Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Publication year range
1.
Biochim Biophys Acta ; 1311(2): 71-6, 1996 Apr 24.
Article in English | MEDLINE | ID: mdl-8630332

ABSTRACT

This study explored the role of clusterin in mechanisms of cell adhesion and apoptosis in P19 embryonic carcinoma cells. We found that serum deprivation induced transient but dramatic elevation in cell adhesion strength to the culture substrate and eventually led to apoptotic cell death. The time course of cell-adhesion increase overlapped temporally with the elevation of clusterin mRNA (peak 8 h after serum deprivation). The coincidental elevation of clusterin expression and cell adhesion strength preceded the schedule of apoptotic cell death. Clusterin antiserum partially antagonized cell adhesion, but did not modify the course of apoptosis. These data suggest that clusterin expression may partially control cell adhesion with no influence on apoptosis in P19 cells, under defined conditions.


Subject(s)
Apoptosis/physiology , Cell Adhesion , Glycoproteins/biosynthesis , Molecular Chaperones , Carcinoma, Embryonal , Clusterin , Culture Media, Serum-Free , Glycoproteins/genetics , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Tumor Cells, Cultured
2.
FASEB J ; 17(6): 725-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12586733

ABSTRACT

Recent studies indicate that the proinflammatory enzyme cyclooxygenase (COX)-2, an enzyme involved in inflammatory cascades but also normal neuronal activities, is elevated in the brain and spinal cord of amyotrophic lateral sclerosis (ALS) patients and ALS mouse model systems. On the basis of this evidence, we explored the impact of COX-2 inhibition on the onset and progression of ALS-like disease in the G93A human superoxide dismutase (SOD)1 mouse model of ALS. We found that prophylactic administration of nimesulide, a preferential COX-2 inhibitor, in the feed resulted in a significant delay in the onset of ALS type motor impairment. This delay of ALS symptomatology temporally overlapped with the inhibition of prostaglandin E2 elevation in the spinal cord of SOD1-G93A transgenic mice relative to untreated SOD1-G93A controls. This study strongly supports a role for COX-2 in the pathophysiology of ALS and provides the first experimental evidence that prophylactic treatment with COX-2 inhibitors can significantly delay the onset of motor dysfunction in the SOD1-G93A transgenic mouse model of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Sulfonamides/therapeutic use , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/administration & dosage , Dinoprostone/blood , Dinoprostone/metabolism , Disease Models, Animal , Isoenzymes/drug effects , Isoenzymes/metabolism , Mice , Mice, Transgenic , Motor Activity/drug effects , Mutation , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Sulfonamides/administration & dosage , Sulfonamides/blood , Superoxide Dismutase/genetics
3.
Neurobiol Aging ; 17(5): 707-16, 1996.
Article in English | MEDLINE | ID: mdl-8892343

ABSTRACT

This review discusses key findings indicating potential roles of the complement (C)-system in chronic inflammation in Alzheimer's disease (AD) brain. Although there is no means to cure or prevent the disease, recent studies suggest that antiinflammatory drugs may delay the onset of AD dementia. One target of these drugs may be the (C)-system, which is best known for its roles in inflammatory processes in peripheral tissues. However, recent data show C-system expression and regulation in brain cells, and C-system protein deposition in AD plaques. It is still nuclear whether C-system activation contributes to neuropathology in the AD brain, as shown in multiple sclerosis (MS). New clinical studies with antiinflammatory agents are now under general consideration by the Alzheimer's Disease Cooperative Study program. In this review I outline research directions which address possible C-system contributions to neurodegeneration. Finally, I discuss potential pharmacological interventions designed to control segments of classical inflammatory cascades in which the C-system is highly implicated. These aspects are critical to the understanding of C-mediated responses in normal and pathologic brain.


Subject(s)
Alzheimer Disease/pathology , Complement System Proteins/physiology , Inflammation/pathology , Nerve Degeneration/physiology , Alzheimer Disease/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain/pathology , Humans , Inflammation/drug therapy , Nerve Degeneration/drug effects
4.
Neurobiol Aging ; 13(6): 641-8, 1992.
Article in English | MEDLINE | ID: mdl-1362796

ABSTRACT

This study describes evidence in the adult human and rat brain for mRNAs that encode two complement (C) proteins, C1qB and C4. C proteins are important effectors of humoral immunity and inflammation in peripheral tissues but have not been considered as normally present in brain. Previous immunocytochemical studies showed that C proteins are associated with plaques, tangles, and dystrophic neurites in Alzheimer's disease (AD), but their source is unknown. Combined immunocytochemistry and in situ hybridization techniques show C4 mRNA in pyramidal neurons and C1qB mRNA in microglia. Primary rat neuron cultures also show C1qB mRNA. In the cortex from AD brains, there were two- to threefold increases of C1qB mRNA and C4 mRNA, and increased C1qB mRNA prevalence was in part associated with microglia. As a model for AD, we examined entorhinal cortex perforant path transection in the rat brain, which caused rapid increases of C1qB mRNA in the ipsilateral, but not contralateral, hippocampus and entorhinal cortex. The role of brain-derived acute and chronic C induction during AD and experimental lesions can now be considered in relation to functions of C proteins that pertain to cell degeneration and/or cell preservation and synaptic plasticity.


