosine, or 5'-N-ethyl-carboxamidoadenosine, caused a significant increase in TSP-1 expression. In addition, release of TSP-1 was stimulated by ATP and UTP but not by 2-methylthio-ADP or adenosine. Additional studies indicated that P2Y(4) receptors stimulate both TSP-1 expression and release. P2Y receptors are coupled to protein kinase cascades, and signaling studies demonstrated that blockade of mitogen-activated protein kinases or Akt inhibited ATP- and UTP-induced TSP-1 expression. Using an in vitro model of CNS trauma that stimulates release of ATP, we found that TSP-1 expression increased after mechanical strain and was completely blocked by a P2 receptor antagonist and by inhibition of p38/mitogen-activated protein kinase and Akt, thereby indicating a major role for P2 receptor/protein kinase signaling in TSP-1 expression induced by trauma. We conclude that TSP-1 expression can be regulated by activation of P2Y receptors, particularly P2Y(4), coupled to protein kinase signaling pathways and suggest that purinergic signaling may be an important factor in TSP-1-mediated cell-matrix and cell-cell interactions such as those occurring during development and repair.
Collapse
Affiliation(s)
- Minh D. Tran
- Research Service, Miami Veterans Affairs Medical Center, and Departments of Pathology, Biochemistry & Molecular Biology, and the Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33125
| | - Joseph T. Neary
- Research Service, Miami Veterans Affairs Medical Center, and Departments of Pathology, Biochemistry & Molecular Biology, and the Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33125
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
441
|
Pugliese M, Geloso MC, Carrasco JL, Mascort J, Michetti F, Mahy N. Canine cognitive deficit correlates with diffuse plaque maturation and S100beta (-) astrocytosis but not with insulin cerebrospinal fluid level. Acta Neuropathol 2006; 111:519-28. [PMID: 16718348 DOI: 10.1007/s00401-006-0052-1] [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] [Received: 10/26/2005] [Revised: 12/27/2005] [Accepted: 01/30/2006] [Indexed: 10/24/2022]
Abstract
Like humans, canines develop with aging beta-amyloid (Abeta) plaques and a progressive cognitive deficit on tasks similar to those used in diagnosis and follow-up of Alzheimer's disease. Owing to that, dogs are quite unique to investigate the early events taking place in the diffuse Abeta plaque maturation and its relationship with cognitive deficit. The aim of the present investigation was to study the link between the diffuse Abeta plaque maturation and the astro- and microglial reactivity. The involvement of insulin and beta-subunit of S100 protein (S100beta) overexpression in the process was also investigated. Abeta plaques were measured and counted in prefrontal cortex of 16 pet dogs of different breeds, weight and sex, classified as control and with a light or severe cognitive deficit. A correlation between canine graded cognitive deficit, diffuse plaque maturation, and S100beta (-) astrocytosis, but not with cerebrospinal fluid insulin level, was found that may reflect the very early events of Abeta deposition in Alzheimer's disease.
Collapse
Affiliation(s)
- Marco Pugliese
- Unitat de Bioquímica, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, c/ Casanova, 143, 08036, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
442
|
de Hemptinne I, Boucherie C, Pochet R, Bantubungi K, Schiffmann SN, Maloteaux JM, Hermans E. Unilateral induction of progenitors in the spinal cord of hSOD1G93A transgenic rats correlates with an asymmetrical hind limb paralysis. Neurosci Lett 2006; 401:25-9. [PMID: 16540243 DOI: 10.1016/j.neulet.2006.02.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Revised: 02/10/2006] [Accepted: 02/21/2006] [Indexed: 10/24/2022]
Abstract
Transgenic rats expressing a mutated form of the human Cu/Zn superoxide dismutase (hSOD1(G93A)) develop an amyotrophic lateral sclerosis (ALS)-like phenotype, including motor neurone degeneration and reactive gliosis in the spinal cord. This study aimed at examining the presence of endogenous neural progenitors in the lumbar spinal cord of these rats at the end-stage of the disease. Immunohistochemical data clearly demonstrated the induced expression of the stem cell factor reported as a chemoattractant and survival factor for neural stem cells as well as nestin (neuro-epithelial stem cell intermediate filament) in the spinal cord sections. While the stem cell factor immunolabelling appeared diffuse throughout the gray matter, nestin labelling was restricted to clusters within the ventral horn. Interestingly, as paralysis regularly develops asymmetrically, induction of nestin was only detected on the ipsilateral side of the predominant symptoms. Finally, immunohistochemical detection of the stem cell factor receptor (c-Kit) revealed its specific induction which coincided with nestin immunolabelling. Together, these results are indicative of endogenous recruitment of neural progenitors within lesioned tissues and could support the development of treatments involving endogenous or exogenous stem cells.