Subject(s)
Alzheimer Disease/metabolism , Brain Chemistry/physiology , Brain/physiology , RNA, Messenger/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Antibodies/immunology , Antibodies/isolation & purification , Base Sequence , Blotting, Northern , Brain/pathology , Carrier Proteins/biosynthesis , Cells, Cultured , Cerebral Cortex/pathology , Cloning, Molecular , Female , Hippocampus/pathology , Humans , Immunohistochemistry , In Situ Hybridization , Male , Molecular Sequence Data , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/isolation & purification , Rats
5.
J Comp Neurol ; 339(3): 387-400, 1994 Jan 15.
Article in English | MEDLINE | ID: mdl-8132868

ABSTRACT

Clusterin (SGP-2) is a newly described glycoprotein associated with several putative functions including responses to brain injury. This study reports the regional and cell type expression of clusterin mRNA and its encoded glycoprotein in the rat brain; a limited comparison was also done with the human brain. Using in situ hybridization combined with immunocytochemistry, we found that astrocytes and neurons may express clusterin mRNA in the normal adult brain. While astrocytes throughout the brain contained clusterin mRNA, there was regional selectivity for neuronal clusterin expression. In the striatum, clusterin mRNA was not detected in neurons. Only a subset of substantia nigra dopaminergic neurons or locus ceruleus noradrenergic neurons (tyrosine hydroxylase immunopositive) contained clusterin mRNA. However, neuronal clusterin mRNA was prevalent in pontine nuclei and in the red nucleus of the midbrain tegmentum. Similarly, clusterin mRNA was prevalent in both rat and human hippocampal neuron-specific enolase immunopositive pyramidal neurons, although rat CA1 neurons had less mRNA than CA2-CA3 neurons. Monotypic primary cell cultures from the neonatal rat showed clusterin mRNA in both neurons and astrocytes, but not in microglia. By immunocytochemistry, no clusterin immunopositive glia were observed in any region of the rat brain, confirming previous studies. However, clusterin immunopositive cells (putative neurons) were observed in the Purkinje cell layer of the cerebellum, medial and interposed cerebellar nuclei, trigeminal motor nucleus, and red nucleus. Finally, in vitro studies suggest that astrocytes, but not neurons, secrete clusterin, which is pertinent to clusterin immunodeposits found after experimental lesioning.


Subject(s)
Astrocytes/metabolism , Brain Chemistry/physiology , Glycoproteins/physiology , Molecular Chaperones , Neurons/metabolism , Alzheimer Disease/metabolism , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Clusterin , Glycoproteins/biosynthesis , Humans , Immunohistochemistry , In Situ Hybridization , Male , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344
6.
Arch Neurol ; 57(8): 1153-60, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10927795

ABSTRACT

BACKGROUND: Inflammatory cytokines have been linked to Alzheimer disease (AD) neurodegeneration, but little is known about the temporal control of their expression in relationship to clinical measurements of AD dementia progression. DESIGN AND MAIN OUTCOME MEASURES: We measured inflammatory cytokine messenger RNA (mRNA) expression in postmortem brain specimens of elderly subjects at different clinical stages of dementia and neuropathological dysfunction. SETTING AND PATIENTS: Postmortem study of nursing home patients. RESULTS: In brains of cognitively normal control subjects, higher interleukin 6 (IL-6) and transforming growth factor beta1 (TGF-beta1) mRNA expression was observed in the entorhinal cortex and superior temporal gyrus compared with the occipital cortex. Compared with age-matched controls, subjects with severe/terminal dementia, but not subjects at earlier disease stages, had higher IL-6 and TGF-beta1 mRNA expression in the entorhinal cortex (P<.01) and superior temporal gyrus (P<.01). When stratified by the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropathological criteria, IL-6 mRNA expression in both the entorhinal cortex (P<.05) and superior temporal gyrus (P<.01) correlated with the level of neurofibrillary tangles but not neuritic plaques. However, in the entorhinal cortex, TGF-beta1 mRNA did not correlate with the level of either neurofibrillary tangles or neuritic plaques. Interestingly, in the superior temporal gyrus, TGF-beta1 mRNA expression negatively correlated with neurofibrillary tangles (P<.01) and showed no relationship to the pathological features of neuritic plaques. CONCLUSIONS: The data are consistent with the hypothesis that cytokine expression may differentially contribute to the vulnerability of independent cortical regions during the clinical progression of AD and suggest that an inflammatory cytokine response to the pathological effects of AD does not occur until the late stages of the disease. These findings have implications for the design of anti-inflammatory treatment strategies. Arch Neurol. 2000;57:1153-1160