Collapse
Affiliation(s)
- Isabelle de Hemptinne
- Laboratoire de Pharmacologie Expérimentale, Université catholique de Louvain, Av. Hippocrate 54.10, B-1200 Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
443
|
Dallas S, Miller DS, Bendayan R. Multidrug resistance-associated proteins: expression and function in the central nervous system. Pharmacol Rev 2006; 58:140-61. [PMID: 16714484 DOI: 10.1124/pr.58.2.3] [Citation(s) in RCA: 211] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Drug delivery to the brain is highly restricted, since compounds must cross a series of structural and metabolic barriers to reach their final destination, often a cellular compartment such as neurons, microglia, or astrocytes. The primary barriers to the central nervous system are the blood-brain and blood-cerebrospinal fluid barriers. Through structural modifications, including the presence of tight junctions that greatly limit paracellular transport, the cells that make up these barriers restrict diffusion of many pharmaceutically active compounds. In addition, the cells that comprise the blood-brain and blood-cerebrospinal fluid barriers express multiple ATP-dependent, membrane-bound, efflux transporters, such as members of the multidrug resistance-associated protein (MRP) family, which contribute to lowered drug accumulation. A relatively new concept in brain drug distribution just beginning to be explored is the possibility that cellular components of the brain parenchyma could act as a "second" barrier to brain permeation of pharmacological agents via expression of many of the same transporters. Indeed, efflux transporters expressed in brain parenchyma may facilitate the overall export of xenobiotics from the central nervous system, essentially handing them off to the barrier tissues. We propose that these primary and secondary barriers work in tandem to limit overall accumulation and distribution of xenobiotics in the central nervous system. The present review summarizes recent knowledge in this area and emphasizes the clinical significance of MRP transporter expression in a variety of neurological disorders.
Collapse
Affiliation(s)
- Shannon Dallas
- Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | | | | |
Collapse
|
444
|
Dhar A, Gardner J, Borgmann K, Wu L, Ghorpade A. Novel role of TGF-beta in differential astrocyte-TIMP-1 regulation: implications for HIV-1-dementia and neuroinflammation. J Neurosci Res 2006; 83:1271-80. [PMID: 16496359 PMCID: PMC3820372 DOI: 10.1002/jnr.20787] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Astrocyte production of tissue inhibitor of metalloproteinase (TIMP)-1 is important in central nervous system (CNS) homeostasis and inflammatory diseases such as HIV-1-associated dementia (HAD). TIMPs and matrix metalloproteinases (MMPs) regulate the remodeling of the extracellular matrix. An imbalance between TIMPs and MMPs is associated with many pathologic conditions. Our recently published studies uniquely demonstrate that HAD patients have reduced levels of TIMP-1 in the brain. Astrocyte-TIMP-1 expression is differentially regulated in acute and chronic inflammatory conditions. In this and the adjoining report (Gardner et al., 2006), we investigate the mechanisms that may be involved in differential TIMP-1 regulation. One mechanism for TIMP-1 downregulation is the production of anti-inflammatory molecules, which can activate signaling pathways during chronic inflammation. We investigated the contribution of transforming growth factor (TGF)-signaling in astrocyte-MMP/TIMP-1-astrocyte regulation. TGF-beta1 and beta2 levels were upregulated in HAD brain tissues. Co-stimulation of astrocytes with IL-1beta and TGF-beta mimicked the TIMP-1 downregulation observed with IL-1beta chronic activation. Measurement of astrocyte-MMP protein levels showed that TGF-beta combined with IL-1beta increased MMP-2 and decreased proMMP-1 expression compared to IL-1beta alone. We propose that one of the mechanisms involved in TIMP-1 downregulation may be through TGF-signaling in chronic immune activation. These studies show a novel extracellular regulatory loop in astrocyte-TIMP-1 regulation.