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Interleukin-6/genetics , Transforming Growth Factor beta/genetics , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Cognition , Disease Progression , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Female , Gene Expression/immunology , Humans , Male , Neurofibrillary Tangles/pathology , Occipital Lobe/metabolism , Occipital Lobe/pathology , Plaque, Amyloid/pathology , RNA, Messenger/analysis , Temporal Lobe/metabolism , Temporal Lobe/pathology
7.
Arch Neurol ; 58(3): 487-92, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11255454

ABSTRACT

BACKGROUND: Prior studies have shown that cyclooxygenase 2 (COX-2), an enzyme involved in inflammatory mechanisms and neuronal activities, is up-regulated in the brain with Alzheimer disease (AD) and may represent a therapeutic target for anti-inflammatory treatments. OBJECTIVE: To explore COX-2 expression in the brain as a function of clinical progression of early AD. DESIGN AND MAIN OUTCOME MEASURES: Using semiquantitative immunocytochemistry, we analyzed COX-2 protein content in the hippocampal formation in 54 postmortem brain specimens from patients with normal or impaired cognitive status. SETTING AND PATIENTS: Postmortem study of nursing home residents. RESULTS: The immunointensity of COX-2 signal in the CA3 and CA2 but not CA1 subdivisions of the pyramidal layers of the hippocampal formation of the AD brain increased as the disease progressed from questionable to mild clinical dementia as assessed by Clinical Dementia Rating. COX-2 signal was increased in all 3 regions examined among cases characterized by severe dementia. CONCLUSION: Neuronal COX-2 content in subsets of hippocampal pyramidal neurons may be an indicator of progression of dementia in early AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Hippocampus/enzymology , Hippocampus/pathology , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Aged , Aged, 80 and over , Cyclooxygenase 2 , Disease Progression , Female , Humans , Immunohistochemistry , Isoenzymes/analysis , Male , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/analysis
8.
Neurology ; 58(7): 1050-4, 2002 Apr 09.
Article in English | MEDLINE | ID: mdl-11940691

ABSTRACT

BACKGROUND: Nonsteroidal anti-inflammatory drugs (NSAID) may be useful in the treatment of AD. Clinical and laboratory experience with nimesulide, an NSAID with preferential cyclooxygenase-2 inhibition, suggests that it may be a good candidate for AD therapy. METHODS: This pilot study investigated the clinical feasibility of nimesulide treatment in AD. Forty persons with probable AD, most of whom were taking cholinesterase inhibitors, were enrolled in a randomized, controlled, parallel-group trial designed to assess tolerability and short-term cognitive/behavioral effects of nimesulide. In the initial 12-week double-blind phase, participants were treated with nimesulide 100 mg by mouth twice daily or matching placebo; during the second 12-week phase all participants received active drug. Participants who tolerated the drug well and perceived benefit were invited to continue open-label nimesulide treatment. RESULTS: Short-term therapy with nimesulide, compared with placebo, had no significant effect on total assessment scores of measures of cognition, clinical status, activities of daily living, affect, and behavior. Long-term therapy was well tolerated for periods exceeding 2 years. CONCLUSION: These findings support the feasibility of nimesulide therapy in AD; assessment of efficacy will require a larger, long-term treatment study.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Sulfonamides/therapeutic use , Aged , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Double-Blind Method , Female , Humans , Liver/chemistry , Liver/drug effects , Male , Pilot Projects , Sulfonamides/adverse effects , Treatment Outcome
9.
J Neuroimmunol ; 105(2): 124-30, 2000 Jun 26.
Article in English | MEDLINE | ID: mdl-10742554

ABSTRACT

There is evidence that the complement system, a major component of inflammatory responses, may play an important role in neurodegenerative conditions such as Alzheimer's disease (AD). Work from our lab demonstrated that mice genetically deficient in the complement component C5 are more susceptible to hippocampal excitotoxic lesions (Pasinetti et al., 1996) and that the C5-derived ana;hylatoxin C5a may protect against excitotoxicity in vitro and in vivo (Osaka et al., 1999). Potential mechanisms identified in C5a-mediated neuroprotection include activation of mitogen activated protein (MAP)-kinase (Osaka et al., 1998; Osaka et al., 1999). This novel neuroprotective role of C5a complicates current theories that complement proteins augment beta-amyloid (Abeta) toxicity in AD. In view of the fact that the complement system represents a target for therapeutic interventions in AD, further characterization of the complex role of complement proteins is essential. Towards this aim, we have characterized a transgenic C5a receptor (C5aR) knockout (KO) mouse. Recent studies in the lab using C5aR-KO mice show that disruption of C5aR alters calcium calmodulin kinase (CaM-KII) signal transduction in brain cells. We are presently using C5aR-KO mice to study the role of C5a in caspase mediated apoptotic neuronal death. In this review we will attempt to delineate possible neuroprotective roles for C5a in mechanisms of neurotoxicity pertaining to AD.