Collapse
Affiliation(s)
- Alok Dhar
- Laboratory of Cellular Neuroimmunology, University of Nebraska Medical Center, Omaha, Nebraska
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jessica Gardner
- Laboratory of Cellular Neuroimmunology, University of Nebraska Medical Center, Omaha, Nebraska
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kathleen Borgmann
- Laboratory of Cellular Neuroimmunology, University of Nebraska Medical Center, Omaha, Nebraska
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Li Wu
- Laboratory of Cellular Neuroimmunology, University of Nebraska Medical Center, Omaha, Nebraska
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Anuja Ghorpade
- Laboratory of Cellular Neuroimmunology, University of Nebraska Medical Center, Omaha, Nebraska
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
445
|
Park KI, Himes BT, Stieg PE, Tessler A, Fischer I, Snyder EY. Neural stem cells may be uniquely suited for combined gene therapy and cell replacement: Evidence from engraftment of Neurotrophin-3-expressing stem cells in hypoxic–ischemic brain injury. Exp Neurol 2006; 199:179-90. [PMID: 16714016 DOI: 10.1016/j.expneurol.2006.03.016] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 03/15/2006] [Indexed: 11/19/2022]
Abstract
Previously, we reported that, when clonal neural stem cells (NSCs) were transplanted into brains of postnatal mice subjected to unilateral hypoxic-ischemic (HI) injury (optimally 3-7 days following infarction), donor-derived cells homed preferentially (from even distant locations) to and integrated extensively within the large ischemic areas that spanned the hemisphere. A subpopulation of NSCs and host cells, particularly in the penumbra, "shifted" their differentiation towards neurons and oligodendrocytes, the cell types typically damaged following asphyxia and least likely to regenerate spontaneously and in sufficient quantity in the "post-developmental" CNS. That no neurons and few oligodendrocytes were generated from the NSCs in intact postnatal cortex suggested that novel signals are transiently elaborated following HI to which NSCs might respond. The proportion of "replacement" neurons was approximately 5%. Neurotrophin-3 (NT-3) is known to play a role in inducing neuronal differentiation during development and perhaps following injury. We demonstrated that NSCs express functional TrkC receptors. Furthermore, the donor cells continued to express a foreign reporter transgene robustly within the damaged brain. Therefore, it appeared feasible that neuronal differentiation of exogenous NSCs (as well as endogenous progenitors) might be enhanced if donor NSCs were engineered prior to transplantation to (over)express a bioactive gene such as NT-3. A subclone of NSCs transduced with a retrovirus encoding NT-3 (yielding >90% neurons in vitro) was implanted into unilaterally asphyxiated postnatal day 7 mouse brain (emulating one of the common causes of cerebral palsy). The subclone expressed NT-3 efficiently in vivo. The proportion of NSC-derived neurons increased to approximately 20% in the infarction cavity and >80% in the penumbra. The neurons variously differentiated further into cholinergic, GABAergic, or glutamatergic subtypes, appropriate to the cortex. Donor-derived glia were rare, and astroglial scarring was blunted. NT-3 likely functioned not only on donor cells in an autocrine/paracrine fashion but also on host cells to enhance neuronal differentiation of both. Taken together, these observations suggest (1) the feasibility of taking a fundamental biological response to injury and augmenting it for repair purposes and (2) the potential use of migratory NSCs in some degenerative conditions for simultaneous combined gene therapy and cell replacement during the same procedure in the same recipient using the same cell (a unique property of cells with stem-like attributes).
Collapse
Affiliation(s)
- Kook In Park
- Department of Pediatrics, and the Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
446
|
Abstract
Enteric glia regulate gastrointestinal physiology by controlling neurochemical phenotypes in the enteric nervous system
Collapse
Affiliation(s)
- A Rühl
- Department of Human Biology, Technical University of Munich, Hochfeldweg 2, D-85350 Freising-Weihenstephan, Germany.