Subject(s)
Alzheimer Disease/etiology , Complement C5a/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Caspases/physiology , Enzyme Activation , Humans , Mice , Mitogen-Activated Protein Kinases/physiology
10.
J Neuroimmunol ; 89(1-2): 142-9, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9726836

ABSTRACT

In this study we assessed the regulation of cyclooxygenase (COX)-2 in models of apoptotic cell death in vivo and in vitro. By 6 h after hippocampal colchicine injection in rat, COX-2 (but not COX-1) mRNA expression was elevated. The induction of COX-2 mRNA expression preceded temporally and overlapped anatomically the cellular morphological features of apoptosis in the granule cell layer of the dentate gyrus. Similarly, in an established in vitro model of apoptosis in P19 cells, COX-2 induction preceded apoptosis in response to serum deprivation by 12 h. These studies suggest that COX-2 may be involved in the early mechanisms leading to apoptosis.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Enzymologic/immunology , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Animals , Apoptosis/drug effects , Apoptosis/immunology , Blood Proteins/pharmacology , Colchicine , Cyclooxygenase 1 , Cyclooxygenase 2 , Dentate Gyrus/enzymology , Dentate Gyrus/immunology , Isoenzymes/analysis , Male , Membrane Proteins , Neoplastic Stem Cells , Nerve Degeneration/chemically induced , Nerve Degeneration/enzymology , Nerve Degeneration/immunology , Peroxidases/analysis , Peroxidases/genetics , Prostaglandin-Endoperoxide Synthases/analysis , Pyramidal Cells/enzymology , Pyramidal Cells/immunology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
11.
J Neuroimmunol ; 58(1): 101-10, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7730444

ABSTRACT

This study characterizes the effect of transforming growth factor (TGF) beta 1 on clusterin expression in rat brain cells. 24 h after an acute unilateral intracerebroventricular infusion of TGF-beta 1, clusterin mRNA prevalence was increased in astrocytes that contained immunoreactive (IR) glial fibrillary acidic protein (GFAP). TGF-beta 1 selectively induced clusterin mRNA in astrocytes, as no clusterin mRNA was detected in neurons, oligodendrocytes, or microglia. TGF-beta 1 induced a bilateral increase in clusterin mRNA per astrocyte. Astrocyte hypertrophy (GFAP-IR area) was only increased on the ipsilateral side. In pure astrocyte cultures, TGF-beta 1 (200 pM) decreased clusterin mRNA levels and the rate of clusterin RNA transcription. However, in cultures of astrocytes that contained microglia and oligodendrocytes (mixed glia cultures), TGF-beta 1 caused a dose-dependent increase in astrocytic clusterin mRNA levels. The astrocytes that responded to TGF-beta 1 included two GFAP-IR subtypes, type 1 and 2. TGF-beta 1 increased clusterin protein in the conditioned medium from cultured glia, in either monotypic or mixed glial cultures. Thus, TGF-beta 1 and heterotypic cell interactions influence clusterin expression by astrocytes and may be important to the role of clusterin in multiple sclerosis, AIDS, and Alzheimer's disease.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Cerebral Ventricles/physiology , Corpus Striatum/metabolism , Gene Expression , Glycoproteins/biosynthesis , Molecular Chaperones , Transforming Growth Factor beta/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Ventricles/drug effects , Clusterin , Corpus Striatum/cytology , Glial Fibrillary Acidic Protein/analysis , Infusions, Parenteral , Male , Microglia/cytology , Microglia/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344 , Transcription, Genetic , Transforming Growth Factor beta/administration & dosage
12.
Neuroscience ; 87(2): 319-24, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9740394

ABSTRACT

Many epidemiological studies suggest that use of nonsteroidal anti-inflammatory drugs delays or slows the clinical expression of Alzheimer's disease, but the mechanism by which these drugs might affect pathophysiological processes relevant to Alzheimer's disease has been unclear. Non-steroidal anti-inflammatory drugs are presumed to act by inhibiting cyclooxygenase, a key enzyme in the metabolism of membrane-derived arachidonic acid into prostaglandins. In recent years, two distinct isoforms of cyclooxygenase have been characterized, a constitutive form, cyclooxygenase-1, and a mitogen-inducible form, cyclooxygenase-2. Cyclooxygenase-2 has been identified in rodent brain. Excitotoxic lesions cause up-regulation of cyclooxygenase-2 expression coincident with the onset of expression of markers of apoptosis; cyclooxygenase-2 thus represents a possible target of non-steroidal anti-inflammatory drug action in neurodegenerative mechanisms. In the present study, we examined cyclooxygenase-2 gene expression in Alzheimer's disease and control cases. We found up-regulation of cyclooxygenase-2 expression in Alzheimer's disease frontal cortex. Further, we found that synthetic beta-amyloid peptides induced cyclooxygenase-2 expression in SH-SY5Y neuroblastoma cells in vitro, suggesting a mechanism for cyclooxygenase-2 up-regulation in Alzheimer's disease. These findings support the investigation of selective cyclooxygenase-2 inhibitors for the treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/enzymology , Isoenzymes/biosynthesis , Prefrontal Cortex/enzymology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Blotting, Northern , Blotting, Western , Cyclooxygenase 2 , Humans , Immunohistochemistry , Membrane Proteins , Neurons/enzymology , RNA, Messenger/biosynthesis
13.
Neuroscience ; 127(3): 649-58, 2004.
Article in English | MEDLINE | ID: mdl-15283964