| |
Collapse
|
447
|
Cross AK, Haddock G, Stock CJ, Allan S, Surr J, Bunning RAD, Buttle DJ, Woodroofe MN. ADAMTS-1 and -4 are up-regulated following transient middle cerebral artery occlusion in the rat and their expression is modulated by TNF in cultured astrocytes. Brain Res 2006; 1088:19-30. [PMID: 16630594 DOI: 10.1016/j.brainres.2006.02.136] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2005] [Revised: 02/21/2006] [Accepted: 02/26/2006] [Indexed: 11/26/2022]
Abstract
ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) enzymes are a recently described group of metalloproteinases. The substrates degraded by ADAMTS-1, -4 and -5 suggest that they play a role in turnover of extracellular matrix in the central nervous system (CNS). ADAMTS-1 is also known to exhibit anti-angiogenic activity. Their main endogenous inhibitor is tissue inhibitor of metalloproteinases (TIMP)-3. The present study was designed to investigate ADAMTS-1, -4 and -5 and TIMP-3 expression after experimental cerebral ischaemia and to examine whether cytokines known to be up-regulated in stroke could alter their expression by astrocytes in vitro. Focal cerebral ischaemia was induced by transient middle cerebral artery occlusion in the rat using the filament method. Our results demonstrate a significant increase in expression of ADAMTS-1 and -4 in the occluded hemisphere but no significant change in TIMP-3. This was accompanied by an increase in mRNA levels for interleukin (IL)-1beta, IL-1 receptor antagonist (IL-1ra) and tumour necrosis factor (TNF). ADAMTS-4 mRNA and protein were up-regulated by TNF in primary human astrocyte cultures. The increased ADAMTS-1 and -4 in experimental stroke, together with no change in TIMP-3, may promote ECM breakdown after stroke, enabling infiltration of inflammatory cells and contributing to brain injury. In vitro studies suggest that the in vivo modulation of ADAMTS-1 and -4 may be controlled in part by TNF.
Collapse
Affiliation(s)
- A K Cross
- Biomedical Research Centre, Faculty of Health and Wellbeing, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK.
| | | | | | | | | | | | | | | |
Collapse
|
448
|
Das A, Garner DP, Del Re AM, Woodward JJ, Kumar DM, Agarwal N, Banik NL, Ray SK. Calpeptin provides functional neuroprotection to rat retinal ganglion cells following Ca2+ influx. Brain Res 2006; 1084:146-57. [PMID: 16600192 DOI: 10.1016/j.brainres.2006.02.051] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Revised: 02/01/2006] [Accepted: 02/07/2006] [Indexed: 11/26/2022]
Abstract
Apoptosis of retinal ganglion cells (RGCs) impairs vision in glaucoma patients. RGCs are also degenerated in multiple sclerosis (MS), resulting in loss of visual perception in MS patients. We examined the involvement of calpain and caspase cascades in apoptosis of the rat retinal ganglion cell line RGC-5 following 24 h of exposure to 250 nM ionomycin (IMN) or 300 units/ml interferon-gamma (IFN-gamma) and then evaluated functional neuroprotection with 2 microM calpeptin (CP, a calpain-specific inhibitor). Morphological and biochemical features of apoptosis were detected in RGC-5 cells following exposure to IMN or IFN-gamma. Fura-2 assay determined significant increases in intracellular free [Ca2+] following exposure to IMN or IFN-gamma. Pretreatment with CP for 1 h prevented Ca2+ influx, proteolytic activities, and apoptosis in RGC-5 cells. Western blot analyses showed an increase in activities of calpain and caspase-12, upregulation of Bax:Bcl-2 ratio, release of cytochrome c from mitochondria, and increase in caspase-9 and caspase-3 activities during apoptosis. Increased caspase-3 activity was also confirmed by a colorimetric assay. Activation of caspase-8 and cleavage of Bid to tBid in RGC-5 cells following exposure to IFN-gamma indicated co-operation between extrinsic and intrinsic pathways of apoptosis. Patch-clamp recordings showed that pretreatment with CP attenuated apoptosis and maintained normal whole-cell membrane potential, indicating functional neuroprotection. Taken together, our results demonstrated that Ca2+ overload could be responsible for activation of calpain and caspase cascades leading to apoptotic death of RGC-5 cells and CP provided functional neuroprotection.