ABSTRACT

We have reported recently that the microtubule-associated protein tau is phosphorylated in vitro by Akt, an important kinase in anti-apoptotic signaling regulated by insulin and growth factors. We also established that Akt phosphorylates tau separately at T212 and S214, two sites previously shown to be phosphorylated by glycogen synthase kinase 3beta (GSK3beta) and protein kinase A (PKA), respectively. In the present studies, we examined the relationship between Akt and T212/S214 in primary cultures of human neurons and astrocytes, and evaluated the contribution of two other kinases. In intact cells, we found a very low content of active (phospho-S473) form of Akt. We also found a low content of phospho-S214 but not phospho-T212 of tau, suggesting that only phospho-S212 may depend on Akt activity in situ. We upregulated Akt activity using two experimental models: treatment with a protein phosphatase inhibitor, okadaic acid, and transfection with a constitutively active Akt gene construct (c-Akt). Under these conditions, phosphorylation of tau at T212 and S214 was regulated independently, with little change or downregulation of phospho-T212 and dynamic upregulation of phospho-S214. Our studies revealed that Akt may influence the phospho-S214 content in a meaningful manner. They also revealed that PKA may only partially contribute to the phosphorylation of S214. In comparison, okadaic acid treatment severely depleted the content of GSK3beta and downregulated the remaining GSK3beta activity by Akt-dependent inhibition, consistent with minimal changes in phospho-T212. In summary, these results strongly suggest that in primary cultures, Akt selectively phosphorylates tau at S214 rather than T212. Our studies raise the possibility that tau S214 may participate in Akt-mediated anti-apoptotic signaling.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , tau Proteins/metabolism , Apoptosis/physiology , Astrocytes/cytology , Blotting, Western , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Epitopes/metabolism , Fetus , Gene Expression , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Neurons/cytology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Serine/metabolism , Threonine/metabolism , Up-Regulation
14.
Neuroscience ; 54(4): 893-907, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8341423

ABSTRACT

Transforming growth factor-beta 1 rapidly increases in adult rat brain in response to experimental lesions. This study characterized the schedule of changes, regional distribution, and cellular localization of striatal transforming growth factor-beta 1 messenger RNA and fibronectin messenger RNA following partial striatal deafferentation by frontal cortex ablation. Frontal cortex ablation induced striatal transforming growth factor-beta 1 messenger RNA elevations that coincided temporally and overlapped anatomically with the course of degeneration of cortico-striatal afferent fibers. Within three days post-lesioning, transforming growth factor-beta 1 messenger RNA was localized at the cortical wound. By 10 days, the anatomical site of transforming growth factor-beta 1 messenger RNA expression shifted to the dorsal half of the deafferented striatum and co-localized with OX-42+ immunostained microglia-macrophage at the site of degenerating afferent terminals. Similarly, fibronectin messenger RNA also shifted from the cortical wound to the deafferented striatum by 10 days post-lesioning. Fibronectin messenger RNA was localized to glial fibrillary acidic protein+ immunostained astrocytes surrounding degenerating corticostriatal afferents. Infusion of transforming growth factor-beta 1 peptide elevated striatal and cortical fibronectin messenger RNA. These findings suggest that microglia-macrophage associated with degenerating afferent fibres can upregulate transforming growth factor-beta 1 messenger RNA and may influence fibronectin messenger RNA synthesis in reactive astrocytes. This study suggests that transforming growth factor-beta 1 has a role in controlling extracellular matrix synthesis following brain injury, which is analogous to that in peripheral wound healing.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Fibronectins/biosynthesis , RNA, Messenger/biosynthesis , Transforming Growth Factor beta/biosynthesis , Animals , Astrocytes/metabolism , Autoradiography , Brain/cytology , Brain/drug effects , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , DNA/metabolism , Glial Fibrillary Acidic Protein/immunology , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , In Situ Hybridization , Injections, Intraventricular , Male , Nerve Growth Factors/biosynthesis , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Rats , Rats, Inbred F344 , Silver Staining , Transforming Growth Factor beta/pharmacology
15.
Neuroscience ; 88(4): 1073-82, 1999.
Article in English | MEDLINE | ID: mdl-10336122

ABSTRACT

In this study we explored the potential role of the complement derived anaphylatoxin C5a and the expression of its receptor in mouse brain. Using in situ hybridization, we found that C5a receptor messenger RNA is expressed in mouse brain. In response to intraventricular kainic acid injection, there was marked increase in the C5a receptor messenger RNA expression, particularly in hippocampal formation and cerebral cortex. C5a ligand-binding autoradiography confirmed the functional expression and elevation of the C5a receptor post-lesioning. The expression of C5a receptor messenger RNA in brain was confirmed by northern blot hybridization of total RNA from neuronal and glial cells in vitro. Based on these findings we explored the role of C5a in mechanisms of signal transduction in brain cells. Treatment of primary cultures of mouse astrocytes with human recombinant C5a resulted in the activation of mitogen-activated extracellular signal-regulated protein kinase. This response appeared to be mediated by the C5a receptor since astrocyte cultures derived from C5a receptor knockout mice were not responsive to the treatment. Understanding the regulation of C5a receptor in brain and mechanisms by which pro-inflammatory C5a modulates specific signal transduction pathways in brain cells is crucial to studies of inflammatory mechanisms in neurodegeneration.


Subject(s)
Brain/metabolism , Complement C5a/metabolism , Receptors, Complement/metabolism , Animals , Brain/cytology , Brain Diseases/chemically induced , Brain Diseases/metabolism , Cells, Cultured , Complement C5a/genetics , Complement C5a/pharmacology , Humans , Kainic Acid , Male , Mice , Mice, Inbred Strains , Mice, Knockout/genetics , Nerve Degeneration/physiopathology , Neuroglia/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, Complement/physiology , Recombinant Proteins , Signal Transduction/physiology
16.
Neuroscience ; 53(1): 199-211, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8469307

ABSTRACT

This investigation describes the schedule and regional distribution of astrocytic responses in striatum following deafferentation by unilateral frontal cortex ablation. In the ipsilateral deafferented striatum, glial fibrillary acidic protein and clusterin (sulfated glycoprotein-2) messengerRNA showed peak elevations by 10 days postlesioning (Northern blots). Vimentin messengerRNA responded faster, with a transient elevation by three days postlesioning. The messengerRNA for glial fibrillary acidic protein, clusterin and vimentin returned toward control levels by 27 days postlesioning. However, the neuronal marker growth-associated protein messengerRNA, was decreased at all postlesion times. By in situ hybridization, the increased glial fibrillary acidic protein messengerRNA and clusterin messengerRNA signals were localized mainly to the dorsal half of the ipsilateral deafferented striatum and followed the same schedule as found by Northern blots. Glial fibrillary acidic protein messengerRNA was widely diffused in the dorsal striatum and was excluded from fascicles of the internal capsule; a similar distribution was found for glial fibrillary acidic protein-immunopositive astrocytes. While clusterin messengerRNA signal showed a distinct clustering, its immunoreactivity appeared as deposits in the deafferented striatal neuropil; Western blots confirmed the immunocytochemical results. By in situ hybridization, vimentin messengerRNA was mostly localized to the cortical wound cavity dorsal to the deafferented striatum and overlapped the distribution of vimentin-immunopositive cells. These findings suggest a coordination of striatal astrocytic messengerRNA responses with the degeneration of corticostriatal afferents. We also compared these same parameters with those from published reports on the hippocampus after deafferenting lesions. Certain astrocyte molecular responses to deafferentation are detected about five days earlier in the hippocampus than in the striatum. This different schedule in response to decortication may pertain to differences in synaptic remodeling in the hippocampus vs striatum.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/physiology , Molecular Chaperones , Neurons, Afferent/physiology , RNA, Messenger/biosynthesis , Animals , Blotting, Northern , Blotting, Western , Clusterin , Corpus Striatum/cytology , GAP-43 Protein , Glial Fibrillary Acidic Protein/biosynthesis , Glycoproteins/biosynthesis , Immunohistochemistry , In Situ Hybridization , Male , Membrane Glycoproteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neuroglia/metabolism , Rats , Rats, Inbred F344 , Synapses/physiology , Vimentin/biosynthesis
17.
Neuroscience ; 62(2): 425-39, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7830889