Collapse
Affiliation(s)
- Arabinda Das
- Department of Neurosciences, Medical University of South Carolina (MUSC), 96 Jonathan Lucas Street, Suite 323K, P.O. Box 250606, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | |
Collapse
|
449
|
Norris CM, Kadish I, Blalock EM, Chen KC, Thibault V, Porter NM, Landfield PW, Kraner SD. Calcineurin triggers reactive/inflammatory processes in astrocytes and is upregulated in aging and Alzheimer's models. J Neurosci 2006; 25:4649-58. [PMID: 15872113 PMCID: PMC1201418 DOI: 10.1523/jneurosci.0365-05.2005] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Astrocyte reactivity (i.e., activation) and associated neuroinflammation are increasingly thought to contribute to neurodegenerative disease. However, the mechanisms that trigger astrocyte activation are poorly understood. Here, we studied the Ca2+-dependent phosphatase calcineurin, which regulates inflammatory signaling pathways in immune cells, for a role in astrogliosis and brain neuroinflammation. Adenoviral transfer of activated calcineurin to primary rat hippocampal cultures resulted in pronounced thickening of astrocyte somata and processes compared with uninfected or virus control cultures, closely mimicking the activated hypertrophic phenotype. This effect was blocked by the calcineurin inhibitor cyclosporin A. Parallel microarray studies, validated by extensive statistical analyses, showed that calcineurin overexpression also induced genes and cellular pathways representing most major markers associated with astrocyte activation and recapitulated numerous changes in gene expression found previously in the hippocampus of normally aging rats or in Alzheimer's disease (AD). No genomic or morphologic evidence of apoptosis or damage to neurons was seen, indicating that the calcineurin effect was mediated by direct actions on astrocytes. Moreover, immunocytochemical studies of the hippocampus/neocortex in normal aging and AD model mice revealed intense calcineurin immunostaining that was highly selective for activated astrocytes. Together, these studies show that calcineurin overexpression is sufficient to trigger essentially the full genomic and phenotypic profiles associated with astrocyte activation and that hypertrophic astrocytes in aging and AD models exhibit dramatic upregulation of calcineurin. Thus, the data identify calcineurin upregulation in astrocytes as a novel candidate for an intracellular trigger of astrogliosis, particularly in aging and AD brain.
Collapse
Affiliation(s)
- Christopher M Norris
- Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | | | | | | | | | | | | | |
Collapse
|
450
|
Chin J, Palop JJ, Puoliväli J, Massaro C, Bien-Ly N, Gerstein H, Scearce-Levie K, Masliah E, Mucke L. Fyn kinase induces synaptic and cognitive impairments in a transgenic mouse model of Alzheimer's disease. J Neurosci 2006; 25:9694-703. [PMID: 16237174 PMCID: PMC6725734 DOI: 10.1523/jneurosci.2980-05.2005] [Citation(s) in RCA: 252] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Human amyloid precursor protein (hAPP) transgenic mice with high levels of amyloid-beta (Abeta) develop behavioral deficits that correlate with the depletion of synaptic activity-related proteins in the dentate gyrus. The tyrosine kinase Fyn is altered in Alzheimer's disease brains and modulates premature mortality and synaptotoxicity in hAPP mice. To determine whether Fyn also modulates Abeta-induced behavioral deficits and depletions of synaptic activity-dependent proteins, we overexpressed Fyn in neurons of hAPP mice with moderate levels of Abeta production. Compared with nontransgenic controls and singly transgenic mice expressing hAPP or FYN alone, doubly transgenic FYN/hAPP mice had striking depletions of calbindin, Fos, and phosphorylated ERK (extracellular signal-regulated kinase), impaired neuronal induction of Arc, and impaired spatial memory retention. These deficits were qualitatively and quantitatively similar to those otherwise seen only in hAPP mice with higher Abeta levels. Surprisingly, levels of active Fyn were lower in high expresser hAPP mice than in NTG controls and lower in FYN/hAPP mice than in FYN mice. Suppression of Fyn activity may result from dephosphorylation by striatal-enriched phosphatase, which was upregulated in FYN/hAPP mice and in hAPP mice with high levels of Abeta. Thus, increased Fyn expression is sufficient to trigger prominent neuronal deficits in the context of even relatively moderate Abeta levels, and inhibition of Fyn activity may help counteract Abeta-induced impairments.
Collapse
Affiliation(s)
- Jeannie Chin
- Gladstone Institute of Neurological Disease, University of California, San Francisco, California 94158, USA
| | | | | | | | | | | | | | | | | |
Collapse
|