ABSTRACT

This ultrastructural and light microscopic immunocytochemical study describes the time course of anatomical changes that occur in striatal astrocytes in response to neuronal deafferentation in young adult rats and the coordinate distribution of two astrocytic proteins involved in reactive synaptogenesis, glial fibrillary acidic protein and clusterin. We found that following a unilateral lesion of the cerebral cortex, striatal astrocytes undergo a rapid ultrastructural transformation from a protoplasmic to a reactive type of astroglia and are the primary cells involved in the removal of degenerating axon terminals, but not axons of passage, from the neuropil. In addition, at 10 and 27 days postlesion, processes of reactive astrocytes are also seen to occupy vacant postsynaptic spines after degenerating presynaptic terminals are removed, suggesting that they may also participate in the reinnervation of the deafferented neurons. By immunocytochemistry, reactive astrocytes were characterized by a significant increase in the intensity of glial fibrillary acidic protein staining beginning at three days postlesion and lasting for at least 27 days postlesion. Reactive astrocytes were characterized by cellular hypertrophy and an increase in the density of immunoreactive processes distributed throughout the deafferented striatum. However, our analysis of astrocyte cell number found no evidence of astrocyte proliferation in response to the deafferentation lesion. Although previous in situ hybridization studies have reported elevated clusterin messenger RNA in reactive astrocytes after decortication, clusterin immunoreactivity was not seen in the cell soma of reactive astrocytes but was distributed as punctate deposits, ranging from 1 to 2 microns in diameter, within the neuropil of the deafferented striatum. At 10 days postlesion, the distribution of clusterin staining appeared as large aggregates of immunoreactive deposits adjacent to neurons. However, by 27 days postlesion, the aggregates of clusterin reaction product were replaced by a fine scattering of individual punctate deposits distributed evenly over the dorsal part of the deafferented striatum. These data support the notion that reactive astrocytes serve multiple, time-dependent roles in response to brain injury and are involved in both the removal of degenerative debris from the lesion site as well as in reforming the synaptic circuitry of the damaged brain. Our data suggest that, in response to decortication, reactive astrocytes are the primary cells responsible for removing degenerating axon terminals, but not axons of passage, from the deafferented striatum and that the coordinate increase in glial fibrillary acidic protein may serve to stabilize the extension of reactive astrocytic processes during phagocytosis.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Afferent Pathways/physiology , Astrocytes/cytology , Corpus Striatum/cytology , Glial Fibrillary Acidic Protein/analysis , Molecular Chaperones , Animals , Astrocytes/ultrastructure , Axons/ultrastructure , Clusterin , Corpus Striatum/ultrastructure , Denervation , Glycoproteins/analysis , Immunohistochemistry , Male , Microscopy, Electron , Nerve Degeneration , Nerve Fibers/ultrastructure , Rats , Rats, Inbred F344
18.
Neuroscience ; 47(2): 303-15, 1992.
Article in English | MEDLINE | ID: mdl-1641125

ABSTRACT

In situ hybridization techniques were used to analyse the spatiotemporal pattern of brain-derived neurotrophic factor messenger RNA elevation associated with kainic acid-induced seizure activity in the rat. Pronounced increases in hippocampal brain-derived neurotrophic factor messenger RNA levels were observed as early as 30 min following the onset of behavioral seizures. The greatest increase (10-fold) occurred in the dentate granule cell layer, while pyramidal layers CA1, CA3, and CA4 exhibited increases of two- to six-fold. Peak elevation of brain-derived neurotrophic factor messenger RNA in CA1 hippocampal region was evident at 4 h in CA3, and in the dentate granule layer at 30 min postseizure. Elevations persisted in the dentate and hilar regions to four days, while the increases in CA1 and CA3 returned to control levels by 16 h following seizure. Significant increases in brain-derived neurotrophic factor messenger RNA were also observed in the superficial layers of cortex (II and III) and in the piriform cortex which reached peak elevations by 8 h. No detectable changes were observed in the dorsomedial thalamus. Although histologically defined pyramidal and granule cell layers displayed relatively uniform increases in brain-derived neurotrophic factor messenger RNA in response to kainate, a closer examination of the labeling patterns using emulsion autoradiography revealed discrete areas of high grain densities overlapping uniform, moderate hybridization densities in the dentate granule cell layer and CA3, suggesting that the capacity to upregulate brain-derived neurotrophic factor messenger RNA in these regions may differ among individual neurons. In conclusion, our studies revealed that brain-derived neurotrophic factor messenger RNA induction in response to systemic kainate administration differs in hippocampal and cortical areas, in magnitude, time of onset and duration. The observed temperospatial pattern does not correspond in a simple way to increases in metabolic or electrical activity associated with seizures or neuronal vulnerability coincident with the seizures.


Subject(s)
Brain/metabolism , Kainic Acid , Nerve Tissue Proteins/biosynthesis , RNA, Messenger/biosynthesis , Seizures/metabolism , Animals , Blotting, Northern , Brain/drug effects , Brain-Derived Neurotrophic Factor , Female , Hippocampus/cytology , Hippocampus/metabolism , Histocytochemistry , Male , Neurons/metabolism , Nucleic Acid Hybridization , Pentobarbital/pharmacology , RNA Probes , Rats , Rats, Inbred Strains , Seizures/chemically induced , Sulfur Radioisotopes
19.
Neuroscience ; 62(3): 741-58, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7870303

ABSTRACT

This study concerns expression of the genes encoding three multifunctional proteins: clusterin and two complement cascade components, C1q and C4. Previous work from this and other laboratories has established that clusterin, Clq and C4 messenger RNAs are elevated during Alzheimer's disease, and in response to deafferenting and excitotoxic brain lesion. This study addresses hippocampal clusterin, ClqB and C4 expression in response to neurotoxins that caused selective neuron death. Kainate, which preferentially kills hippocampal CA3 pyramidal neurons but not dentate gyrus granule neurons induced clusterin immunoreactivity in CA1 and CA3 pyramidal neurons and adjacent astrocytes, but not in dentate gyrus granule neurons. In contrast, colchicine, which preferentially kills the dentate gyrus granule neurons, induced clusterin immunoreactivity in the local neuropil as punctate deposits, but not in the surviving or degenerating dentate gyrus granule neurons. Clusterin messenger RNA was increased in astrocytes. ClqB and C4 messenger RNAs increased within 48 h after kainate injections, particularly in the CA3 pyramidal layer, less in the dentate gyrus-CA4, and less in CA1. Clq immunoreactivity was detected in CA1 pyramidal neurons and also as small punctate deposits in the CA1 region at eight and 14 days after kainate. The increase of both clusterin and ClqB messenger RNAs after kainate injections was blocked by barbiturates that prevented seizures and neurodegeneration. In primary hippocampal neuronal cultures treated with glutamate, a subpopulation of cultured neurons that survived glutamate toxicity also had parallel elevations of clusterin and ClqB messenger RNA. In conclusion, cytotoxins that target selective hippocampal neurons increase the expression of both clusterin and ClqB in vivo and in vitro. These results show that elevations of clusterin messenger RNA or protein can be dissociated from each other and from cell death. These increased messenger RNAs were associated with immunoreactive deposits that differed by cell type and intra- versus extracellular locations. These results suggest that the complement system is involved in brain responses to injury.


Subject(s)
Astrocytes/drug effects , Complement C1q/biosynthesis , Complement C4/biosynthesis , Gene Expression Regulation/drug effects , Glutamic Acid/toxicity , Glycoproteins/biosynthesis , Hippocampus/drug effects , Kainic Acid/toxicity , Molecular Chaperones , Nerve Tissue Proteins/biosynthesis , Neurons/drug effects , Seizures/chemically induced , Animals , Astrocytes/metabolism , Cell Death , Clusterin , Complement C1q/genetics , Complement C4/genetics , Glutamic Acid/pharmacology , Glycoproteins/genetics , Glycoproteins/physiology , Hippocampus/metabolism , Hippocampus/pathology , In Situ Hybridization , Kainic Acid/pharmacology , Male , Nerve Tissue Proteins/genetics , Neurons/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Seizures/genetics , Seizures/metabolism , Seizures/pathology
20.
Drugs ; 63 Suppl 1: 23-9, 2003.
Article in English | MEDLINE | ID: mdl-14506908

ABSTRACT

OBJECTIVE: The goal of this study was to derive a systematic approach for the identification of protein species profiles that selectively identify subgroups of similar but distinct pain models and correlate with the therapeutic efficacy (or lack thereof) of drug intervention. METHODS: Using high-throughput surface-enhanced laser desorption ionization (SELDI) mass spectrometry proteomic technology based on ProteinChip arrays, we conducted a comparative analysis of the profile of protein expression in cerebral spinal fluid (CSF) from rats exposed to either injection of complete Freund's adjuvant (CFA) [a model of inflammatory pain] or chronic constriction injury (CCI) [a model of neuropathic pain]. The CFA model was then further studied for the effects of treatment with the NSAID, nimesulide (a preferential cyclo-oxygenase [COX]-2 inhibitor). RESULTS: Among other observations, we found that the content of two metal (copper) binding protein species (2.9 and 3.2kDa) and three anionic protein species (4.0, 6.9 and 8.2kDa) were increased in the CSF of rats with inflammatory pain in a time-dependent fashion, at 7 and 14 days after CFA injection. These changes were highly selective for the CFA model, as no detectable increase in these protein biomarkers was found in the CCI neuropathic pain model. Further, we found that most of the changes in the biomarker protein species induced by the inflammatory pain were prevented by treatment with nimesulide and correlated with the antihyperalgesic effect of this drug. CONCLUSION: This study demonstrates that CSF biomarker profiles, as detected by SELDI technology, can consistently and reproducibly differentiate inflammatory from neuropathic pain, and reflect the analgesic action produced by the preferential COX-2 inhibitor, nimesulide. The characterisation and identification of these biomarkers will provide invaluable insight into the pathophysiology of pain mechanisms, in addition to further understanding of the value of nimesulide in the treatment of inflammatory pain.


Subject(s)
Cerebrospinal Fluid Proteins/analysis , Cyclooxygenase Inhibitors/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Proteomics/methods , Sulfonamides/pharmacology , Analysis of Variance , Animals , Biomarkers/analysis , Cerebrospinal Fluid Proteins/genetics , Constriction , Disease Models, Animal , Freund's Adjuvant/adverse effects , Inflammation/chemically induced , Inflammation/drug therapy , Male , Pain/diagnosis , Pain Measurement/methods , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL